Abstract
Primary immunodeficiency disorders (PID) are a group of inborn errors of immunity with a broad range of clinical severity but often associated with recurrent and serious infections. While hematopoietic stem cell transplantation (HSCT) can be curative for some forms of PID, chronic and/or refractory viral infections remain a cause of morbidity and mortality both before and after HSCT. Although antiviral pharmacologic agents exist for many viral pathogens, these are associated with significant costs and toxicities and may not be effective for increasingly drug-resistant pathogens. Thus, the emergence of adoptive immunotherapy with virus-specific T lymphocytes (VSTs) is an attractive option for addressing the underlying impaired T cell immunity in many PID patients. VSTs have been utilized for PID patients following HSCT in many prior phase I trials, and may potentially be beneficial before HSCT in patients with chronic viral infections. We review the various methods of generating VSTs, clinical experience using VSTs for PID patients, and current limitations as well as potential ways to broaden the clinical applicability of adoptive immunotherapy for PID patients.
Keywords: Primary immunodeficiency, Cytotoxic T lymphocytes, Adoptive immunotherapy, Antiviral therapy, Hematopoietic stem cell transplantation
Introduction
Primary immunodeficiency disorders (PID) are a group of inborn errors of immunity. To date, over 320 causative genes have been identified, which result in a wide spectrum of disorders ranging from profound immunodeficiency manifesting in the newborn period to immunologic disorders that can manifest in adulthood [1, 2]. Though autoimmunity and severe allergic disease are sometimes presenting signs of PID, recurrent and often serious infections are the most common presentation. Severe forms of PID such as severe combined immunodeficiency (SCID) often manifest in the newborn period with failure to thrive and susceptibility to a wide variety of pathogens [3]. Other forms of PID cause more narrow susceptibility to infections, such as the selective susceptibility to intracellular microbes caused by defects in the IL-12/IFN-γ pathway [4].
The use of hematopoietic stem cell transplantation (HSCT) has proven to be lifesaving for moderate to severe forms of PID [5, 6]. Though it has been the standard of care for SCID for over 40 years, HSCT is now increasingly being utilized to prevent late complications in other forms of PID such as chronic granulomatous disease, DOCK8 deficiency, GATA2 haploinsufficiency, and IPEX and related syndromes, such as individuals with STAT3 gain-of-function mutations [7–10]. In many of these forms of PID, patients often have a significant infectious burden at the time of diagnosis.
Chronic and refractory viral infections are a cause of significant mortality both before and after HSCT in patients with PID [11, 12, 13•]. Cytomegalovirus (CMV), Epstein-Barr virus (EBV), and respiratory viruses have been described as the leading causes of virus-associated mortality in this population, and accounted for up to 40% of transplant-related mortality [3, 5]. Though newborn screening for SCID has been tremendously successful in permitting early preventative treatments and definitive therapy prior to occurrence of severe infections [14], the vast majority of PID are not caught by the TREC assay. Antiviral pharmacotherapy is available for many viruses, but is often limited by toxicity and resistance [15–19]. Given the need for a conditioning (chemotherapy) regimen prior to HSCT for many forms of PID, active viral infections can pose serious risks for patients undergoing transplantation for PID [13•].
Reconstitution of T cell immunity is critical for control of viral infections. However, the emergence of naïve T cells usually does not occur for 3–6 months after HSCT and may be further delayed in the setting of cord blood transplantation or with the occurrence of graft-versus-host disease (GVHD) [20, 21]. During this period, patients remain profoundly susceptible to viral infections. Infusion of unmanipulated donor lymphocytes has been a successful strategy to combat persistent viral infections after HSCT, but this approach risks severe GVHD [22–24]. Adoptive immunotherapy with virus-specific T lymphocytes (VSTs) allows reconstitution of antiviral immunity after HSCT and has been effective for treatment or prevention of viral infections with CMV, EBV, and adenovirus (AdV) with minimal risk of GVHD [25, 26]. Though most prior studies have utilized VSTs generated from the HSCT donor, more recently the use of banked, partially HLA-matched VST from third party donors has permitted “off the shelf’ therapy for critically ill patients [27]. Thus, it is possible to circumvent the need for a unique donor and the time required for VST production.
Over 400 patients have been treated with VSTs in previous phase I and II trials, with the majority of patients being treated following HSCT. As PID represents a sizeable proportion of the non-malignant referrals for HSCT in pediatrics, patients with PID have been represented in many prior VST trials. Here, we review the prior use of VST therapy for patients with PID, as well as upcoming trials and advances in adoptive immunotherapy, and how it may impact the future care of patients with PID.
VST Generation Methods
Adoptive T cell therapy for viral infections has been successfully used clinically for over 20 years [28, 29•]. While unmanipulated donor lymphocyte infusions (DLIs) are capable of restoring virus-specific immunity after HSCT, this approach increases the risk of GVHD [30]. Early studies demonstrated that selective transfer of antigen-specific T cells, isolated via ex vivo expansion or multimer selection, decreased the risk of GVHD [31–33].
Initial adoptive T cell therapy methods relied on prolonged ex vivo culture methods by which the desired virus-specific T cells would be activated and expand in the setting of stimulation with cytokines while alloreactive cells would die in culture. Early culture methods required not only being able to identify the immunogenic antigens but also having antigen presenting cells (APCs) that could not only present the target antigens but also provide costimulatory molecules to promote T cell activation and expansion. Dendritic cells (DCs), activated monocytes, phytohemagglutinin blast (PHA blasts), and lymphoblastoid cell lines (LCLs) may be used as APCs, but using APC to multiply stimulate VSTs does increase the time and complexity of the manufacturing process [31, 34••]. Viruses or viral lysates have been used as antigen sources, but the advent of synthetic peptide pools that span immunodominant proteins has improved culture protocols by eliminating the risk of live viral transduction, and allowing expansion of T cells with a variety of HLA restrictions [35]. Rapid ex vivo culture methods using a single stimulation with APC pulsed with synthetic peptide pools can shorten the duration of manufacturing to approximately 10 to 14 days while maintaining antigen specificity and achieving the cellular expansion needed for clinical use.
While the rapid ex vivo expansion of VSTs has appreciably reduced manufacturing time and costs, VSTs can be produced even more rapidly using selection techniques such as multimer selection or IFN-γ capture [36–38]. In multimer selection, magnetically labeled peptide multimers are used to select T cells that are reactive to immunogenic peptides. Similarly, IFN-γ capture selects T cells that produce IFN-γ in the presence of stimulation by viral antigens using an immunomagnetic separation device. While these techniques can provide a product within 24 to 48 h, they require the donor to be immune to the virus(es) of interest and a high number of circulating reactive T cells in the starting product. Therefore, apheresis is often needed to procure the number of T cells needed for clinical use, and antigen-specific T cell yields are typically limited. Multimer selection is an HLA-restricted process, but IFN-γ capture has the advantage of not being HLA-restricted and produces a product that contains CD4+ and CD8+ T cells that recognize targeted viral epitopes.
Clinical Experience Using VSTs for PID Patients
Studies Utilizing Ex Vivo Expansion Methodologies
Numerous immunodeficiencies such as X-linked lymphoproliferative disorder and familial hemophagocytic lymphohistiocytosis (FHLH) are commonly associated with EBV-related complications ranging from fulminant mononucleosis to lymphoproliferative disorders and EBV-related lymphomas [39]. As these complications are secondary to ineffective EBV-specific T cell surveillance allowing for uncontrolled proliferation of EBV-infected B cells, adoptive T cell therapy with EBV-specific T cell therapy is a logical therapeutic option as it addresses the underlying immune deficit. Heslop et al. treated 101 patients at risk for EBV-related lymphoproliferative disease prophylactically and 13 patients therapeutically with EBV-specific T cells generated using LCLs as antigens presenting cells [40••, 41]. This cohort included 13 patients with PID. Remarkably, none of the patients in the prophylaxis cohort developed lymphoproliferative disease, and 11 of the 13 patients treated therapeutically had complete remissions. Seven of the PID patients in the prophylaxis cohort had EBV viremia at the time of infusion, four of whom had resolution of viremia, and the remaining three had decreases in EBV viral load without clearance.
To expand the clinical applicability of VST, Leen et al. demonstrated that CMV, EBV, and AdV-specific T cells can be manufactured in a monoculture using EBV-LCLs transduced with an adenoviral vector expressing CMV-pp65 protein (Ad5f35-CMVpp65) and can produce clinically relevant effects [34••]. Eleven patients, including one with SCID, received VST infusions, and all patients with active infection at the time of infusion had resolution of viral symptoms and reduction in viral titers. However, it was noted that while EBV and CMV-specific T cells expanded in all patients within 4 weeks of infusion, AdV-specific T cells only expanded in patients with recent or active AdV infections. As these trivirus-specific products were dominated by CMV-reactive T cells, the culture method was modified to use LCLs transduced with a Ad5f35 vector to produce bivirus (EBV and AdV) specific T cell products [42]. This resulted in a higher frequency of AdV-reactive T cells in the products compared to the trivirus products. Thirteen patients, including one patient with SCID, received bivirus-specific T cell products in a phase I protocol. While AdV-specific T cells again only expanded in vivo in patients with concomitant AdV infections, none of these patients developed de novo AdV infections, suggesting that infused products had in vivo activity even if below the detectable level by ELISpot assay.
Despite these clinical successes, the time and cost associated with these prolonged culture methods limited their clinical use. Thus, several groups have subsequently developed rapid culture methods that have demonstrated safety and efficacy in clinical trials. Bao et al. used a rapid culture method using peptide pools for CMV pp65 and IE1 to produce CMV-specific T cell products from 10/10 healthy HSCT donors, and seven patients met eligibility criteria for infusion, four of whom were transplanted for immunodeficiencies [43]. Two of the PID patients had complete resolution of CMV infection, and one patient had a transient response but developed recurrence of CMV viremia after steroids were initiated for flare of HLH. The fourth patient had undetectable CMV-specific T cell function noted after infusion and persistent viremia, but the authors hypothesized that efficacy was diminished by the poor viability (60%) of the infused product as the patient subsequently demonstrated an increase in CMV-pp65-specific cytotoxic activity 3 weeks after receiving an unmodified DLI.
Papadopolou et al. demonstrated that a rapid ex vivo culture method can be used to generate VSTs specific for CMV, EBV, AdV, HHV6, and BK virus, although only 14 of 48 products generated had specificity for all five viruses [44••]. Four of the 11 patients treated had received an HSCT for PID conditions. All four patients had complete responses to targeted viruses, with the exception of one patient with HLH who had a complete response to HHV6 and EBV but no response to BK virus, which was not surprising as the infused product lacked specificity for BK. Overall, this study demonstrated a 94% response rate (15 CR; 2 PR) for the 18 viral infections/reactivations observed. Three patients who received VSTs for prophylaxis did not develop any viral complications.
Several previously unpublished cases were also included in a recent retrospective review by Naik et al. [45••]. Five patients with PID received trivirus-specific VST derived from their HSCT donor at our institution, including two with SCID, one with Wiskott-Aldrich Syndrome, and one with HLH due to STXBP2 deficiency. Three of these patients had existing viral infections (CMV in two, and CMV and EBV in 1), and two patients were treated prophylactically. Four of the five patients cleared their targeted infections and subsequent reactivations, while a fifth patient with SCID had progressive viral disease.
Patients Treated with “Third Party” VSTs
Third party banks of VSTs can further increase the availability of cellular therapy for patients with viral infections as they are not only readily available “off-the-shelf” but also provide therapeutic options for patients for whom custom-made products would not be available if they had a seronegative donor (e.g., cord blood). Although the experience using third party VSTs in PID patients has been limited thus far (Table 1), a multicenter phase I study demonstrated a 74% response rate in 50 patients who had undergone HSCT for various underlying diagnoses [46••]. Doubrovina et al. also described a series of 49 patients who were treated for EBV-lymphoproliferative disease following HSCT with donor lymphocyte infusion and/or EBV-specific VST that were either HSCT-donor derived or third party. Three patients in this series had PID disorders: one with autoimmune lymphoproliferative syndrome, one with X-linked lymphoproliferative disease, and another had primary HLH. The authors described an overall response rate of 68% following EBV-specific VST infusion, including a complete response in the patient with HLH who received third party EBV-specific T cells [24].
Table 1.
Reference | PID diagnosis | Infection | Specificity | VST stimulation | Source | Cell dose | Outcomes |
---|---|---|---|---|---|---|---|
Bao et al. [43] | Hyper IgM syndrome | CMV | CMV | Culture, peptide | HSCT donor, peripheral blood | 5 × 105/kg | Persistent viremia |
SCID | CMV | CMV | Culture, peptide | HSCT donor, peripheral blood | 5 × 105/kg | Resolution of viremia | |
CID | CMV | CMV | Culture, peptide | HSCT donor, peripheral blood | 2.5 × 105/kg | Resolution of viremia | |
Creidy et al. [51] | FHLH | Meningoecephlitis; retinitis | AdV | Selection by IFN-γ secretion | Haploidentical HSCT donor, leukaphresis | (1) 5 × 104/kg (2) 1.5 × 104/kg |
Alive, stabilization of retinitis |
SCID | Meningoecephlitis; retinitis | AdV | Selection by IFN-γ secretion | Haploidentical HSCT donor, leukaphresis | (1) 5 × 104/kg (2) 5 × 104/kg |
Resolution of viremia; stabilization of retinitis | |
FHLH | Pneumonitis; encephalitis; retinitis; viremia | CMV | Selection by IFN-γ secretion | HSCT donor, leukaphresis | 1.76 × 104/kg | Died at day +3 (alveolar hemorrhage) | |
FHLH | CMV viremia | CMV | Selection by IFN-γ secretion | Haploidentical HSCT donor, leukaphresis | (1) 3 × 104/kg (2) 5.88 × 104/kg (3) 1.99 × 105/kg |
Died at day +96; pulmonary arterial hypertension | |
CID | Retinitis | CMV | Selection by IFN-γ secretion | HSCT donor, leukaphresis | 5 × 104/kg | Alive but blind | |
Feuchtinger et al. [37] | HLH | AdV viremia, diarrhea | AdV | Selection by IFN-γ secretion | HSCT donor, peripheral blood | 5 × 104/kg | Clearance of AdV from blood and stool |
Leen et al. (2006) [34••] | SCID | Adeno | CMV, EBV, AdV | Culture, DC and LCL with Ad5f35f-CMV pp65 vector | HSCT donor, peripheral blood | 1 × 108/m2 | Clearance of AdV |
Leen et al. (2009) [42] | SCID | Prophylaxis | EBV, AdV | Culture, LCL with Ad5f35 vector | HSCT donor, peripheral blood | 1.35 × 108/m2 | Alive, no active infections |
Papadopolou et al. [44••] | LAD | Adenoviremia | CMV, EBV, AdV, HHV6, BK | Culture, peptide | Haploidentical HSCT donor, peripheral blood | 2 × 107/m2 | CR |
GATA2 | EBV, BK viremia | CMV, EBV, AdV, HHV6, BK | Culture, peptide | HSCT donor, peripheral blood | 2 × 107/m2 | CR | |
SCID variant | BK viremia; EBV viremia | CMV, EBV, AdV, HHV6, BK | Culture, peptide | HSCT donor, peripheral blood | 2 × 107/m2 | CR | |
HLH | HHV6, BK viremia | CMV, EBV, AdV, HHV6, BK | Culture, peptide | HSCT donor, peripheral blood | 1 × 107/m2 | HHV6: CR; BK: NR; EBV: CR | |
Vickers et al. [48] | CID | PTLD (EBV) | EBV | Culture, LCL | Third party | 1–2 × 106/kg/dose; 4 doses given weekly | CR |
CGD | PTLD (EBV) | EBV | Culture, LCL | Third party | 1–2 × 106/kg/dose; 4 doses given weekly | PD; died | |
CID | PTLD (EBV) | EBV | Culture, LCL | Third party | 1–2 × 106/kg/dose; 4 doses given weekly | PD; died | |
Wynn et al. [47] | CTPS1 | Primary CNS Lymphoma | EBV | Culture, LCL | Third party | 2 × 106/kg/dose; 7 doses given weekly; 2 additional doses given after re-emergence of EBV disease | CR |
Heslop et al. [40••] | WAS | EBV viremia | EBV | Culture, LCL | HSCT donor, peripheral blood | 2 × 107/m2 | CR |
NK defect/SCAEBV | EBV viremia | EBV | Culture, LCL | HSCT donor, peripheral blood | 1 × 108/m2 | CR | |
SCAEBV | EBV | EBV | Culture, LCL | HSCT donor, peripheral blood | 2 × 107/m2 | PR, died of progressive lymphoma | |
SCAEBV | EBV-LPD | EBV | Culture, LCL | HSCT donor, peripheral blood | 2 × 107/m2 | No further EBV reactivation | |
XLP (SLAM mutation) | EBV viremia | EBV | Culture, LCL | HSCT donor, peripheral blood | 2 × 107/m2 | CR | |
XLP and lymphoma | Prophylaxis | EBV | Culture, LCL | HSCT donor, peripheral blood | 2 × 107/m2 | No viremia | |
CID, enterocolitis | EBV-LPD (EBV viremia resolved) | EBV | Culture, LCL | HSCT donor, peripheral blood | 2.5 × 107/m2 | No further EBV reactivation | |
WAS | EBV-LPD (EBV viremia resolved) | EBV | Culture, LCL | HSCT donor, peripheral blood | 2.5 × 107/m2 | No further EBV reactivation | |
XLP | Resolved EBV viremia | EBV | Culture, LCL | HSCT donor, peripheral blood | 2 × 107/m2 | No further EBV reactivation | |
XLP | EBV viremia | EBV | Culture, LCL | HSCT donor, peripheral blood | 2 × 107/m2 | PR | |
XLP | Resolved EBV viremia | EBV | Culture, LCL | HSCT donor, peripheral blood | 2 × 107/m2 | CR | |
XLP | EBV-LPD with viremia | EBV | Culture, LCL | HSCT donor, peripheral blood | 2 × 107/m2 | PR | |
XLP-like | Resolved EBV viremia | EBV | Culture, LCL | HSCT donor, peripheral blood | 2 × 107/m2 | No further EBV reactivation | |
Doubrovina et al. [24] | HLH | EBV-LPD | EBV | Culture, LCL | HSCT donor, peripheral blood | 1 × 106/kg × 3 doses | PD; died |
XLP | EBV-LPD | EBV | Culture, LCL | Third Party | 1 × 106/kg × 3 doses | CR | |
ALPS | EBV-LPD | EBV | Culture, LCL | HSCT donor, peripheral blood | 1 × 106/kg | NE; died | |
Naik et al. [45••] | IL7RA-SCID | Prophylaxis | CMV, EBV, AdV | Culture, DC and LCL with Ad5f35f-CMVpp65 vector | HSCT donor, umbilical cord | 1.5 × 107/m2 | No viremia |
IL2RG-SCID | Prophylaxis | CMV, EBV, AdV | Culture, DC and LCL with Ad5f35f-CMVpp65 vector | HSCT donor, umbilical cord | 2.5 × 107/m2 | No viremia | |
IL2RG-SCID | Prophylaxis | CMV, EBV, AdV | Culture, DC and LCL with Ad5f35f-CMVpp65 vector | HSCT donor, umbilical cord | 1 × 107/m2 | No viremia | |
IL2RG-SCID | Prophylaxis | CMV, EBV, AdV | Culture, DC and LCL with Ad5f35f-CMVpp65 vector | HSCT donor | 1 × 107/m2 | No viremia | |
SCID | Prophylaxis | AdV | Culture, DC and LCL with Ad5f35 vector | HSCT donor | 1 × 107/m2 | No viremia | |
MHC II deficiency | CMV viremia, pneumonitis | CMV, EBV, AdV | Culture, DC and LCL with Ad5f35f-CMVpp65 vector | HSCT donor | 1 × 107/m2 | CR | |
WAS | Prophylaxis | CMV, EBV, AdV | Culture, DC and LCL with Ad5f35f-CMVpp65 vector | HSCT donor | 1 × 107/m2 | No viremia | |
CID, enterocolitis | Prophylaxis | EBV | Culture, peptide | HSCT donor | 2.5 × 107/m2 | No viremia | |
ADA-SCID | EBV-LPD | CMV, EBV, AdV | Culture | Third party | 5 × 106/m2 | NR, died from EBV lymphoma | |
SCID | CMV viremia | CMV, EBV, AdV | Culture | HSCT donor | 5 × 106/m2/dose; 2 doses | PR | |
SCID | (1) CMV viremia (2) CMV viremia and pneumonitis |
(1) CMV, EBV, AdV (donor) (2) CMV, EBV, AdV (third party) |
Culture | HSCT donor | (1) 1 × 107/m2 (2) 2 × 107/m2 |
NR; died; refractory CMV and disseminated BCG | |
MHC II Deficiency | Prophylaxis | CMV, EBV, AdV | Culture | HSCT donor | 5 × 106/m2 | CMV: CR | |
HLH (STXBP2) | CMV, EBV viremia | CMV, EBV, AdV | Culture | HSCT donor | 1 × 107/m2/dose × 2 doses | CMV: CR; EBV: CR | |
WAS | Prophylaxis | CMV, EBV, AdV | Culture | HSCT donor | 2 × 107/m2 | ||
IL-10R deficiency | AdV viremia, pneumonitis | CMV, EBV, AdV | Culture | Third party | 2 × 107/m2 | AdV: CR; CMV: CR | |
CTPS1 | EBV-LPD | EBV | Culture | Third party | 2 × 106/kg/dose × 2 doses | CR | |
HLH | EBV viremia | EBV | Culture | Third party | 2 × 106/kg/dose × 3 doses | EBV: PR; died; PTLD | |
CD4 lymphopenia | CMV viremia | CVM | Multimer selection | HSCT donor | 2 × 106/kg | CMV: PR; died of fungal pneumonia |
SCID severe combined immunodeficiency, CID combined immunodeficiency, FHLH familial hemophagocytosis lymphohistiocytosis, LAD leukocyte adhesion deficiency, CGD chronic granulomatous disease, WAS Wiskott-Aldrich Syndrome, SCAEBV severe chronic active Epstein-Barr virus infection, XLP X-linked lymphoproliferative syndrome, ALPS autoimmune lymphoproliferative syndrome, CTPS1 cytidine-5-prime-triphosphate synthetase, CR complete response, PR partial response, PD progressive disease, NR no response, NE not evaluable
Third party products may be of potential use pre-HSCT for PID patients, who are particularly susceptible to viral infections even before HSCT. Among the patients reviewed by Naik et al., two patients with PID (one with SCID and another with CTPS1 deficiency) were treated for persistent viral infections with VST derived from partially HLA-matched third party donors prior to HSCT. The latter patient, who had EBV-associated CNS lymphoma, had complete resolution of the lymphoma and clearance of EBV viremia after receiving EBV-specific VST prior to umbilical cord blood transplantation [47, 48].
Patients Treated with VST Derived Via Selection Technology
Several groups have utilized selection technology to treat viral infections in patients with PID following HSCT. Feuchtinger et al. treated nine patients with AdV-specific T cells isolated with IFN-γ capture, including one patient with HLH. Five out of six evaluable patients including the patient with HLH had in vivo expansion of the infused VST and durable antiviral responses [49]. These successes occurred in spite of low cell doses (1.2–5 × 10 [3] cells/kg) obtained by selection methods. Qasim et al. treated five patients with adenoviremia post-HSCT (including one with combined immunodeficiency, one with X-linked SCID, and one with MHC class-II deficiency) with AdV-specific VST derived via their HSCT donor or a parental donor [50]. Two of the treated patients had resolution of AdV infection and survived, while three died (two due to viral progression and one due to GVHD).
A recent French multicenter pilot study (NCT01325636) treated 15 patients after HSCT with CMV and/or AdV infections with VSTs generated by IFN-γ capture [51]. Five patients in this cohort had undergone HSCT due to PID. Three of the five PID patients demonstrated in vivo expansion of VSTs at day +21 of which two patients had stabilization of their retinitis while the third patient remained alive but blind. Two additional PID patients died of pulmonary complications in the setting of ongoing viral infections. Of those treated, one patient developed grade III GVHD in the 3 weeks after VST infusion, and another patient developed chronic GVHD.
Feucht et al. treated thirty patients with AdV infections, including seven with immunodeficiency, with hexon-specific T cells isolated by IFN-γ capture [52]. Twenty-one patients responded to VST infusion, including complete viral clearance in 18 patients and 3 partial responses (>1 log decrease in viral load without clearance). Two of the responders still died from AdV infection, and all eight of the non-responders died from progressive AdV disease. In this cohort, two patients developed grade I GVHD after VST infusion.
Potential Risks of VSTs
Multiple phase I studies have shown VSTs to be safe and well-tolerated. Infusion reactions are uncommon and mild and likely related to the cryopreservation additive rather than the VST themselves. While unmanipulated DLIs are capable of restoring virus-specific immunity after HSCT [30], this approach increases the risk of GVHD [22]. In contrast, the use of ex vivo expansion or direct selection methodologies to generate VSTs decreases the GVHD risk by reducing the alloreactive cells in the final product [21, 53]. In the largest retrospective review of VSTs in PID patients, only 4 of 36 patients (11%) had evidence of GVHD. Three patients developed GVHD in the setting of weaning of immunosuppression and responded well to steroid therapy. As control groups were not built into any phase I protocols to date, it is unclear if the rate of GVHD in those receiving VST therapy is different from the background rate of GVHD in patients undergoing HSCT. The risk of GVHD has been similarly low even when VSTs are only partially HLA matched such as those derived from haploidentical or third party donors [27, 46••].
Cytokine release syndrome (CRS), which can present with fever, hypotension, and multi-organ dysfunction due to immune-mediated inflammation, is another potential risk of antiviral cellular therapy. CRS has been well-described in the setting of chimeric-antigen receptor (CAR) T cell therapy and is associated with elevations in serum cytokines including IL-6, TNFα, and IFNγ [54]. Rooney et al. described inflammatory complications requiring mechanical ventilation in one patient with bulky EBV-related disease, who subsequently had a complete remission [41]. To date, CRS has not been described in patients with PID who have been treated with VST therapy, though it remains a theoretical risk, particularly in patients with disseminated viral disease.
Limitations of Current VST Protocols
Though CMV, EBV, and AdV are the most common viral pathogens associated with morbidity and mortality after HSCT for PID, many other viral infections are a threat to patients with PID, both before and after transplantation [11, 55]. Papadopoulou et al. expanded VST therapy to include targeting of HHV6 and BK virus with clinical success [44••]. While increasing the number of viruses targeted with a single product is appealing, it raises concern for antigenic competition in which the resulting products would be skewed toward immunodominant viral antigens. However, while CMV-pp65 and CMV-IE1 are considered strongly immunogenic, Papadopoulou et al. showed that CMV was the least consistently recognized virus in their VST products (26 of 48 VSTs) while AdV was the most consistently recognized virus (45 of 48 VSTs). Indeed, viral specificity of VSTs seemed more closely associated with pre-existing donor immunity, as all 26 VSTs derived from CMV-seropositive donors were specific for CMV whereas none of the VSTs derived from CMV-seronegative donors showed specificity for CMV antigens. Moreover, they demonstrated that there was no significant difference in the number of AdV-reactive T cells in a product regardless of the number of viruses the product was specific for. Hence, they proposed that virus-specificity in the multivirus T cell products is dependent on the frequency of pre-existing virus-specific T cells in the donor.
In the setting of partially HLA-matched third party VST therapy, a comparison of clinical outcomes using “overall best HLA match” strategies versus the approach utilized by Leen et al., where antiviral restrictions of selected VST products were confirmed prior to use, suggests that confirmation of antiviral activity mediated through one or more shared HLA alleles is essential for clinical efficacy [46••]. Accordingly, an improved knowledge of the antiviral restrictions of a multitude of viral antigens will be necessary in order to extend third party therapy to a wider number of pathogens.
Future Directions
Extending to Other Viruses
As VSTs have shown to be safe and efficacious in multiple phase I studies, there is growing interest in targeting other pathogens with cellular therapy. Human parainfluenza (HPIV) is a common respiratory pathogen that causes mild disease in immune competent hosts but causes significant morbidity and mortality in immune-compromised patients such as those with PID, with HPIV3 being the most common serotype in the post-HSCT setting [11, 56]. Currently, no therapeutic options exist for HPIV infections other than supportive care. Preclinical studies demonstrated that HPIV3-specific T cells can be generated from healthy donors using a rapid ex vivo culture protocol using peptide stimulation [57]. Importantly, HPIV3 could be targeted alongside EBV, CMV, AdV, BK, and HHV6 using this same method without loss of HPIV3-specificity.
Patients undergoing HSCT receive routine antiviral prophylaxis against herpes simplex virus (HSV); however, patients remain at risk from reactivation or infection with drug-resistant strains. HSV-1-specific T cells have been generated from seropositive donors using a culture method with antigen-pulsed dendritic cells and demonstrated cytokine production and cytotoxicity against HSV-1-infected targets [58]. Both of these promising preclinical studies will hopefully be translated into the clinical trials in the near future.
Extending to Other Pathogens
While viruses are a significant cause of morbidity and mortality both pre- and post-HSCT in patients with PID, many patients with PID are also risk for invasive fungal and mycobacterial infections [4, 59, 60]. Perruccio et al. treated 10 patients after HSCT with Aspergillus-specific T cells, and 9 patients had resolution of invasive aspergillosis [61]. Admittedly, the importance of T cell immunity in combating invasive aspergillosis is still unclear, as previous studies have demonstrated robust T cell immunity against Aspergillus species in patients with chronic granulomatous disease [62]. Invasive mycobacterial infections are unfortunately often a presenting sign in patients with T cell deficiency who receive early immunization with the live Bacillus Calmette-Guerin (BCG) vaccine [63]. Smith et al. published a report of a patient with SCID and disseminated BCG, which persisted after unfractionated HSCT despite therapy with multiple antibiotics [64]. She was given multiple whole blood transfusions from a healthy, HLA-matched sibling who previously had been immunized with BCG. Gradual improvement in radiographic lesions was noted with no evidence of GVHD, and a strong proliferative response was detectable in response to purified protein derivative. This case demonstrated that adoptive immunotherapy for mycobacteria may be feasible, and given the problem of multidrug resistance in many species of mycobacterial, T cell therapy may be an extremely useful adjunctive therapy.
Optimizing Manufacturing
Despite improvements in manufacturing time and costs, cellular therapy with VSTs remains a limited therapeutic option as it is only available at specialized centers with Good Manufacturing Practice (GMP)-compliant facilities capable of generating clinical grade products and meeting regulatory guidelines. As the use of third party VST improves, the use of specialized centers as regional banks, as well as commercialization of T cell banks should dramatically improve the availability of “off the shelf” VST products. The use of commercially available selection kits may also widen the availability of T cell therapy.
Conclusions
While numerous phase I studies have demonstrated the efficacy and safety of VSTs, comparing outcomes from various studies is complicated not only by the various manufacturing methods but also by the variation in cell doses and viruses targeted. Moreover, patients with PID are an extremely heterogeneous population and have variable HSCT regimens. Thus, there exists a considerable need for larger, multi-institutional phase II studies and meta-analyses to determine optimal manufacturing methods, dosing, and timing as well as long-term outcomes for patients treated with VSTs. However, adoptive T cell immunotherapy will likely become an integral component of transplantation, particularly as advances are made in the range of targeted pathogens and in the understanding of essential T cell subsets that can rapidly and safely target infections in immunocompromised patients.
Acknowledgements
We would like to thank the staffs of the Divisions of Allergy and Immunology and Blood and Marrow Transplantation at Children’s National Medical Center, our collaborators at the Cell and Gene Therapy center at Baylor College of Medicine and other institutions, the Children’s Research Institute, and the Jeffrey Modell Foundation for their support of this work.
Footnotes
Conflict of Interest Drs. McLaughlin, Bollard, and Keller declare no conflicts of interest relevant to this manuscript.
Human and Animal Rights and Informed Consent This article does not contain any studies with human or animal subjects performed by any of the authors.
References
Papers of particular interest, published recently, have been highlighted as:
• Of importance
•• Of major importance
- 1.Bousfiha AA, Jeddane L, Ailal F, et al. A phenotypic approach for IUIS PID classification and diagnosis: guidelines for clinicians at the bedside. J Clin Immunol. 2013;33(6):1078–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bonilla FA, Khan DA, Ballas ZK, et al. Practice parameter for the diagnosis and management of primary immunodeficiency. J Allergy Clin Immunol. 2015. [DOI] [PubMed] [Google Scholar]
- 3.Buckley RH. Transplantation of hematopoietic stem cells in human severe combined immunodeficiency: longterm outcomes. Immunol Res. 2011;49(1–3):25–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Bustamante J, Boisson-Dupuis S, Abel L, Casanova JL. Mendelian susceptibility to mycobacterial disease: genetic, immunological, and clinical features of inborn errors of IFN-gamma immunity. Semin Immunol. 2014;26(6):454–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Gennery AR, Slatter MA, Grandin L, et al. Transplantation of hematopoietic stem cells and long-term survival for primary immunodeficiencies in Europe: entering a new century, do we do better? J Allergy Clin Immunol. 2010;126(3):602–610 e601–611. [DOI] [PubMed] [Google Scholar]
- 6.Worth AJ, Booth C, Veys P. Stem cell transplantation for primary immune deficiency. Curr Opin Hematol. 2013;20(6):501–8. [DOI] [PubMed] [Google Scholar]
- 7.Cuellar-Rodriguez J, Gea-Banacloche J, Freeman AF, et al. Successful allogeneic hematopoietic stem cell transplantation for GATA2 deficiency. Blood. 2011;118(13):3715–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Cuellar-Rodriguez J, Freeman AF, Grossman J, et al. Matched related and unrelated donor hematopoietic stem cell transplantation for DOCK8 deficiency. Biol Blood Marrow Transplant. 2015;21(6):1037–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Notarangelo LD, Gambineri E, Badolato R. Immunodeficiencies with autoimmune consequences. Adv Immunol. 2006;89:321–70. [DOI] [PubMed] [Google Scholar]
- 10.Milner JD, Vogel TP, Forbes L, et al. Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations. Blood. 2015;125(4):591–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hutspardol S, Essa M, Richardson S, et al. Significant transplantation-related mortality from respiratory virus infections within the first One hundred days in children after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2015;21(10):1802–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Odek C, Kendirli T, Dogu F, et al. Patients with primary immunodeficiencies in pediatric intensive care unit: outcomes and mortality-related risk factors. J Clin Immunol. 2014;34(3):309–15. [DOI] [PubMed] [Google Scholar]
- 13.•.Pai SY, Logan BR, Griffith LM, et al. Transplantation outcomes for severe combined immunodeficiency, 2000–2009. N Engl J Med. 2014;37(5):434–46. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates the impact of active infections on survival of HSCT in patients with severe combined immunodeficiency.
- 14.Kwan A, Abraham RS, Currier R, et al. Newborn screening for severe combined immunodeficiency in 11 screening programs in the United States. JAMA. 2014;312(7):729–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Lugthart G, Oomen MA, der Zijde CM J-v, et al. The effect of cidofovir on adenovirus plasma DNA levels in stem cell transplantation recipients without T cell reconstitution. Biol Blood Marrow Transplant. 2015;21(2):293–9. [DOI] [PubMed] [Google Scholar]
- 16.Biron KK. Antiviral drugs for cytomegalovirus diseases. Anticancer Res. 2006;71(2–3):154–63. [DOI] [PubMed] [Google Scholar]
- 17.Sellar RS, Peggs KS. Management of multidrug-resistant viruses in the immunocompromised host. Br J Haematol. 2012;156(5):559–72. [DOI] [PubMed] [Google Scholar]
- 18.Taylor RP, Lindorfer MA. Antigenic modulation and rituximab resistance. Semin Hematol. 2010;47(2):124–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Boeckh M, Ljungman P How we treat cytomegalovirus in hematopoietic cell transplant recipients. Blood. 2009;113(23):5711–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Brown JA, Boussiotis VA. Umbilical cord blood transplantation: basic biology and clinical challenges to immune reconstitution. Clin Immunol. 2008;127(3):286–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Leen AM, Tripic T, Rooney CM. Challenges of T cell therapies for virus-associated diseases after hematopoietic stem cell transplantation. Expert Opin Biol Ther. 2010;10(3):337–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hromas R, Cornetta K, Srour E, Blanke C, Broun ER. Donor leukocyte infusion as therapy of life-threatening adenoviral infections after T-cell-depleted bone marrow transplantation. Blood. 1994;84(5):1689–90. [PubMed] [Google Scholar]
- 23.Roddie C, Peggs KS. Donor lymphocyte infusion following allogeneic hematopoietic stem cell transplantation. Expert Opin Biol Ther. 2011;11(4):473–87. [DOI] [PubMed] [Google Scholar]
- 24.Doubrovina E, Oflaz-Sozmen B, Prockop SE, et al. Adoptive immunotherapy with unselected or EBV-specific T cells for biopsyproven EBV+ lymphomas after allogeneic hematopoietic cell transplantation. Blood. 2012; 119(11):2644–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hanley PJ, Keller MD, Martin Manso M, et al. A phase 1 perspective: multivirus-specific T-cells from both cord blood and bone marrow transplant donors. Cytotherapy. 2016;18(6):S8. [Google Scholar]
- 26.Bollard CM, Heslop HE. T cells for viral infections after allogeneic hematopoietic stem cell transplant. Blood. 2016;127(26):3331–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.O’Reilly RJ, Prockop S, Hasan AN, Koehne G, Doubrovina E. Virus-specific T-cell banks for ‘off the shelf’ adoptive therapy of refractory infections. Bone Marrow Transplant. 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Walter EA, Greenberg PD, Gilbert MJ, et al. Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. N Engl J Med. 1995;333(16): 1038–44. [DOI] [PubMed] [Google Scholar]
- 29.•.Riddell SR, Watanabe KS, Goodrich JM, Li CR, Agha ME, Greenberg PD. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science. 1992;257(5067):238–41. [DOI] [PubMed] [Google Scholar]; The first report of adoptive immunotherapy with Cytomegalovirus-specific CD8+ cytotoxic T cell clones to restore CMV-specific immunity after HSCT.
- 30.Papadopoulos EB, Ladanyi M, Emanuel D, et al. Infusions of donor leukocytes to treat Epstein-Barr virus-associated lymphoproliferative disorders after allogeneic bone marrow transplantation. N Engl J Med. 1994;330(17):1185–91. [DOI] [PubMed] [Google Scholar]
- 31.Rooney CM, Smith CA, Ng CY, et al. Use of gene-modified virus-specific T lymphocytes to control Epstein-Barr-virus-related lymphoproliferation. Lancet. 1995;345(8941):9–13. [DOI] [PubMed] [Google Scholar]
- 32.Heslop HE, Brenner MK, Rooney CM. Donor T cells to treat EBV-associated lymphoma. N Engl J Med. 1994;331(10):679–80. [DOI] [PubMed] [Google Scholar]
- 33.Cobbold M, Khan N, Pourgheysari B, et al. Adoptive transfer of cytomegalovirus-specific CTL to stem cell transplant patients after selection by HLA-peptide tetramers. J Exp Med. 2005;202(3):379–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.••.Leen AM, Myers GD, Sili U, et al. Monoculture-derived T lymphocytes specific for multiple viruses expand and produce clinically relevant effects in immunocompromised individuals. Nat Med. 2006;12(10):1160–6. [DOI] [PubMed] [Google Scholar]; This group demonstrated not only that multivirus-specific T cells can be generated using a single culture method but also that these T cell products are safe and effective when used to treat immunocompromised patients.
- 35.Gerdemann U, Keirnan JM, Katari UL, et al. Rapidly generated multivirus-specific cytotoxic T lymphocytes for the prophylaxis and treatment of viral infections. Mol Ther. 2012;20(8):1622–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Neudorfer J, Schmidt B, Huster KM, et al. Reversible HLA multimers (Streptamers) for the isolation of human cytotoxic T lymphocytes functionally active against tumor- and virus-derived antigens. J Immunol Methods. 2007;320(1–2): 119–31. [DOI] [PubMed] [Google Scholar]
- 37.Feuchtinger T, Lucke J, Hamprecht K, et al. Detection of adenovirus-specific T cells in children with adenovirus infection after allogeneic stem cell transplantation. Br J Haematol. 2005;128(4):503–9. [DOI] [PubMed] [Google Scholar]
- 38.Feucht J, Joachim L, Lang P, Feuchtinger T. Adoptive T-cell transfer for refractory viral infections with cytomegalovirus, Epstein-Barr virus or adenovirus after allogeneic stem cell transplantation. Klin Padiatr. 2013;225(3):164–9. [DOI] [PubMed] [Google Scholar]
- 39.Cohen JI. Primary immunodeficiencies associated with EBV disease. Curr Top Microbiol Immunol. 2015;390(Pt 1):241–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.••.Heslop HE, Slobod KS, Pule MA, et al. Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood. 2010;115(5):925–35. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrated long term persistence of EBV-specific T-cells following HSCT.
- 41.Rooney CM, Smith CA, Ng CY, et al. Infusion of cytotoxic T cells for the prevention and treatment of Epstein-Barr virus-induced lymphoma in allogeneic transplant recipients. Blood. 1998;92(5):1549–55. [PubMed] [Google Scholar]
- 42.Leen AM, Christin A, Myers GD, et al. Cytotoxic T lymphocyte therapy with donor T cells prevents and treats adenovirus and Epstein-Barr virus infections after haploidentical and matched unrelated stem cell transplantation. Blood. 2009;114(19):4283–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bao L, Cowan MJ, Dunham K, et al. Adoptive immunotherapy with CMV-specific cytotoxic T lymphocytes for stem cell transplant patients with refractory CMV infections. J Immunother. 2012;35(3):293–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.••.Papadopoulou A, Gerdemann U, Katari UL, et al. Activity of broad-spectrum T cells as treatment for AdV, EBV, CMV, BKV, and HHV6 infections after HSCT. Sci Transl Med. 2014;6(242):242ra283. [DOI] [PMC free article] [PubMed] [Google Scholar]; Using a rapid culture method, this group showed that VSTs that recognize 12 immunogenic antigens from 5 viruses (Epstein-Barr virus, adenovirus, cytomegalovirus, BK virus, and Human Herpesvirus 6) can be manufactured in less than two weeks. While only 14 of the 48 VST products had activity against antigens from all 5 viruses, the authors showed that viral specificity was related to prior viral exposure of the VST donors rather than a limitation in the manufacturing protocol. Safety and efficacy was demonstrated in 11 patients who had a 94% response rate.
- 45.••.Naik S, Nicholas SK, Martinez CA, et al. Adoptive immunotherapy for primary immunodeficiency disorders with virus-specific T lymphocytes. J Allergy Clin Immunol. 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]; This is the largest review of VSTs used in patients with primary immunodeficiencies that showed the safety and efficiacy of VSTs in this population.
- 46.••.Leen AM, Bollard CM, Mendizabal AM, et al. Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood. 2013;121(26):5113–23. [DOI] [PMC free article] [PubMed] [Google Scholar]; This is the largest phase II study of partially-HLA matched third-party VST therapy for treatment of viral infections following HSCT.
- 47.Wynn RF, Arkwright PD, Haque T, et al. Treatment of Epstein-Barr-virus-associated primary CNS B cell lymphoma with allogeneic T-cell immunotherapy and stem-cell transplantation. Lancet Oncol. 2005;6(5):344–6. [DOI] [PubMed] [Google Scholar]
- 48.Vickers MA, Wilkie GM, Robinson N, et al. Establishment and operation of a Good Manufacturing Practice-compliant allogeneic Epstein-Barr virus (EBV)-specific cytotoxic cell bank for the treatment of EBV-associated lymphoproliferative disease. Br J Haematol. 2014;167(3):402–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Feuchtinger T, Matthes-Martin S, Richard C, et al. Safe adoptive transfer of virus-specific T-cell immunity for the treatment of systemic adenovirus infection after allogeneic stem cell transplantation. Br J Haematol. 2006;134(1):64–76. [DOI] [PubMed] [Google Scholar]
- 50.Qasim W, Gilmour K, Zhan H, et al. Interferon-gamma capture T cell therapy for persistent adenoviraemia following allogeneic haematopoietic stem cell transplantation. Br J Haematol. 2013;161(3):449–52. [DOI] [PubMed] [Google Scholar]
- 51.Creidy R, Moshous D, Touzot F, 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(3):920–4. e923. [DOI] [PubMed] [Google Scholar]
- 52.Feucht J, Opherk K, Lang P, et al. Adoptive T-cell therapy with hexon-specific Th1 cells as a treatment of refractory adenovirus infection after HSCT. Blood. 2015;125(12):1986–94. [DOI] [PubMed] [Google Scholar]
- 53.Melenhorst JJ, Leen AM, Bollard CM, et al. Allogeneic virus-specific T cells with HLA alloreactivity do not produce GVHD in human subjects. Blood. 2010;116(22):4700–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lee DW, Gardner R, Porter DL, et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124(2):188–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Crooks BN, Taylor CE, Turner AJ, et al. Respiratory viral infections in primary immune deficiencies: significance and relevance to clinical outcome in a single BMT unit. Bone Marrow Transplant. 2000;26(10):1097–102. [DOI] [PubMed] [Google Scholar]
- 56.Karron RA, Collins PL. Parainfluenza viruses. In: Knipe DM, Howley PM, editors. Fields virology, vol. 1. Philadelphia: Lippincott Williams & Wilkins; 2013. p. 996–1023. [Google Scholar]
- 57.McLaughlin LP, Lang H, Williams E, et al. Human parainfluenza virus-3 can be targeted by rapidly ex vivo expanded T lymphocytes. Cytotherapy. 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ma CK, Clancy L, Deo S, Blyth E, Micklethwaite KP, Gottlieb DJ. Herpes simplex virus type 1 (HSV-1) specific T-cell generation from HLA-A1- and HLA-A2-positive donors for adoptive immunotherapy. Cytotherapy. 2016 [DOI] [PubMed] [Google Scholar]
- 59.Hsu AP, Sampaio EP, Khan J, et al. Mutations in GATA2 are associated with the autosomal dominant and sporadic monocytopenia and mycobacterial infection (MonoMAC) syndrome. Blood. 2011;118(10):2653–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hanson EP, Monaco-Shawver L, Solt LA, et al. Hypomorphic nuclear factor-kappaB essential modulator mutation database and reconstitution system identifies phenotypic and immunologic diversity. J Allergy Clin Immunol. 2008;122(6):1169–77. e1116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Perruccio K, Tosti A, Burchielli E, et al. Transferring functional immune responses to pathogens after haploidentical hematopoietic transplantation. Blood. 2005;106(13):4397–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Cruz CR, Lam S, Hanley PJ, et al. Robust T cell responses to aspergillosis in chronic granulomatous disease: implications for immunotherapy. Clin Exp Immunol. 2013;174(1):89–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Marciano BE, Huang CY, Joshi G, et al. BCG vaccination in patients with severe combined immunodeficiency: complications, risks, and vaccination policies. J Allergy Clin Immunol. 2014;133(4):1134–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Smith LL, Wright BL, Buckley RH. Successful treatment of disseminated BCG in a patient with severe combined immunodeficiency. J Allergy Clin Immunol Pract. 2015;3(3):438–40. [DOI] [PMC free article] [PubMed] [Google Scholar]