Next Article in Journal
The Worm-Specific Immune Response in Multiple Sclerosis Patients Receiving Controlled Trichuris suis Ova Immunotherapy
Next Article in Special Issue
Anti-Cancer and Immunomodulatory Activity of a Polyethylene Glycol-Betulinic Acid Conjugate on Pancreatic Cancer Cells
Previous Article in Journal
Structure, Function, and Interactions of the HIV-1 Capsid Protein
Previous Article in Special Issue
Cryptococcal Immune Reconstitution Inflammatory Syndrome: From Blood and Cerebrospinal Fluid Biomarkers to Treatment Approaches
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Reconstitution after Haploidentical Donor and Umbilical Cord Blood Allogeneic Hematopoietic Cell Transplantation

1
Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL 33612, USA
2
Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
*
Author to whom correspondence should be addressed.
Submission received: 31 December 2020 / Revised: 19 January 2021 / Accepted: 25 January 2021 / Published: 29 January 2021
(This article belongs to the Special Issue Immune Reconstitution Disorders)

Abstract

:
Allogeneic hematopoietic cell transplantation (HCT) is the only potentially curative therapy for a variety of hematologic diseases. However, this therapeutic platform is limited by an initial period when patients are profoundly immunocompromised. There is gradual immune recovery over time, that varies by transplant platform. Here, we review immune reconstitution after allogeneic HCT with a specific focus on two alternative donor platforms that have dramatically improved access to allogeneic HCT for patients who lack an HLA-matched related or unrelated donor: haploidentical and umbilical cord blood HCT. Despite challenges, interventions are available to mitigate the risks during the immunocompromised period including antimicrobial prophylaxis, modified immune suppression strategies, graft manipulation, and emerging adoptive cell therapies. Such interventions can improve the potential for long-term overall survival after allogeneic HCT.

1. Introduction

Allogeneic hematopoietic cell transplantation (HCT) offers the only potential cure for many high-risk hematologic malignancies. The therapeutic benefit of allogeneic HCT is, in part, due to an alloreactive graft-versus-tumor (GVT) response whereby the donor immune system recognizes the recipient tumor cells as foreign and eradicates them [1]. However, this same alloreactivity can also lead to toxicity such as graft-versus-host disease (GVHD) where the donor immune system attacks the recipient, or graft rejection where the recipient immune system attacks the donor cells [2,3]. Control of these bidirectional immune responses requires modulation of the lymphodepleting conditioning regimen and immune suppressive therapies (IST) which decrease the risks of graft rejection and GVHD, but also increase risks of infection [4,5,6,7]. Antimicrobial prophylaxis and vaccination, thus, also become critical elements of successful HCT [8].
In addition to IST, selection of a human leukocyte antigen (HLA) matched donor has long been considered a critical consideration to mitigate the risks of toxic alloreactivity and consequent transplant related mortality (TRM) [9,10]. Historically, HLA matched sibling donors (MSD) have been the preferred donor option followed by HLA matched unrelated donors (MUD). However, due to the Mendelian inheritance pattern of HLA haplotypes, the likelihood of a patient’s each sibling being a full HLA match is 25% and the likelihood of identifying a MUD varies from 10–80% depending on the ethnic and racial background of the patient [11]. To improve access to HCT for patients lacking an HLA matched donor, alternative donor platforms such as haploidentical (haplo) related donor HCT and umbilical cord blood transplant (UCBT) have been developed [12]. Specifically, haplo HCT with use of posttransplant cyclophosphamide (PTCy) has emerged as a favorable strategy that yields rates of GVHD, TRM, and overall survival (OS) that are nearly equivalent to matched donor HCT [13,14]. Further, in a recent Blood and Marrow Transplant Clinical Trials Network (BMT CTN) phase III trial, haplo HCT with PTCy was well tolerated and resulted in better overall survival (OS) compared to UCBT [15]. Nonetheless, both approaches are considered appropriate alternative donor HCT strategies and the choice of haplo versus UCBT is still largely dependent on institutional expertise [13].
The feasibility of modern approaches to HCT, including expansion of the donor pool to haplo and UCBT, requires unique manipulations to the immune systems of both donor and recipient cells to allow for successful donor engraftment and prevention of GVHD. However, these manipulations can result in profound effects on immunity that impact rates of infection and relapse of the primary malignancy [13,15]. Here, we describe the immune reconstitution after allogeneic HCT in general, special considerations for haplo HCT and UCBT, as well as resultant impacts on clinical outcomes and considerations for management, particularly in the context of hematologic malignancies.

2. Kinetics of Immune Reconstitution after Allogeneic HCT

In practice, allogeneic HCT consists of a conditioning regimen (chemotherapy and/or total body irradiation (TBI)) to suppress recipient alloreactivity against the donor and allow engraftment of the donor hematopoietic stem cells, followed by infusion of the donor hematopoietic stem cells and subsequent initiation of IST. This procedure acutely results in severe immune compromise followed by gradual immune reconstitution [16]. While immune reconstitution varies depending on the specific transplant platform, there are uniform patterns that inform the likelihood of specific immunologic complications over time.

2.1. Early Immune Reconstitution

The conditioning regimen given prior to transplant results in profound cytopenias that nadir in the week following stem cell infusion. This results in depletion of both innate and adaptive immunity. During this period, susceptibility is high to bacterial and fungal infections so antibacterial and antifungal prophylaxis is standard [8]. In general, the innate immune system begins to recover first. Monocyte engraftment begins in the two weeks after transplant followed closely by neutrophil recovery [17]. Concurrently, non-hematopoietic innate immunity, such as mucosal barriers, heal from injury caused by the conditioning regimen [17]. At most centers, the resolution of neutropenia and mucosal injury are key landmarks required for hospital discharge and discontinuation of antibacterial and antifungal prophylaxis. In the weeks following neutrophil engraftment, natural killer (NK) cells recover [18,19,20,21,22,23]. These cells are increasingly recognized as essential to the GVT effect that prevents disease relapse [20,21,22,23].

2.2. Late Immune Reconstitution

Though the innate immune system quantitatively recovers in the first weeks after HCT, these cells may not be functionally competent. Indeed, functional recovery of the hematopoietic innate immune system typically occurs in the 4–12 months after transplant [17,24,25]. The adaptive immune system, including the cellular immune response and humoral immune response, requires functional T-lymphocytes and B-lymphocytes. These cells begin recovering in the months after transplant but may require years to reach full competence [26,27,28].
The recovery of the cellular immune response, of particular importance for immunity against viral pathogens and graft-versus-tumor, occurs in two phases. First, immunocompetent T-cells in the donor graft may undergo clonal expansion [16,29]. Second, naïve T-cells from the donor may be expanded in the thymus of the recipient [16,29]. The humoral immune response resulting in adequate antibody response, requires recovery of T-cells as well as functional B-cells, which recover between 3 months–1 year after transplant [17,30]. It is during this phase of recovery that post-HCT vaccinations are typically initiated [8].

2.3. Factors Influencing Immune Reconstitution

The expected immune reconstitution described is general and may vary considerably between individual patients due to both modifiable and fixed aspects of HCT [17]. First, advanced age of either the donor or the recipient can result in slower immune recovery due slower engraftment with aging marrow [31,32]. Further, T-cell immune reconstitution requires a functional thymus and, thus, may be significantly limited in older patients who can have thymic atrophy [29]. HLA matched donors have also been shown to yield better immune reconstitution, possibly because HLA mismatched may lead to more mixed lymphocyte reactions and host-versus-graft alloreactivity that can delay immune reconstitution [25,33,34]. More intensive conditioning regimens can result in more rapid engraftment, though may also slow early lymphocyte recovery [35]. Graft related factors may also contribute to immunity, with peripheral blood stem cells grafts resulting in more rapid engraftment than marrow grafts [17,36]. Grafts with higher stem cell dose or higher T-cell content also may engraft more quickly, resulting in more rapid immune reconstitution [37,38,39]. Conversely, T-cell depletion of the graft, often used for GVHD prevention, results in higher rates of graft rejection as well as slower immune reconstitution and increased risk of infections [40,41]. Finally, the GVHD prophylactic regimen, its duration of administration, and onset of acute or chronic GVHD are all associated with impaired immune reconstitution [3,8,42].

2.4. Clinical Significance of Immune Reconstitution

The kinetics of immune reconstitution correlate temporally with expected transplant related complications. In the period preceding donor cell engraftment, the marrow is aplastic. The resultant profound neutropenia leads to a period of high risk for bacterial and fungal infections, generally in the first 30 days after HCT [43,44]. Between days 30–100 after HCT, as cell mediated immunity slowly recovers, the highest risk infections shift towards viral reactivation such as cytomegalovirus (CMV), human herpesvirus 6 (HHV–6), or Epstein–Barr virus (EBV), in addition to pneumocystis pneumonia (PCP) [8,45,46]. During this period, acute GVHD, a T-cell mediated process, also emerges, occurring in ~20–50% of HCT patients and can result in skin rashes, gastrointestinal toxicity, hepatic injury, infections, and mortality [4,5,47].
Beyond day 100, though infectious immunity steadily improves, chronic GVHD will occur in up to 60% of HCT patients, though may be lower with modern approaches even with a haplo, and cord blood transplant [3,4,5,15,47,48]. The occurrence of chronic GVHD is a risk factor for subsequent infections, due to both inherent immune dysregulation as well as increases in IST to control chronic GVHD [49].
In addition to transplant related toxicity, post-HCT immune reconstitution is necessary to prevent relapse and cure the underlying hematologic malignancy through the immunologic GVT effect. The GVT effect became clinically apparent in studies showing greater HLA disparity resulted in reduced risks of relapse, while genetically similar identical twin donors result in higher risks of relapse [1]. It is now understood that cytotoxic T-cells are critical to the GVT effect, eliminating tumor cells through secretion of granzyme B as well as apoptosis via FAS ligands [50]. The significance of T-cells to the GVT effect has been demonstrated clinically as donor lymphocyte infusions are able to eradicate active tumor, while T-cell depletion results in higher risks of relapse [40,51]. The GVT effect can be triggered by HLA mismatch, as well as host minor histocompatibility antigens and tumor related neoantigens [52]. Thus, the ability of the cytotoxic T-cells to distinguish healthy host tissue from tumor cells is limited, and GVT and GVHD often overlap.
In recent years, there has been increasing focus on the role of NK cells as mediators for the GVT effect [53]. NK cell function is dependent upon receptor/ligand interactions resulting in activating signals or inhibitory signals [54,55,56]. The balance of activation and inhibition leads to either cell killing or tolerance [55,56]. Interactions between the NK cell killer immunoglobulin-like receptor (KIR) with self HLA class I molecules, which are expressed uniformly on healthy host tissue, lead to inactivation [20,21,57,58]. In the setting of allogeneic HCT, certain combinations of donor activating KIR types and recipient HLA subtypes promote NK cell activation, leading to a more potent GVT effect without an increase in GVHD [53,54,59,60,61,62,63,64]. An ongoing prospective, multicenter trial is evaluating the utility of incorporating donor KIR type in selection of allogeneic HCT donors for patients with acute myeloid leukemia (NCT02450708). Further, the ability of tumor cells to down regulate HLA class I creates a mechanism by which NK cells can differentiate between tumor and healthy host tissue, thus leading to activation and tumor killing [65,66,67].

3. Haploidentical Donor Transplant

Because of the inheritance patterns of HLA haplotypes, parents and children will be HLA haploidentical matches and siblings have a 50% likelihood of being haploidentical matches. As a result, patients in need of transplant have a >90% likelihood of having a suitable HLA haploidentical related donor [68]. However, the significant HLA mismatch between the haplo donor and the recipient results in intense bidirectional alloreactivity whereby the donor immune system attacks the recipient (GVHD) and the recipient immune system attacks the donor cells (graft rejection) [69]. Early studies of haplo transplant, thus, resulted in unacceptable toxicity that precluded the use of this strategy for many years [69].

3.1. Approaches to Haploidentical Transplant

The acute alloreactivity that occurs after haplo HCT is mediated primarily by donor and recipient T-lymphocytes. Multiple regimens have been developed to target T-cell function to mitigate toxicity. Three strategies have become the most utilized: (1) high-dose PTCy; (2) ex vivo T-cell depletion (TCD) with “megadose” CD34+ cells; and (3) the ”GIAC” regimen (GCSF-stimulation of the donor; intensified immunosuppression through post–transplantation CsA, mycophenolate mofetil (MMF), and short-course methotrexate; antithymocyte globulin (ATG) added to conditioning to help prevent GVHD and aid engraftment; and combination of PBSC and bone-marrow allografts) [14,70,71].

3.2. Immune Reconstituion after Haploidentical Transplant

Because of the higher risks of GVHD with HLA mismatched donors, the GVHD prophylaxis regimens used for haplo donor HCT are more immune suppressive than those used in matched donor HCT. Haplo HCT with PTCy has emerged as the haplo platform of choice in the United States. Immune reconstitution with this regimen has been compared retrospectively to matched donor and UCBT [72]. Compared to the MSD group, the haplo with PTCy group had a higher risk of CMV viremia (58% versus 74%), fungal infection (4% versus 11%), and infection related death (4% versus 11%). At day 100, median CD4+ lymphocyte count was 229/mm3 for the MSD group and 190/mm3 for the haplo group. Despite these differences in immune recovery, TRM was similar between the groups [72]. Similarly, in our experience at the Moffitt Cancer Center, the recovery of total absolute CD4+ T cell count after haplo and MUD HCT with PTCy was significantly lower compared to MUD with calcineurin inhibitor (CNI)-based GVHD prophylaxis throughout 1 year of HCT [73]. In contrast, the total CD8+ T cell recovery was similar in all groups. A recent retrospective registry analysis by the Center for International Blood and Marrow Transplant Research compared haplo HCT with PTCy to matched donor HCT with PTCy to matched donor HCT with CNI for GVHD prophylaxis. Both PTCy groups had higher risks of CMV viremia, suggesting PTCy is an independent risk factor for CMV viremia regardless of donor type [74].
The TCD strategy results in the highest risks of infection with trials showing ~27% of treated patients dying of infection, a rate that is higher than all–cause transplant related mortality with many other platforms [70]. The most common infections reported were CMV and aspergillus. However, subsequent studies with this platform have shown potential for adoptive transfer of infection specific T-cells or regulatory T-cells to improve immune reconstitution and decrease infections [75,76].
Immune reconstitution of haplo HCT with the GIAC regimen has been prospectively compared to matched donor transplants [77]. In that study, survival outcomes were similar between the two groups, though CMV viremia was more prevalent in the haplo group: 50% versus 13% (p = 0.007). Compared to the matched donor group, the haplo group was noted to have decreased T-cell subsets and dendritic cells at day 90, with the most significant decreases observed in the CD4+ T-cells. Notably, B-cell recovery and monocyte recovery were similar between the two groups.

3.3. Graft-Versus-Tumor Effect after Haploidentical Transplant

Though HLA disparity is known to elicit a more potent GVT effect after transplant, rates of relapse after modern haplo donor HCT are similar or even higher than matched donor transplants in several reports [72,77,78]. The reasons for this are possibly related to the other components of the transplant platform such as intensity of the immune suppression associated with these regimens, as well as low intensity conditioning and/or bone marrow graft source often used in conjunction with haplo HCT with PTCy [79,80,81]. Additionally, relapse after haplo HCT often occurs, at least in part, through a unique mechanism through which the mismatched haplotype is eradicated from the tumor cells as a form of antigen escape [82,83,84]. This phenomenon, called “loss of heterozygosity,” occurs in up to 30% of relapses after haplo donor HCT but is rarely encountered in matched donor HCT [83,85,86]. As such, loss of heterozygosity is indirect evidence that the GVT effect in haplo donor HCT is driven by immune recognition of the HLA mismatch.

4. Umbilical Cord Blood Transplantation

No risk to the donor, rapid availability, less restrictive HLA-matching selection criteria, and low risk of chronic GVHD are well-recognized advantages of UCBT [87,88,89,90]. Thus, UCB as an alternative donor option has had large utilization in the past two decades offering curative allogeneic HCT to racial and ethnic minorities with various hematological malignancies. Conversely, the limitations of UCBT include delayed hematopoietic engraftment and immune reconstitution, leading to higher risks of infections and TRM after HCT [91,92,93,94,95]. The introduction of double UCBT and RIC further extended the access and made this alternative donor HCT option available to many adults with hematological malignancies [88,90,96,97,98]. However, slow immune reconstitution and higher frequency of infections still remain major obstacles to the successful use of UCB source [92,94].

4.1. Quantitative Immune Reconstitution after UCBT

We previously compared the pace of immune reconstitution after UCB (n = 89) and MSD peripheral blood (n = 68) allo HCT in patients receiving similar RIC (consisting of fludarabine (Flu), Cy and TBI) and GVHD prophylaxis [92]. Despite lower absolute numbers of total NK cells and individual NK cell subsets at day 28 after UCBT, their absolute numbers were significantly higher after UCBT compared to MSD HCT as early as day 60 after HCT. Similarly, despite lower absolute B cell count at day 28 the numbers of B cells were significantly higher at day 100 after UCBT compared to MSD HCT. Conversely, UCBT was associated with significantly slower recovery of CD8+ and CD4+ T cell subsets as compared to MSD HCT. While the numbers of most CD4+ T cell subsets (central memory, effector memory and regulatory) were lower after UCBT within only the first 100 days of HCT, the naïve CD4+ T cell count remained low throughout 6 months after HCT. For the CD8+ T cells subsets, the central memory CD8+ T-cell count was lower within the 100 days whereas the naïve and effector memory CD8+ T-cell counts remained significantly lower throughout 6 months after UCBT compared to MSD. The use of ATG had no significant impact on immune reconstitution in our analysis. We observed significantly higher frequency of viral infections within first 180 days and bacterial infections within first 60 days after UCBT compared to MSD HCT [92]. A similar pattern of more robust recovery of NK cells and B cells but slower recovery of T-cell immune subsets is reported after RIC UCBT as compared to MUD HCT [95].
Other studies also reported this distinct pattern of immune cell count recovery after either myeloablative or RIC UCBT [91,92,93,94,99].

4.2. Virus-Specific Immune Reconstitution after UCBT

Prior studies were largely focused on quantitative immune cell count recovery after UCBT. We recently compared virus-specific immune reconstitution after UCBT and MSD peripheral blood HCT in patients receiving the same RIC regimen with Flu, Cy, TBI and no ATG [100]. Interferon-gamma (IFN-γ) enzyme-linked immune absorbent spot assay (ELISpot), which was used to quantify the frequencies of IFN-γ-secreting peripheral blood mononuclear cell (PBMC), identified higher frequencies of CMV-specific PBMCs after HCT in CMV seropositive patients compared to CMV seronegative patients. However, the frequencies of CMV-reactive PBMCs in CMV seropositive recipients were significantly lower after UCBT compared to MSD HCT throughout the first 12 months after transplant. These findings suggest that higher rates of CMV reactivation/infection after UCBT are explained not only by delayed quantitative recovery of immune cells but also by slower recovery of CMV-specific immunity after UCBT compared to MSD HCT. The reconstitution of other virus-specific immunity (HHV6, EBV, BK and adenovirus) was not significantly different between the two donor types in our analysis [100]. Another prior study reported high rates of CMV, BK and adenovirus infections after myeloablative conditioning UCBT. The authors reported significant delay (up to 12 months) in recovery of virus–reactive PBMCs against CMV, EBV, BK, adenovirus, influenza and RSV antigens [94]. However, all patients in that study also received ATG in addition to myeloablative conditioning.
While CMV is the most frequently reported viral reactivation/infection after UCBT we also observed higher frequency of HHV6 reactivation/infection after UCBT with use of sirolimus instead of cyclosporine in combination with MMF as GVHD prophylaxis (51% vs. 20%; p < 0.01 by day +45) [98]. HHV6 reactivation is generally an earlier event after UCBT (median onset of 26 days) and can be associated with primary graft failure after RIC UCBT [98]. Introduction of antiviral prophylaxis with foscarnet from day +7 through neutrophil engraftment after UCBT delayed the time to HHV6 reactivation and resulted in higher neutrophil engraftment rates in our recent report [101].

5. Interventions to Mitigate Complications of Immune Deficiency after Allogeneic HCT

5.1. Prevention of Bacterial Infection

The first month of allogeneic transplant lends to a high risk of bacterial infections due to severe neutropenia and breakdown of mucosal barriers [8]. The primary pathways of entrance for these infections are translocation of oral/intestinal flora due to mucosal injury, or transmission of skin flora through indwelling catheters or skin breakdown [8]. Thus, both Gram-positive and Gram-negative bacteria are implicated, though Gram-negative bacteremia results in especially rapid clinical decline [102]. Because of this, antibacterial prophylaxis should be considered for all patients undergoing allogenic HCT [8]. Fluoroquinolones are the preferred agents based on prospective data and meta–analyses demonstrating improvements in infection related mortality and overall survival [103,104]. While ciprofloxacin is acceptable, levofloxacin is preferred in patients with poor dentition or high risk of mucosal injury given effectiveness against oral strep viridans [8]. In patients intolerant to fluoroquinolones, a recent retrospective study suggested similar efficacy with cefpodoxime as an alternative agent [105]. Antibacterial prophylaxis should be continued until neutropenia resolves, generally 2–3 weeks after the stem cell infusion. Notably, these general recommendations should be modified based on the local bacterial resistance patterns [106]. During the period of neutropenia, most centers also monitor closely in the inpatient setting. In the case of neutropenic fevers, broad spectrum antibiotics with pseudomonal coverage (e.g., piperacillin–tazobactam or cefepime) must be initiated within one hour of fever onset to decrease the risk of septic shock and mortality [107].
Though the risk of severe bacterial infection decreases after engraftment, patients with chronic GVHD are at high risk for encapsulated bacterial infections such as Neisseria meningitides or Streptococcus pneumoniae [108]. Therefore, prophylaxis with penicillin should be considered for patients on systemic glucocorticoid therapy for chronic GVHD.

5.2. Prevention of Fungal Infection

Fungal infections are common in the first month after HCT but the risk continues beyond engraftment, particularly in patients who develop GVHD [109]. The role for antifungal prophylaxis early after transplant is clear, with studies demonstrating significantly improved overall survival [110]. Candida and invasive molds, such as aspergillus, are most problematic, which would suggest prophylaxis against both is necessary. However, large, well conducted studies have demonstrated that prophylaxis against mold and candida with posaconazole or voriconazole does not reduce risks of invasive fungal infections nor improve overall survival compared to prophylaxis against candida with fluconazole [111,112]. Thus, candida prophylaxis with fluconazole through neutrophil engraftment is considered adequate for most allogeneic HCT patients. The echinocandins are also effective against candida species, with a broader spectrum than fluconazole, and may be substituted based on local resistance patters or side effect profiles [8,109]. However, mold coverage with voriconazole or posaconazole is implemented for patients with risk factors such as prolonged neutropenia or presence of lung nodules prior to transplant [8]. For patients who develop GVHD and require high doses of IST or prednisone (≥0.5 mg/kg), anti-fungal prophylaxis should be reinitiated [108]. In a randomized controlled phase III trial, posaconazole was superior to fluconazole in preventing invasive aspergillosis and reducing the mortality related to fungal infections in patients with GVHD who require systemic IST [113].
Pneumocystis jiroveci is a yeast-like fungus that causes pneumonia in patients with low CD4+ T-lymphocytes as well as those on IST or prednisone doses above 20 mg/kg/day [8,109]. PCP prophylaxis with trimethropim-sulfa, dapsone, atovaquone, or pentamidine is recommended for all allogeneic HCT patients for at least 6 months, and should be continued in the setting of ongoing IST [8].

5.3. Prevention of Viral Infection

Both herpes simplex virus (HSV) and varicella zoster virus (VZV) may be reactivated in immunocompromised patients after allogeneic HCT [8]. The antiviral acyclovir and valacyclovir are both similarly effective in reducing the risk of these viruses and are acceptable options for prophylaxis [114]. Continuing these drugs until the CD4+ T-cells are above 200/mm3 and IST has been discontinued is recommended to avoid rebound [8,115].
CMV disease remains a major cause of morbidity and mortality among allogeneic HCT patients [8]. After primary infection, the virus lies dormant in the myeloid cells. CD4+ and CD8+ T cells control the infection in healthy hosts. However, after allogeneic transplant, the period after myeloid recovery and preceding T-cell recovery (days 30–100) allows a window for CMV to reactivate [8]. Prevention of CMV infection starts with allogeneic donor selection. During donor selection, CMV serology should be tested in both the donor and the recipient. CMV seropositive patients with a CMV seronegative donor are at particularly high risk for CMV infection as the donor T-cells lack a CMV memory response to suppress the virus already present in the recipient’s body. Therefore, choosing a CMV seropositive donor reduces the risk of CMV reactivation and may improve overall survival [116]. Similarly, CMV negative patients benefit from a CMV negative donor to avoid transmission of CMV virus from the donor to the immunologically naïve host [116]. After HCT, weekly monitoring of CMV viral load by PCR should be checked and pre-emptive therapy with ganciclovir or valganciclovir or foscarnet should be started in asymptomatic patients with significant viremia in order to prevent CMV disease [8]. In high risk populations, including HLA-mismatched or haplo donors, UCBT, or ex vivo TCD, initiation of CMV prophylaxis is warranted. In a phase III trial, letermovir, compared to placebo, given for CMV prophylaxis in these high-risk groups resulted in significant reductions in CMV infection (37.5% versus 60.6%, p < 0.001) and all-cause mortality (10.2% versus 15.9%, p = 0.03) at 24 weeks post-HCT [45]. A recent study of haplo donor or HLA-mismatched unrelated donor HCT with PTCy similarly showed the risk of CMV reactivation was 22% with letermovir prophylaxis versus 69% with no prophylaxis (p < 0.001). Notably, CMV vaccines are currently being studied in phase III trials [117].

5.4. Immunizations after Allogeneic HCT

Following allogeneic HCT, humoral immunity is suppressed and antibody titers to previous vaccines decline [118,119,120]. This demonstrates that re-vaccination is necessary after transplant. Response to vaccines requires reconstitution of both T-cell and B-cell immunity and, specifically, naïve T-cells that are capable of mounting a memory response after exposure to a new antigen [16,17,29]. Thus, vaccination schedules commence approximately 3–12 months after transplant [17,30]. Specific practices may vary by institution both in terms of schedule and the choice of vaccinations administered. A typical approach is to begin the vaccination schedule with pneumococcal vaccine, followed by Haemophilus influenzae, tetanus/diphtheria/pertussis (DTaP), and hepatitis B. To avoid risk of infection, live vaccines including measles/mumps/rubella (MMR) or some shingles vaccines should not be given until patients are off of immune suppression and at least 2 years have passed since HCT [8,118,119,120]. Inactivated influenza vaccine should be administered yearly [8,109,121].

5.5. Interventions to Improve Immune Reconstitution

In addition to prophylaxis and vaccinations, strategies to improve immune reconstitution can potentially reduce the risks of infection and improve the GVT effect against relapse.
Modifications to GVHD prophylactic regimens can potentially improve the balance of immune reconstitution with risks of GVHD. Historically, IST regimens were continued for at least 90 days and tapered thereafter [4,5]. With the use of PTCy, clinical trials have shown that, depending on the conditioning and donor choice, IST can be discontinued prior to day 90 or even omitted completely without increasing risks of GVHD [122,123,124]. This has the potential to boost early immune reconstitution and, additionally, creates an optimal platform for adding post-HCT maintenance strategies to prevent relapse. For TCD grafts, novel graft manipulation techniques allow for selective depletion of alpha-beta T-cells and B-cells that cause GVHD while preserving transfer of the allogeneic gamma–delta T-cells and NK cells that are required for GVT activity and infection control [125]. A recent prospective trial of myeloablative haplo HCT with alpha-beta T-cell and B-cell depletion in acute leukemia resulted in no severe GVHD and overall survival of 75%, comparable to historic controls with matched donors [126].
Immune checkpoint inhibitors are drugs that activate the immune system to attack malignant cells [127]. The mechanism of action suggests potential utility in boosting the GVT effect if given in the post-HCT setting. A phase I trial of the CTLA-4 antibody ipilimumab for post-HCT relapse led to complete responses in 9% [128]. Responses correlated with CD8+ T-cell infiltration into the tumor, supporting the immune mediated mechanism of action. GVHD occurred in only 14%. The PD-1 inhibitor nivolumab has also been studied in the post–transplant setting, though fatal immune-related toxicities have limited the potential of this strategy [129]. Notably, patients treated with checkpoint inhibitors prior to HCT may also experience post-HCT immune effects including higher rates of GVHD [130]. However, studies of haplo HCT with PTCy after prior checkpoint inhibitor suggest that GVHD rates are acceptable and relapse rates may be lower than in patients who did not receive checkpoint inhibitors [131,132]. More data is needed to confirm these findings, but this suggests that haplo HCT with PTCy and checkpoint inhibition is a potential strategy to optimize GVT.
Transfer of exogenous cells to boost immunity may help boost post-HCT immune responses. Allogeneic donor lymphocyte infusions depleted of CD8+ T-cells have been shown to successfully treat relapse and reverse immune exhaustion with low rates of GVHD [51]. In haplo HCT, a phase I study of ex vivo-expanded, donor-derived NK cells infused with HCT resulted in no dose limiting toxicities and only 1 relapse among 13 patients treated [133]. The upcoming BMT CTN 1803 NK REALM phase II trial will evaluate the effectiveness of haplo NK cell infusion in reducing the risk of relapse after haplo HCT (NCT04395092). A number of studies have also explored exogenous cell transfer to manage infectious complications of allogeneic HCT [134]. Ex vivo virus specific T-cells can be generated from healthy donors with existing immunity to a specific viral pathogen and then infused into infected HCT patients. Virus specific T-cells have mostly been studied for treatment of CMV and EBV, with complete response rates as high as 75% [135,136,137]. However, virus specific T-cells are also in development for adenovirus, human herpes virus 6, and BK virus [135,138].

6. Conclusions

Allogeneic HCT is the only curative therapy for many high-risk hematologic malignancies. Advances in GVHD prevention have broadened the donor pool to include haplo related donors and UCBT. However, severe immune deficiency and subsequent infection and relapse remain primary drivers of post-transplant morbidity and mortality. The kinetics of immune reconstitution are useful for predicting the temporality of potential complications and implementing appropriate management strategies. Novel approaches in graft manipulation and adoptive cellular therapies are being studied to accelerate post-HCT immune recovery and improve outcomes.

Author Contributions

H.E., N.B. and C.G.B. conceived the study; H.E. wrote the original manuscript draft; N.B. and C.G.B. edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Horowitz, M.M.; Gale, R.P.; Sondel, P.M.; Goldman, J.M.; Kersey, J.; Kolb, H.J.; Rimm, A.A.; Ringden, O.; Rozman, C.; Speck, B. Graft–versus–leukemia reactions after bone marrow transplantation. Blood 1990, 75, 555–562. [Google Scholar] [CrossRef] [Green Version]
  2. Przepiorka, D.; Weisdorf, D.; Martin, P.; Klingemann, H.G.; Beatty, P.; Hows, J.; Thomas, E.D. 1994 Consensus Conference on Acute GVHD Grading. Bone Marrow Transplant. 1995, 15, 825. [Google Scholar]
  3. Jagasia, M.H.; Greinix, H.T.; Arora, M.; Williams, K.M.; Wolff, D.; Cowen, E.W.; Palmer, J.; Weisdorf, D.; Treister, N.S.; Cheng, G.S.; et al. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft–versus–Host Disease: I. The 2014 Diagnosis and Staging Working Group report. BBMT 2015, 21, 389–401. [Google Scholar] [CrossRef] [Green Version]
  4. Ratanatharathorn, V.; Nash, R.A.; Przepiorka, D.; Devine, S.M.; Klein, J.L.; Weisdorf, D.; Fay, J.W.; Nademanee, A.; Antin, J.H.; Christiansen, N.P.; et al. Phase III study comparing methotrexate and tacrolimus (prograf, FK506) with methotrexate and cyclosporine for graft–versus–host disease prophylaxis after HLA–identical sibling bone marrow transplantation. Blood 1998, 92, 2303–2314. [Google Scholar]
  5. Cutler, C.; Logan, B.R.; Nakamura, R.; Johnston, L.J.; Choi, S.; Porter, D.L.; Hogan, W.J.; Pasquini, M.C.; Macmillan, M.L.; Hsu, J.W.; et al. Tacrolimus/sirolimus vs Tacrolimus/Methotrexate as GVHD Prophylaxis After Matched, Related Donor Allogeneic HCT. Blood 2014, 124, 1372–1377. [Google Scholar] [CrossRef]
  6. Hamilton, B.K. Current Approaches to Prevent and Treat GVHD after Allogeneic Stem Cell Transplant. Hematol. Am. Soc. Hematol. Educ. Program 2018, 1, 228–235. [Google Scholar] [CrossRef] [Green Version]
  7. Diaconescu, R.; Flowers, C.R.; Storer, B.; Sorror, M.L.; Maris, M.B.; Maloney, D.G.; Sandmaier, B.M.; Storb, R. Morbidity and mortality with nonmyeloablative compared with myeloablative conditioning before hematopoietic cell transplantation from HLA–matched related donors. Blood 2004, 104, 1550–1558. [Google Scholar] [CrossRef]
  8. Tomblyn, M.; Chiller, T.; Einsele, H.; Gress, R.; Sepkowitz, K.; Storek, J.; Wingard, J.R.; Young, J.H.; Boeckh, M.J. Guidelines for preventing infectious complications among hematopoietic cell transplantation recipients: A global perspective. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2009, 15, 1143–1238. [Google Scholar]
  9. Bray, R.A.; Hurley, C.K.; Kamani, N.R.; Woolfrey, A.; Müller, C.; Spellman, S.; Setterholm, M.; Confer, D.L. National Marrow Donor Program HLA Matching Guidelines for Unrelated Adult Donor Hematopoietic Cell Transplants. BBMT 2008, 14, 45–53. [Google Scholar] [CrossRef] [Green Version]
  10. Dehn, J.; Spellman, S.; Hurley, C.K.; Shaw, B.E.; Barker, J.N.; Burns, L.J.; Confer, D.L.; Eapen, M.; Fernandez-Vina, M.; Hartzman, R.; et al. Selection of unrelated donors and cord blood units for hematopoietic cell transplantation: Guidelines from the NMDP/CIBMTR. Blood 2019, 134, 924–934. [Google Scholar] [CrossRef]
  11. Gragert, L.; Eapen, M.; Williams, E.; Freeman, J.; Spellman, S.; Baitty, R.; Hartzman, R.; Rizzo, J.D.; Horowitz, M.; Confer, D.; et al. HLA Match Likelihoods for Hematopoietic Stem–Cell Grafts in the U.S. Registry. N. Engl. J. Med. 2014, 371, 339–348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Elmariah, H.; Pratz, K. Role of Alternative Donor Allogeneic Transplants in the Therapy of Acute Myeloid Leukemia. JNCCN 2017, 15, 959–966. [Google Scholar] [CrossRef] [PubMed]
  13. Brunstein, C.G.; Fuchs, E.J.; Carter, S.L.; Karanes, C.; Costa, L.J.; Wu, J.; Devine, S.M.; Wingard, J.R.; Aljitawi, O.S.; Cutler, C.S.; et al. Alternative donor transplantation after reduced intensity conditioning: Results of parallel phase 2 trials using partially HLA–mismatched related bone marrow or unrelated double umbilical cord blood grafts. Blood 2011, 118, 282–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Luznik, L.; O’Donnell, P.V.; Symons, H.J.; Chen, A.R.; Leffell, M.S.; Zahurak, M.; Gooley, T.A.; Piantadosi, S.; Kaup, M.; Ambinder, R.F.; et al. HLA–haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high–dose, posttransplantation cyclophosphamide. BBMT 2008, 14, 641–650. [Google Scholar] [CrossRef] [Green Version]
  15. Fuchs, E.J.; O’Donnell, P.V.; Eapen, M.; Logan, B.; Antin, J.H.; Dawson, P.; Devine, S.; Horowitz, M.M.; Horwitz, M.E.; Karanes, C.; et al. Double unrelated umbilical cord blood versus HLA–haploidentical bone marrow transplantation (BMT CTN 1101). Blood 2020, 137, 420–428. [Google Scholar] [CrossRef]
  16. Mackall, C.L.; Fleisher, T.A.; Brown, M.R.; Magrath, I.T.; Shad, A.T.; Horowitz, M.E.; Wexler, L.H.; Adde, M.A.; McClure, L.L.; Gress, R.E. Lymphocyte depletion during treatment with intensive chemotherapy for cancer. Blood 1994, 84, 2221–2228. [Google Scholar] [CrossRef] [Green Version]
  17. Storek, J.; Geddes, M.; Khan, F.; Huard, B.; Helg, C.; Chalandon, Y.; Passweg, J.; Roosnek, E.E. Reconstitution of the immune system after hematopoietic stem cell transplantation in humans. Semin. Immunopathol. 2008, 30, 425–437. [Google Scholar] [CrossRef]
  18. Jacobs, R.; Stoll, M.; Stratmann, G.; Leo, R.; Link, H.; Schmidt, R.E. CD16–CD56+ natural killer cells after bone marrow transplantation. Blood 1992, 79, 3239–3244. [Google Scholar]
  19. Vitale, C.; Pitto, A.; Benvenuto, F.; Ponte, M.; Bellomo, R.; Frassoni, F.; Mingari, M.C.; Bacigalupo, A.; Moretta, L. Phenotypic and functional analysis of the HLA–class I–specific inhibitory receptors of natural killer cells isolated from peripheral blood of patients undergoing bone marrow transplantation from matched unrelated donors. Hematol. J. 2000, 1, 136–144. [Google Scholar] [CrossRef]
  20. Uharek, L.; Glass, B.; Gaska, T.; Zeiss, M.; Gassmann, W.; Loeffler, H.; Mueller-Ruchholtz, W. Natural killer cells as effector cells of graft–versus–leukemia activity in a murine transplantation model. Bone Marrow Transplant. 1993, 12 (Suppl. 3), S57–S60. [Google Scholar]
  21. Jiang, Y.Z.; Barrett, A.J.; Goldman, J.M.; Mavroudis, D.A. Association of natural killer cell immune recovery with a graft–versus–leukemia effect independent of graft–versus–host disease following allogeneic bone marrow transplantation. Ann. Hematol. 1997, 74, 1–6. [Google Scholar] [CrossRef] [PubMed]
  22. Farag, S.S.; Fehniger, T.A.; Ruggeri, L.; Velardi, A.; Caligiuri, M.A. Natural killer cell receptors: New biology and insights into the graft–versus–leukemia effect. Blood 2002, 100, 1935–1947. [Google Scholar] [CrossRef] [PubMed]
  23. Cruz, C.R.; Bollard, C.M. T–cell and natural killer cell therapies for hematologic malignancies after hematopoietic stem cell transplantation: Enhancing the graft–versus–leukemia effect. Haematologica 2015, 100, 709–719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zimmerli, W.; Zarth, A.; Gratwohl, A.; Speck, B. Neutrophil function and pyogenic infections in bone marrow transplant recipients. Blood 1991, 77, 393–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Cayeux, S.; Meuer, S.; Pezzutto, A.; Korbling, M.; Haas, R.; Schulz, R.; Dorken, B. Allogeneic mixed lymphocyte reactions during a second round of ontogeny: Normal accessory cells did not restore defective interleukin–2 (IL–2) synthesis in T cells but induced responsiveness to exogeneous IL–2. Blood 1989, 74, 2278–2284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Talmadge, J.E. Lymphocyte subset recovery following allogeneic bone marrow transplantation: CD4(+)–cell count and transplant–related mortality. Bone Marrow Transplant. 2008, 41, 19–21. [Google Scholar] [CrossRef]
  27. Thoma, M.D.; Huneke, T.J.; DeCook, L.J.; Johnson, N.D.; Wiegand, R.A.; Litzow, M.R.; Hogan, W.J.; Porrata, L.F.; Holtan, S.G. Peripheral blood lymphocyte and monocyte recovery and survival in acute leukemia postmyeloablative allogeneic hematopoietic stem cell transplant. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2012, 18, 600–607. [Google Scholar] [CrossRef] [Green Version]
  28. Yamamoto, W.; Ogusa, E.; Matsumoto, K.; Maruta, A.; Ishigatsubo, Y.; Kanamori, H. Lymphocyte recovery on day 100 after allogeneic hematopoietic stem cell transplant predicts non–relapse mortality in patients with acute leukemia or myelodysplastic syndrome. Leuk. Lymphoma 2014, 55, 1113–1118. [Google Scholar] [CrossRef]
  29. Mackall, C.L.; Fleisher, T.A.; Brown, M.R.; Andrich, M.P.; Chen, C.C.; Feuerstein, I.M.; Horowitz, M.E.; Magrath, I.T.; Shad, A.T.; Steinberg, S.M.; et al. Age, thymopoiesis, and CD4+ T–lymphocyte regeneration after intensive chemotherapy. N. Engl. J. Med. 1995, 332, 143–149. [Google Scholar] [CrossRef]
  30. Storek, J. B–cell immunity after allogeneic hematopoietic cell transplantation. Cytotherapy 2002, 4, 423–424. [Google Scholar] [CrossRef]
  31. Harrison, D.E.; Astle, C.M. Loss of stem cell repopulating ability upon transplantation. Effects of donor age, cell number, and transplantation procedure. J. Exp. Med. 1982, 156, 1767–1779. [Google Scholar] [CrossRef] [PubMed]
  32. Jacobsen, N.; Badsberg, J.H.; Lönnqvist, B.; Ringden, O.; Volin, L.; Rajantie, J.; Nikoskelainen, J.; Keiding, N. Graft–versus–leukaemia activity associated with CMV–seropositive donor, post–transplant CMV infection, young donor age and chronic graft–versus–host disease in bone marrow allograft recipients. The Nordic Bone Marrow Transplantation Group. Bone Marrow Transplant. 1990, 5, 413–418. [Google Scholar] [PubMed]
  33. Seggewiss, R.; Einsele, H. Immune reconstitution after allogeneic transplantation and expanding options for immunomodulation: An update. Blood 2010, 115, 3861–3868. [Google Scholar] [CrossRef] [PubMed]
  34. Anasetti, C.; Amos, D.; Beatty, P.G.; Appelbaum, F.R.; Bensinger, W.; Buckner, C.D.; Clift, R.; Doney, K.; Martin, P.J.; Mickelson, E.; et al. Effect of HLA compatibility on engraftment of bone marrow transplants in patients with leukemia or lymphoma. N. Engl. J. Med. 1987, 320, 197–204. [Google Scholar] [CrossRef]
  35. Soderling, C.C.; Song, C.W.; Blazar, B.R.; Vallera, D.A. A correlation between conditioning and engraftment in recipients of MHC–mismatched T cell–depleted murine bone marrow transplants. J. Immunol. 1985, 135, 941–946. [Google Scholar]
  36. Anasetti, C.; Logan, B.R.; Lee, S.J.; Waller, E.K.; Weisdorf, D.J.; Wingard, J.R.; Cutler, C.S.; Westervelt, P.; Woolfrey, A.; Couban, S.; et al. Peripheral–Blood Stem Cells versus Bone Marrow from Unrelated Donors. N. Engl. J. Med. 2012, 367, 1487–1496. [Google Scholar] [CrossRef] [Green Version]
  37. Siena, S.; Schiavo, R.; Pedrazzoli, P.; Carlo–Stella, C. Therapeutic relevance of CD34 cell dose in blood cell transplantation for cancer therapy. J. Clin. Oncol. 2000, 18, 1360–1377. [Google Scholar] [CrossRef]
  38. Nakamura, R.; Auayporn, N.; Smith, D.D.; Palmer, J.; Sun, J.Y.; Schriber, J.; Pullarkat, V.; Parker, P.; Rodriguez, R.; Stein, A.; et al. Impact of graft cell dose on transplant outcomes following unrelated donor allogeneic peripheral blood stem cell transplantation: Higher CD34+ cell doses are associated with decreased relapse rates. BBMT 2008, 14, 449–457. [Google Scholar] [CrossRef] [Green Version]
  39. Remberger, M.; Törlén, J.; Ringdén, O.; Engström, M.; Watz, E.; Uhlin, M.; Mattsson, J. Effect of Total Nucleated and CD34(+) Cell Dose on Outcome after Allogeneic Hematopoietic Stem Cell Transplantation. BBMT 2015, 21, 889–893. [Google Scholar] [CrossRef] [Green Version]
  40. Tokuda, N.; Mayumi, H.; Sakumoto, M.; Himeno, K.; Tomita, Y.; Nomoto, K. The effect of T cell depletion from spleen cell allografts on graft–versus–host disease and long–term immune reconstitution in H–2 haplotype–identical murine combinations. Immunobiology 1989, 179, 328–341. [Google Scholar] [CrossRef]
  41. Roex, M.C.; Wijnands, C.; Veld, S.A.; Van Egmond, E.; Bogers, L.; Zwaginga, J.J.; Netelenbos, T.; Borne, P.A.V.D.; Veelken, H.; Halkes, C.J.; et al. Effect of alemtuzumab–based T–cell depletion on graft compositional change in vitro and immune reconstitution early after allogeneic stem cell transplantation. Cytotherapy 2020, 23, 46–56. [Google Scholar] [CrossRef] [PubMed]
  42. Cutler, C.; Pavletic, S.Z. NCCN Guidelines: Pretransplant Recipient Evaluation and Management of Graft–Versus–Host Disease. J. Natl. Compr. Cancer Netw. 2020, 18, 645–647. [Google Scholar] [CrossRef] [PubMed]
  43. Heidt, P.J. Management of bacterial and fungal infections in bone marrow transplant recipients and other granulocytopenic patients. Cancer Detect Prev. 1988, 12, 609–619. [Google Scholar] [PubMed]
  44. Lalitha, M.K.; Pai, R.; Manoharan, A.; Jesudason, M.V.; Brahmadathan, K.N.; Srivastava, A.; Chandy, M. Systemic bacterial infections in bone marrow transplant patients. Indian J. Cancer 2000, 37, 10–14. [Google Scholar]
  45. 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]
  46. Crawford, D.H.; Mulholland, N.; Iliescu, V.; Hawkins, R.; Powles, R. Epstein–Barr virus infection and immunity in bone marrow transplant recipients. Transplantation 1986, 42, 50–54. [Google Scholar] [CrossRef]
  47. Bolaños-Meade, J.; Reshef, R.; Fraser, R.; Fei, M.; Abhyankar, S.; Al-Kadhimi, Z.; Alousi, A.M.; Antin, J.H.; Arai, S.; Bickett, K.; et al. Three prophylaxis regimens (tacrolimus, mycophenolate mofetil, and cyclophosphamide; tacrolimus, methotrexate, and bortezomib; or tacrolimus, methotrexate, and maraviroc) versus tacrolimus and methotrexate for prevention of graft–versus–host disease with haemopoietic cell transplantation with reduced–intensity conditioning: A randomised phase 2 trial with a non–randomised contemporaneous control group (BMT CTN 1203). Lancet Haematol. 2019, 6, 132–143. [Google Scholar]
  48. Kanarky, C.J.; Fuchs, E.J.; Luznik, L. Modern Approaches to HLA-Haploidentical Blood or Marrow Transplantation. Nat. Rev. Clin. Oncol. 2016, 13, 10–24. [Google Scholar]
  49. Storek, J.; Wells, D.; Dawson, M.A.; Storer, B.; Maloney, D.G. Factors influencing B lymphopoiesis after allogeneic hematopoietic cell transplantation. Blood 2001, 98, 489–491. [Google Scholar] [CrossRef] [Green Version]
  50. Barrett, A.J. Mechanisms of the graft–versus–leukemia reaction. Stem Cells 1997, 15, 248–258. [Google Scholar] [CrossRef]
  51. Bachireddy, P.; Hainz, U.; Rooney, M.; Pozdnyakova, O.; Aldridge, J.; Zhang, W.; Liao, X.; Hodi, F.S.; O’Connell, K.; Haining, W.N.; et al. Reversal of in Situ T–cell Exhaustion During Effective Human Antileukemia Responses to Donor Lymphocyte Infusion. Blood 2014, 123, 1412–1421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Ofran, Y.; Ritz, J. Targets of Tumor Immunity after Allogeneic Hematopoietic Stem Cell Transplantation. Clin. Cancer Res. 2008, 14, 4997–4999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Shaffer, B.C.; Hsu, K.C. Selection of allogeneic hematopoietic cell transplant donors to optimize natural killer cell alloreactivity. Semin. Hematol. 2020, 57, 167–174. [Google Scholar] [CrossRef] [PubMed]
  54. Yu, J.; Venstrom, J.M.; Liu, X.-R.; Pring, J.; Hasan, R.S.; O’Reilly, R.J.; Hsu, K.C. Breaking tolerance to self, circulating natural killer cells expressing inhibitory KIR for non–self HLA exhibit effector function after T cell–depleted allogeneic hematopoietic cell transplantation. Blood 2009, 113, 3875–3884. [Google Scholar] [CrossRef] [PubMed]
  55. Brodin, P.; Karre, K.; Hoglund, P. NK cell education: Not an on–off switch but a tunable rheostat. Trends Immunol. 2009, 30, 143–149. [Google Scholar] [CrossRef]
  56. Brodin, P.; Lakshmikanth, T.; Johansson, S.; Karre, K.; Hoglund, P. The strength of inhibitory input during education quantitatively tunes the functional responsiveness of individual natural killer cells. Blood 2009, 113, 2434–2441. [Google Scholar] [CrossRef] [Green Version]
  57. Hsu, K.C.; Chida, S.; Geraghty, D.E.; Dupont, B. The killer cell immunoglobulin–like receptor (KIR) genomic region: Gene–order, haplotypes and allelic polymorphism. Immunol. Rev. 2002, 190, 40–52. [Google Scholar] [CrossRef]
  58. Hsu, K.C.; Liu, X.R.; Selvakumar, A.; Mickelson, E.; O’Reilly, R.J.; Dupont, B. Killer Ig–like receptor haplotype analysis by gene content: Evidence for genomic diversity with a minimum of six basic framework haplotypes, each with multiple subsets. J Immunol. 2002, 169, 5118–5129. [Google Scholar] [CrossRef]
  59. Ruggeri, L.; Mancusi, A.; Perruccio, K.; Burchielli, E.; Martelli, M.F.; Velardi, A. Natural killer cell alloreactivity for leukemia therapy. J. Immunother. 2005, 28, 175–182. [Google Scholar] [CrossRef]
  60. Ruggeri, L.; Aversa, F.; Martelli, M.F.; Velardi, A. Allogeneic hematopoietic transplantation and natural killer cell recognition of missing self. Immunol. Rev. 2006, 214, 202–218. [Google Scholar] [CrossRef]
  61. Ruggeri, L.; Mancusi, A.; Burchielli, E.; Perruccio, K.; Aversa, F.; Martelli, M.F.; Velardi, A. Natural killer cell recognition of missing self and haploidentical hematopoietic transplantation. Semin. Cancer Biol. 2006, 16, 404–411. [Google Scholar] [CrossRef]
  62. Velardi, A.; Ruggeri, L.; Mancusi, A. Killer–cell immunoglobulin–like receptors reactivity and outcome of stem cell transplant. Curr. Opin. Hematol. 2012, 19, 319–323. [Google Scholar] [CrossRef] [PubMed]
  63. Shaffer, B.C.; Hsu, K.C. How important is NK alloreactivity and KIR in allogeneic transplantation? Best Pract. Res. Clin. Haematol. 2016, 29, 351–358. [Google Scholar] [CrossRef] [PubMed]
  64. Boudreau, J.E.; Giglio, F.; Gooley, T.A.; Stevenson, P.A.; Le Luduec, J.-B.; Shaffer, B.C.; Rajalingam, R.; Hou, L.; Hurley, C.K.; Noreen, H.; et al. KIR3DL1/HLA–B Subtypes Govern Acute Myelogenous Leukemia Relapse After Hematopoietic Cell Transplantation. J. Clin. Oncol. 2017, 35, 2268–2278. [Google Scholar] [CrossRef] [PubMed]
  65. Gao, F.; Ye, Y.; Gao, Y.; Huang, H.; Zhao, Y. Influence of KIR and NK Cell Reconstitution in the Outcomes of Hematopoietic Stem Cell Transplantation. Front. Immunol. 2020, 11, 2022. [Google Scholar] [CrossRef] [PubMed]
  66. Verheyden, S.; Demanet, C. NK cell receptors and their ligands in leukemia. Leukemia 2008, 22, 249–257. [Google Scholar] [CrossRef] [Green Version]
  67. Verheyden, S.; Ferrone, S.; Mulder, A.; Claas, F.H.; Schots, R.; De Moerloose, B.; Benoit, Y.; Demanet, C. Role of the inhibitory KIR ligand HLA–Bw4 and HLA–C expression levels in the recognition of leukemic cells by Natural Killer cells. Cancer Immunol. Immunother. 2009, 58, 855–865. [Google Scholar] [CrossRef] [Green Version]
  68. Bolaños-Meade, J.; Fuchs, E.J.; Luznik, L.; Lanzkron, S.M.; Gamper, C.J.; Jones, R.J.; Brodsky, R.A. HLA–haploidentical bone marrow transplantation with posttransplant cyclophosphamide expands the donor pool for patients with sickle cell disease. Blood 2012, 120, 4285–4291. [Google Scholar] [CrossRef] [Green Version]
  69. Aversa, F.; Tabilio, A.; Velardi, A.; Cunningham, I.; Terenzi, A.; Falzetti, F.; Ruggeri, L.; Barbabietola, G.; Aristei, C.; Latini, P.; et al. Treatment of high–risk acute leukemia with T–cell–depleted stem cells from related donors with one fully mismatched HLA haplotype. N. Engl. J. Med. 1998, 339, 1186–1193. [Google Scholar] [CrossRef]
  70. Aversa, F.; Terenzi, A.; Tabilio, A.; Falzetti, F.; Carotti, A.; Ballanti, S.; Felicini, R.; Falcinelli, F.; Velardi, A.; Ruggeri, L.; et al. Full haplotype–mismatched hematopoietic stem–cell transplantation: A phase II study in patients with acute leukemia at high risk of relapse. J. Clin. Oncol. 2005, 23, 3447–3454. [Google Scholar] [CrossRef]
  71. Huang, X.-J.; Liu, D.-H.; Liu, K.-Y.; Xu, L.-P.; Chen, H.; Han, W.; Chen, Y.-H.; Wang, J.-Z.; Gao, Z.-Y.; Zhang, Y.; et al. Haploidentical hematopoietic stem cell transplantation without in vitro T–cell depletion for the treatment of hematological malignancies. Bone Marrow Transplant. 2006, 38, 291–297. [Google Scholar] [CrossRef] [Green Version]
  72. Raiola, A.M.; Dominietto, A.; Di Grazia, C.; Lamparelli, T.; Gualandi, F.; Ibatici, A.; Bregante, S.; Van Lint, M.T.; Varaldo, R.; Ghiso, A.; et al. Unmanipulated haploidentical transplants compared with other alternative donors and matched sibling grafts. BBMT 2014, 20, 1573–1579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ranspach, P.; Zhou, J.M.; Pidala, J.A.; Nishihori, T.; Nieder, M.L.; Elmariah, H.; Faramand, R.; Lazaryan, A.; Baluch, A.; Mishra, A.; et al. Delayed CD4+ T–Cell but Faster B–Cell Immune Reconstitution after Ptcy–Based Compared to Conventional Gvhd Prophylaxis after Allogeneic Transplantation. BBMT 2020, 26, S308–S309. [Google Scholar]
  74. Goldsmith, S.R.; Fuchs, E.J.; Bashey, A.; Ciurea, S.O.; Singh, A.K.; Ganguly, S.; Taplitz, R.; Mulroney, C.; Maziarz, R.T.; Kim, S.; et al. Incidence and Impact of Cytomegalovirus Infection in Haploidentical and Matched–Related Donors Receiving Post–Transplant Cyclophosphamide (PTCy): A CIBMTR Analysis. BBMT 2020, 26, S69–S70. [Google Scholar] [CrossRef]
  75. 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] [PubMed]
  76. Di Ianni, M.; Falzetti, F.; Carotti, A.; Terenzi, A.; Castellino, F.; Bonifacio, E.; Del Papa, B.; Zei, T.; Ostini, R.I.; Cecchini, D.; et al. Tregs prevent GVHD and promote immune reconstitution in HLA–haploidentical transplantation. Blood 2011, 117, 3921–3928. [Google Scholar] [CrossRef]
  77. Chang, Y.-J.; Zhaoab, X.; Huo, M.-R.; Xu, L.-P.; Liu, D.-H.; Liu, K.; Huang, X.-J. Immune reconstitution following unmanipulated HLA–mismatched/haploidentical transplantation compared with HLA–identical sibling transplantation. J. Clin. Immunol. 2012, 32, 268–280. [Google Scholar] [CrossRef]
  78. McCurdy, S.R.; Kasamon, Y.L.; Kanakry, C.G.; Bolaños-Meade, J.; Tsai, H.-L.; Showel, M.M.; Kanakry, J.A.; Symons, H.J.; Gojo, I.; Smith, B.D.; et al. Comparable composite endpoints after HLA–matched and HLA–haploidentical transplantation with post–transplantation cyclophosphamide. Haematologica 2017, 102, 391–400. [Google Scholar] [CrossRef] [Green Version]
  79. Lorentino, F.; Labopin, M.; Fleischhauer, K.; Ciceri, F.; Mueller, C.R.; Ruggeri, A.; Shimoni, A.; Bornhäuser, M.; Bacigalupo, A.; Gülbas, Z.; et al. The impact of HLA matching on outcomes of unmanipulated haploidentical HSCT is modulated by GVHD prophylaxis. Blood Adv. 2017, 1, 669–680. [Google Scholar] [CrossRef] [Green Version]
  80. Ciurea, S.O.; Zhang, M.J.; Bacigalupo, A.A.; Bashey, A.; Appelbaum, F.R.; Aljitawi, O.S.; Armand, P.; Antin, J.H.; Chen, J.; Devine, S.M.; et al. Haploidentical transplant with post–transplant cyclophosphamide versus matched unrelated donor transplant for acute myeloid leukemia. Blood 2015, 126, 1033–1040. [Google Scholar]
  81. Bashey, A.; Zhang, M.-J.; McCurdy, S.R.; Martin, A.S.; Argall, T.; Anasetti, C.; Ciurea, S.O.; Fasan, O.; Gaballa, S.; Hamadani, M.; et al. Mobilized Peripheral Blood Stem Cells Versus Unstimulated Bone Marrow As a Graft Source for T–Cell–Replete Haploidentical Donor Transplantation Using Post–Transplant Cyclophosphamide. JCO 2017, 35, 3002–3009. [Google Scholar] [CrossRef] [PubMed]
  82. Imus, P.H.; Blackford, A.L.; Bettinotti, M.; Iglehart, B.; Dietrich, A.; Tucker, N.; Symons, H.; Cooke, K.R.; Luznik, L.; Fuchs, E.J.; et al. Major Histocompatibility Mismatch and Donor Choice for Second Allogeneic Bone Marrow Transplantation. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2017, 23, 1887–1894. [Google Scholar] [CrossRef] [PubMed]
  83. Vago, L.; Perna, S.K.; Zanussi, M.; Mazzi, B.; Barlassina, C.; Stanghellini, M.T.L.; Perrelli, N.F.; Cosentino, C.; Torri, F.; Angius, A.; et al. Loss of mismatched HLA in leukemia after stem–cell transplantation. N. Engl. J. Med. 2009, 361, 478–488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Villalobos, I.B.; Takahashi, Y.; Akatsuka, Y.; Muramatsu, H.; Nishio, N.; Hama, A.; Yagasaki, H.; Saji, H.; Kato, M.; Ogawa, S.; et al. Relapse of leukemia with loss of mismatched HLA resulting from uniparental disomy after haploidentical hematopoietic stem cell transplantation. Blood 2010, 115, 3158–3161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Stölzel, F.; Hackmann, K.; Kuithan, F.; Mohr, B.; Füssel, M.; Oelschlägel, U.; Thiede, C.; Röllig, C.; Platzbecker, U.; Schetelig, J.; et al. Clonal evolution including partial loss of human leukocyte antigen genes favoring extramedullary acute myeloid leukemia relapse after matched related allogeneic hematopoietic stem cell transplantation. Transplantation 2012, 93, 744–749. [Google Scholar] [CrossRef]
  86. Hamdi, A.; Cao, K.; Poon, L.M.; Aung, F.; Kornblau, S.M.; Vina, M.A.F.; Champlin, R.E.; O Ciurea, S. Are changes in HLA Ags responsible for leukemia relapse after HLA–matched allogeneic hematopoietic SCT? Bone Marrow Transplant. 2015, 50, 411–413. [Google Scholar] [CrossRef] [Green Version]
  87. Brunstein, C.G.; Gutman, J.A.; Weisdorf, D.J.; Woolfrey, A.E.; DeFor, T.E.; Gooley, T.A.; Verneris, M.R.; Appelbaum, F.R.; Wagner, J.E.; Delaney, C. Allogeneic hematopoietic cell transplantation for hematologic malignancy: Relative risks and benefits of double umbilical cord blood. Blood 2010, 116, 4693–4699. [Google Scholar] [CrossRef] [Green Version]
  88. Brunstein, C.G.; Eapen, M.; Ahn, K.W.; Appelbaum, F.R.; Ballen, K.K.; Champlin, R.E.; Cutler, C.; Kan, F.; Laughlin, M.J.; Soiffer, R.J.; et al. Reduced–intensity conditioning transplantation in acute leukemia: The effect of source of unrelated donor stem cells on outcomes. Blood 2012, 119, 5591–5598. [Google Scholar]
  89. Laughlin, M.J.; Barker, J.; Ambach, B.A.B.; Oc, O.M.N.K.; Izzieri, D.A.A.R.; Agner, J.O.E.W.; Erson, S.T.L.G.; Lazarus, H.M.; Airo, M.I.C.; Tevens, C.L.E.S.; et al. Hematopoietic Engraftment and Survival in Adult Recipients of Umbilical–Cord Blood from Unrelated Donors. N. Engl. J. Med. 2001, 344, 1815–1822. [Google Scholar] [CrossRef]
  90. Bejanyan, N.; Haddad, H.; Brunstein, C. Alternative Donor Transplantation for Acute Myeloid Leukemia. J. Clin. Med. 2015, 4, 1240–1268. [Google Scholar] [CrossRef] [Green Version]
  91. Komanduri, K.V.; John, L.S.S.; De Lima, M.; McMannis, J.; Rosinski, S.; Mcniece, I.; Bryan, S.G.; Kaur, I.; Martin, S.; Wieder, E.D.; et al. Delayed immune reconstitution after cord blood transplantation is characterized by impaired thymopoiesis and late memory T–cell skewing. Blood 2007, 110, 4543–4551. [Google Scholar] [CrossRef] [PubMed]
  92. Bejanyan, N.; Brunstein, C.G.; Cao, Q.; Lazaryan, A.; Luo, X.; Curtsinger, J.; Mehta, R.S.; Warlick, E.; Cooley, S.A.; Blazar, B.R.; et al. Delayed immune reconstitution after allogeneic transplantation increases the risks of mortality and chronic GVHD. Blood Adv. 2018, 2, 909–922. [Google Scholar] [CrossRef] [PubMed]
  93. Sauter, C.; Abboud, M.; Jia, X.; Heller, G.; Gonzales, A.-M.; Lubin, M.; Hawke, R.; Perales, M.-A.; Brink, M.R.V.D.; Giralt, S.; et al. Serious infection risk and immune recovery after double–unit cord blood transplantation without antithymocyte globulin. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2011, 17, 1460–1471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Saliba, R.M.; Rezvani, K.; Leen, A.; Jorgensen, J.; Shah, N.; Hosing, C.; Parmar, S.; Oran, B.; Olson, A.; Rondon, G.; et al. General and Virus–Specific Immune Cell Reconstitution after Double Cord Blood Transplantation. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2015, 21, 1284–1290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Jacobson, C.A.; Turki, A.T.; McDonough, S.M.; Stevenson, K.E.; Kim, H.T.; Kao, G.; Herrera, M.I.; Reynolds, C.G.; Alyea, E.P.; Ho, V.T.; et al. Immune reconstitution after double umbilical cord blood stem cell transplantation: Comparison with unrelated peripheral blood stem cell transplantation. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2012, 18, 565–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Barker, J.; Krepski, T.; DeFor, T.; Davies, S.; Wagner, J.; Weisdorf, D. Searching for unrelated donor hematopoietic stem cells: Availability and speed of umbilical cord blood versus bone marrow. BBMT 2002, 8, 257. [Google Scholar] [CrossRef] [Green Version]
  97. Barker, J.N.; Weisdorf, D.J.; DeFor, T.E.; Blazar, B.R.; McGlave, P.B.; Miller, J.S.; Verfaillie, C.M.; Wagner, J.E. Transplantation of 2 partially HLA–matched umbilical cord blood units to enhance engraftment in adults with hematologic malignancy. Blood 2005, 105, 1343–1347. [Google Scholar] [CrossRef] [Green Version]
  98. Bejanyan, N.; Rogosheske, J.; DeFor, T.E.; Lazaryan, A.; Arora, M.; Holtan, S.G.; Jacobson, P.A.; Macmillan, M.L.; Verneris, M.R.; Blazar, B.R.; et al. Sirolimus and Mycophenolate Mofetil as Calcineurin Inhibitor–Free Graft–versus–Host Disease Prophylaxis for Reduced–Intensity Conditioning Umbilical Cord Blood Transplantation. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2016, 22, 2025–2030. [Google Scholar] [CrossRef] [Green Version]
  99. Politikos, I.; Lavery, J.A.; Hilden, P.; Cho, C.; Borrill, T.; Maloy, M.A.; Giralt, S.A.; van den Brink, M.R.; Perales, M.A.; Barker, J.N. Robust CD4+ T–cell recovery in adults transplanted with cord blood and no antithymocyte globulin. Blood Adv. 2020, 4, 191–202. [Google Scholar] [CrossRef]
  100. Bejanyan, N.; Vlasova-St, L.I.; Mohei, H.; Cao, Q.; El Jurdi, N.; Wagner, J.E.; Miller, J.S.; Brunstein, C.G. CMV–Specific Immunity Recovers Slowly after Cord Blood Compared with Matched Sibling Donor Allogeneic Transplantation. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2020, (in press). [Google Scholar]
  101. El Jurdi, N.; Rogosheske, J.; DeFor, T.; Bejanyan, N.; Arora, M.; Bachanova, V.; Betts, B.; He, F.; Holtan, S.; Janakiram, M.; et al. Prophylactic Foscarnet for Human Herpesvirus 6: Effect on Hematopoietic Engraftment after Reduced–Intensity Conditioning Umbilical Cord Blood Transplantation. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2020, 27, 84e1–84e5. [Google Scholar] [CrossRef] [PubMed]
  102. Rolston, K.V. Challenges in the treatment of infections caused by gram–positive and gram–negative bacteria in patients with cancer and neutropenia. Clin. Infect. Dis. 2005, 40 (Suppl. 4), S246–S252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Bucaneve, G.; Micozzi, A.; Menichetti, F.; Martino, P.; Dionisi, M.S.; Martinelli, G.; Allione, B.; D’Antonio, D.; Buelli, M.; Nosari, A.M.; et al. Levofloxacin to prevent bacterial infection in patients with cancer and neutropenia. N. Engl. J. Med. 2005, 353, 977–987. [Google Scholar] [CrossRef] [Green Version]
  104. Gafter-Gvili, A.; Fraser, A.; Paul, M.; Vidal, L.; Lawrie, T.A.; van de Wetering, M.D.; Kremer, L.C.; Leibovici, L. Antibiotic prophylaxis for bacterial infections in afebrile neutropenic patients following chemotherapy. Cochrane Database Syst. Rev. 2012, 1, CD004386. [Google Scholar] [CrossRef] [PubMed]
  105. Doan, V.P.; Yeh, J.C.; Gulbis, A.M.; Aitken, S.L.; Ariza–Heredia, E.; Ahmed, S. Levofloxacin versus Cefpodoxime for Antibacterial Prophylaxis in Allogeneic Stem Cell Transplantation. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2019, 25, 1637–1641. [Google Scholar] [CrossRef]
  106. Bow, E.J. Fluoroquinolones, antimicrobial resistance and neutropenic cancer patients. Curr. Opin. Infect. Dis. 2011, 24, 545–553. [Google Scholar] [CrossRef]
  107. Dellinger, R.P.; Levy, M.M.; Carlet, J.M.; Bion, J.; Parker, M.M.; Jaeschke, R.; Reinhart, K.; Angus, D.C.; Brun-Buisson, C.; Beale, R.; et al. Surviving Sepsis Campaign: International guidelines for management of severe sepsis and septic shock: 2008. Crit. Care Med. 2008, 36, 296–327. [Google Scholar] [CrossRef] [Green Version]
  108. Carpenter, P.A.; Kitko, C.L.; Elad, S.; Flowers, M.E.; Gea-Banacloche, J.C.; Halter, J.P.; Hoodin, F.; Johnston, L.; Lawitschka, A.; McDonald, G.B.; et al. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft–versus–Host Disease: V. The 2014 Ancillary Therapy and Supportive Care Working Group Report. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2015, 21, 1167–1187. [Google Scholar] [CrossRef] [Green Version]
  109. Taplitz, R.A.; Kennedy, E.B.; Bow, E.J.; Crews, J.; Gleason, C.; Hawley, D.K.; Langston, A.A.; Nastoupil, L.J.; Rajotte, M.; Rolston, K.V.; et al. Antimicrobial Prophylaxis for Adult Patients With Cancer–Related Immunosuppression: ASCO and IDSA Clinical Practice Guideline Update. J. Clin. Oncol. 2018, 36, 3043–3054. [Google Scholar] [CrossRef]
  110. Robenshtok, E.; Gafter-Gvili, A.; Goldberg, E.; Weinberger, M.; Yeshurun, M.; Leibovici, L.; Paul, M. Antifungal prophylaxis in cancer patients after chemotherapy or hematopoietic stem–cell transplantation: Systematic review and meta–analysis. J. Clin. Oncol. 2007, 25, 5471–5489. [Google Scholar] [CrossRef]
  111. Wingard, J.R.; Carter, S.L.; Walsh, T.J.; Kurtzberg, J.; Small, T.N.; Baden, L.R.; Gersten, I.D.; Mendizabal, A.M.; Leather, H.L.; Confer, D.L.; et al. Randomized, double–blind trial of fluconazole versus voriconazole for prevention of invasive fungal infection after allogeneic hematopoietic cell transplantation. Blood 2010, 116, 5111–5118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Gotzsche, P.C.; Johansen, H.K. Routine versus selective antifungal administration for control of fungal infections in patients with cancer. Cochrane Database Syst. Rev. 2014, CD000026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Ullmann, A.J.; Lipton, J.H.; Vesole, D.H.; Chandrasekar, P.; Langston, A.; Tarantolo, S.R.; Greinix, H.; De Azevedo, W.M.; Reddy, V.; Boparai, N.; et al. Posaconazole or fluconazole for prophylaxis in severe graft–versus–host disease. N. Engl. J. Med. 2007, 356, 335–347. [Google Scholar] [CrossRef] [PubMed]
  114. Eisen, D.; Essell, J.; Broun, E.R.; Sigmund, D.; DeVoe, M. Clinical utility of oral valacyclovir compared with oral acyclovir for the prevention of herpes simplex virus mucositis following autologous bone marrow transplantation or stem cell rescue therapy. Bone Marrow Transplant. 2003, 31, 51–55. [Google Scholar] [CrossRef] [Green Version]
  115. Erard, V.; Guthrie, K.A.; Varley, C.; Heugel, J.; Wald, A.; Flowers, M.E.D.; Corey, L.; Boeckh, M.; Arulogun, S.O.; Prince, H.M.; et al. One–year acyclovir prophylaxis for preventing varicella–zoster virus disease after hematopoietic cell transplantation: No evidence of rebound varicella–zoster virus disease after drug discontinuation. Blood 2007, 110, 3071–3077. [Google Scholar] [CrossRef]
  116. Shaw, B.E.; Logan, B.R.; Spellman, S.R.; Marsh, S.G.; Robinson, J.; Pidala, J.; Hurley, C.; Barker, J.; Maiers, M.; Dehn, J.; et al. Development of an Unrelated Donor Selection Score Predictive of Survival after HCT: Donor Age Matters Most. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2018, 24, 1049–1056. [Google Scholar] [CrossRef] [Green Version]
  117. Lin, A.; Flynn, J.; DeRespiris, L.; Figgins, B.; Griffin, M.; Lau, C.; Proli, A.; Devlin, S.M.; Cho, C.; Tamari, R.; et al. Letermovir for Prevention of Cytomegalovirus Reactivation in Haploidentical and Mismatched Adult Donor Allogeneic Hematopoietic Cell Transplantation with Post–Transplantation Cyclophosphamide for Graft–versus–Host Disease Prophylaxis. Biol. Blood Marrow Transplant. 2012, 12, 290–299. [Google Scholar] [CrossRef]
  118. Kharfan-Dabaja, M.A.; Boeckh, M.; Wilck, M.B.; A Langston, A.; Chu, A.H.; Wloch, M.K.; Guterwill, D.F.; Smith, L.; Rolland, A.P.; Kenney, R.T. A novel therapeutic cytomegalovirus DNA vaccine in allogeneic haemopoietic stem–cell transplantation: A randomised, double–blind, placebo–controlled, phase 2 trial. Lancet Infect Dis. 2012, 12, 290–299. [Google Scholar] [CrossRef] [Green Version]
  119. Ljungman, P.; Fridell, E.; Lönqvist, B.; Bolme, P.; Böttiger, M.; Gahrton, G.; Linde, A.; Ringden, O.; Wahren, B. Efficacy and safety of vaccination of marrow transplant recipients with a live attenuated measles, mumps, and rubella vaccine. J. Infect. Dis. 1989, 159, 610–615. [Google Scholar]
  120. Ljungman, P.; Wiklund-Hammarsten, M.; Duraj, V.; Hammarström, L.; Lönnqvist, B.; Paulin, T.; Ringdén, O.; Sullivan Pepe, M.; Gahrton, G. Response to tetanus toxoid immunization after allogeneic bone marrow transplantation. J. Infect. Dis. 1990, 162, 496–500. [Google Scholar] [CrossRef]
  121. Halasa, N.B.; Savani, B.N.; Asokan, I.; Kassim, A.; Simons, R.; Summers, C.; Bourgeois, J.; Clifton, C.; Vaughan, L.A.; Lucid, C.; et al. Randomized Double–Blind Study of the Safety and Immunogenicity of Standard–Dose Trivalent Inactivated Influenza Vaccine versus High–Dose Trivalent Inactivated Influenza Vaccine in Adult Hematopoietic Stem Cell Transplantation Patients. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2016, 22, 528–535. [Google Scholar] [CrossRef] [Green Version]
  122. Kanakry, C.G.; Tsai, H.-L.; Bolaños-Meade, J.; Smith, B.D.; Gojo, I.; Kanakry, J.A.; Kasamon, Y.L.; Gladstone, D.E.; Matsui, W.; Borrello, I.; et al. Single–agent GVHD prophylaxis with posttransplantation cyclophosphamide after myeloablative, HLA–matched BMT for AML, ALL, and MDS. Blood 2014, 124, 3817–3827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. DeZern, A.E.; Elmariah, H.; Zahurak, M.; Rosner, G.L.; Gladstone, D.E.; Ali, S.A.; Huff, C.A.; Swinnen, L.J.; Imus, P.; Borrello, I.; et al. Shortened–Duration Immunosuppressive Therapy after Nonmyeloablative, Related HLA–Haploidentical or Unrelated Peripheral Blood Grafts and Post–Transplantation Cyclophosphamide. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2020, 26, 2075–2081. [Google Scholar] [CrossRef] [PubMed]
  124. Kasamon, Y.L.; Fuchs, E.J.; Zahurak, M.; Rosner, G.L.; Symons, H.J.; Gladstone, D.E.; Huff, C.A.; Swinnen, L.J.; Brodsky, R.A.; Matsui, W.H.; et al. Shortened–Duration Tacrolimus after Nonmyeloablative, HLA–Haploidentical Bone Marrow Transplantation. BBMT 2018, 24, 1022–1028. [Google Scholar]
  125. Locatelli, F.; Merli, P.; Rutella, S. At the Bedside: Innate immunity as an immunotherapy tool for hematological malignancies. J. Leukoc. Biol. 2013, 94, 1141–1157. [Google Scholar] [PubMed]
  126. Locatelli, F.; Merli, P.; Pagliara, D.; Pira, G.L.; Falco, M.; Pende, D.; Rondelli, R.; Lucarelli, B.; Brescia, L.P.; Masetti, R.; et al. Outcome of children with acute leukemia given HLA–haploidentical HSCT after alphabeta T–cell and B–cell depletion. Blood 2017, 130, 677–685. [Google Scholar] [CrossRef]
  127. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, Activity, and Immune Correlates of Anti–PD–1 Antibody in Cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef]
  128. Davids, M.S.; Kim, H.T.; Bachireddy, P.; Costello, C.; Liguori, R.; Savell, A.; Lukez, A.P.; Avigan, D.; Chen, Y.-B.; McSweeney, P.; et al. Ipilimumab for Patients with Relapse after Allogeneic Transplantation. N. Engl. J. Med. 2016, 375, 143–153. [Google Scholar]
  129. Davids, M.S.; Kim, H.T.; Costello, C.; Herrera, A.F.; Locke, F.L.; Maegawa, R.O.; Savell, A.; Mazzeo, M.; Anderson, A.; Boardman, A.P.; et al. A multicenter phase 1 study of nivolumab for relapsed hematologic malignancies after allogeneic transplantation. Blood 2020, 135, 2182–2191. [Google Scholar] [CrossRef]
  130. Merryman, R.W.; Kim, H.T.; Zinzani, P.L.; Carlo-Stella, C.; Ansell, S.M.; Perales, M.-A.; Avigdor, A.; Halwani, A.S.; Houot, R.; Marchand, T.; et al. Safety and efficacy of allogeneic hematopoietic stem cell transplant after PD–1 blockade in relapsed/refractory lymphoma. Blood. 2017, 129, 1380–1388. [Google Scholar] [CrossRef] [Green Version]
  131. Paul, S.; Zahurak, M.; Luznik, L.; Ambinder, R.F.; Fuchs, E.J.; Bolaños-Meade, J.; Wagner-Johnston, N.; Swinnen, L.J.; Schoch, L.; Varadhan, R.; et al. Non–Myeloablative Allogeneic Transplantation with Post–Transplant Cyclophosphamide after Immune Checkpoint Inhibition for Classic Hodgkin Lymphoma: A Retrospective Cohort Study. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2020, 26, 1679–1688. [Google Scholar] [CrossRef] [PubMed]
  132. Oran, B.; Garcia-Manero, G.; Saliba, R.M.; Alfayez, M.; Al-Atrash, G.; Ciurea, S.O.; Jabbour, E.J.; Mehta, R.S.; Popat, U.R.; Ravandi, F.; et al. Posttransplantation cyclophosphamide improves transplantation outcomes in patients with AML/MDS who are treated with checkpoint inhibitors. Cancer 2020, 126, 2193–2205. [Google Scholar] [CrossRef] [PubMed]
  133. 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]
  134. Keller, M.D.; Bollard, C.M. Virus–specific T–cell therapies for patients with primary immune deficiency. Blood 2020, 135, 620–628. [Google Scholar] [CrossRef] [PubMed]
  135. Creidy, R.; Moshous, D.; Touzot, F.; Elie, C.; Neven, B.; Gabrion, A.; Ville, M.-L.; 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.e3. [Google Scholar]
  136. Bao, L.; Cowan, M.J.; Dunham, K.; Horn, B.; McGuirk, J.; Gilman, A.; Lucas, K.G.N. 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] [Green Version]
  137. Heslop, H.E.; Slobod, K.S.; Pule, M.A.; Hale, G.A.; Rousseau, A.; Smith, C.A.; Bollard, C.M.; Liu, H.; Wu, M.-F.; Rochester, R.J.; 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, 925–935. [Google Scholar] [CrossRef] [Green Version]
  138. Papadopoulou, A.; Gerdemann, U.; Katari, U.L.; Tzannou, I.; Liu, H.; Martinez, C.; Leung, K.; Carrum, G.; Gee, A.P.; Vera, J.F.; 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, 242ra283. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Elmariah, H.; Brunstein, C.G.; Bejanyan, N. Immune Reconstitution after Haploidentical Donor and Umbilical Cord Blood Allogeneic Hematopoietic Cell Transplantation. Life 2021, 11, 102. https://0-doi-org.brum.beds.ac.uk/10.3390/life11020102

AMA Style

Elmariah H, Brunstein CG, Bejanyan N. Immune Reconstitution after Haploidentical Donor and Umbilical Cord Blood Allogeneic Hematopoietic Cell Transplantation. Life. 2021; 11(2):102. https://0-doi-org.brum.beds.ac.uk/10.3390/life11020102

Chicago/Turabian Style

Elmariah, Hany, Claudio G. Brunstein, and Nelli Bejanyan. 2021. "Immune Reconstitution after Haploidentical Donor and Umbilical Cord Blood Allogeneic Hematopoietic Cell Transplantation" Life 11, no. 2: 102. https://0-doi-org.brum.beds.ac.uk/10.3390/life11020102

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