Abstract
Introduction
With the use of reduced-intensity conditioning (RIC), early toxicity of allogeneic stem cell transplantation (SCT) has been much reduced. Graft-versus-host disease (GvHD) causes morbidities and mortality. Alemtuzumab is a mAb directed against CD52. When administered prior to transplant it leads to T-cell depletion. Incorporation of alemtuzumab in RIC results in low rates of GvHD and treatment-related mortality (TRM) in haematological diseases, even in the setting of mismatched-donor transplantation.
Areas covered
The use of alemtuzumab for GvHD in SCT. The benefit of alemtuzumab-based conditioning is partially offset by increased disease relapse due to impaired graft-versus-tumor effect (GvT) and by slower immune reconstitution, necessitating special precautions. While GvHD is prevented with alemtuzumab, post-SCT interventions are often required. Most studies find that alemtuzumab-based conditioning results in decreased chronic GvHD and TRM, but also in decreased progression-free survival. Overall survival after 3 – 5 years is usually equivalent and quality of life may be improved because of a lower incidence of sequelae of chronic GvHD. Many aspects of alemtuzumab treatment are under investigation.
Expert opinion
Alemtuzumab reduces GvHD and TRM after SCT. Use of alemtuzumab requires awareness and strict management of the risk of opportunistic infections and of an increased risk of disease recurrence.
Keywords: alemtuzumab, allogeneic stem cell transplantation, graft-versus-host disease, T-cell depletion
1. Introduction
Allogeneic hematopoietic stem cell transplantation (SCT) is an important treatment option for haematological malignancies as well as for some non-malignant diseases, but carries a considerable risk for complications that are life-threatening and/or affect quality of life. In recent decades, improvements in conditioning regimens and supportive care have resulted in steady reduction of treatment related mortality (TRM) [1] but graft-versus-host disease (GvHD) remains a problem [2], and has become even more prominent in the era of reduced-intensity conditioning (RIC) which relies on graft-versus-tumor (GvT) effects to achieve tumor response. Acute GvHD usually occurs within the first months after transplant and affects mainly the skin, gastrointestinal tract and liver. It occurs in 25% – 70% of patients, depending on the type of donor and conditioning. Chronic GvHD, which has distinct clinical features and tends to develop later occurs in 30% – 50% of patients and is the leading cause of late mortality after SCT [2,3]. GvHD physiopathology is complex but is mediated to a large extent by alloreactive donor T-cells [4]. All stem cell transplants require GvHD prophylaxis, usually based on blocking of T-cell function with calcineurin inhibitors [5]. An alternative and more efficient method of GvHD prophylaxis consists of the removal of donor T-cells in vitro through physical methods [6], the use of anti-lymphocyte antibodies [7] or column-based immunomagnetic selection of specific cell populations [8]. Lastly, GvHD can be prevented by in vivo administration of antibodies that lyse lymphocytes, in particular alemtuzumab or anti-thymocyte globulin (ATG).
Alemtuzumab is a humanized anti-CD52 monoclonal antibody, which effectively depletes both B and T cells from circulating blood with limited or no effect on hematopoietic progenitors [9]. The first anti-CD52 antibody was developed in the Cambridge Pathology-1 lab (CAMPATH-1) as a tool to deplete donor T-cells before SCT [4]. The original CAMPATH molecules were rat-derived antibodies and included an IgM antibody (CAMPATH-1M), and subsequently an IgG antibody (CAMPATH-1G) both of which were studied for in vitro and in vivo T-cell depletion respectively. Both caused significant reduction in GvHD [7], but their benefit was offset by an increased risk of graft rejection caused by residual host T-cells and an increased risk of relapse due to the impaired GvT effect [7]. Subsequently, a humanized antibody was engineered (CAMPATH-1H or Alemtuzumab) to decrease immunogenicity and for myriad clinical applications [4]. It has potent activity in chronic lymphocytic leukemia (CLL), and is approved for CLL therapy [9]. It also has unique activity in various T-cell lymphomas in particularly in T-prolymphocytic leukemia (T-PLL) [10]. It is used for treatment of severe aplastic anemia (AA) [11] and has shown remarkable benefit in multiple sclerosis [12]. In organ transplantation, alemtuzumab has shown promising results in tolerance induction [13–16]. Alemtuzumab also continues to be widely used in many countries as a very effective method for prevention of acute and especially chronic GvHD after transplantation that is widely used in the United Kingdom and in many other centers around the world [2,17,18].
Relapse and delayed immune reconstitution remain concerns of this method and are the reasons why its application in SCT is not universally accepted [2]. Small series have also reported its role in acute GvHD therapy [19]. In this review, we provide an overview of the current role and recent data on alemtuzumab in SCT.
2. Structure and mechanism of action
Alemtuzumab is a recombinant humanized monoclonal IgG1 antibody directed against the CD52 antigen, a 12 amino acid, 28,000 molecular weight glycosylated glycosylphosphatidylinositol (GPI)-linked cell surface protein [20]. The function of CD52 remains largely unknown but it is expressed on more than 95% of peripheral blood lymphocytes, monocytes, eosinophils and macrophages and on some dendritic cells but not on granulocytes, red blood cells, platelets or hematopoietic progenitor cells [21–23]. It is also expressed in the male reproductive tract where CD52 is necessary for spermatozoa to preserve normal motility [24]. CD52 antigen density is higher on normal T lymphocytes than on normal B lymphocytes, a pattern of expression recapitulated on T and B neoplasms [21,22]. Differences in CD52 expression may explain differential sensitivity to alemtuzumab in vitro and in vivo. Indeed, alemtuzumab depletes peripheral B and T cells effectively but the pattern of recovery varies among the different subsets. Memory T cells are partially spared by alemtuzumab [14] while CD8+ cells recover more rapidly than CD4+ cells [16,25,26]. NK cells are effectively depleted by alemtuzumab but tend to recover rapidly after SCT [25,27]. Alemtuzumab also removes monocyte-derived dentritic cells from the circulation. This decreases phagocytosis and presentation of host-derived antigens which probably contributes to further reduction in GvHD early after SCT [28]. Most of the CD52 is membrane bound, but in CLL, a disease characterized by high expression of CD52 on the tumor cells, some of the antigen is shed. As a result, soluble CD52 can readily be detected in the plasma of patients with CLL and may modulate response to alemtuzumab in vivo [24].
The mechanism by which alemtuzumab mediates lympholysis is complex and includes complement-mediated cell lysis (complement-dependent cytotoxicity (CDC)), antibody-dependent cellular cytotoxicity (ADCC) and direct apoptosis [29]. Depending on experimental conditions, in vitro studies have found a prominent effect of the complement pathway [30], a strong ADCC through a caspase-dependent pathway [31] or a direct caspase-independent apoptotic pathway [32]. In a human CD52-transgenic mouse a major role for ADCC in lymphocyte depletion has been shown, with neutrophils and NK cells as potential effectors [26,33]. However, high-affinity IgG Fc eceptor (FCGR) polymorphisms are not correlated with clinical response to alemtuzumab in CLL, suggesting that in vivo its mechanism of action is not limited to ADCC [34].
3. Pharmacology and dosing of alemtuzumab in transplant protocols
The incorporation of alemtuzumab in transplant protocols has a dual purpose, namely reduction in GvHD (both acute and chronic) and the prevention of graft rejection. Its major side-effects are immune suppression, resulting in opportunistic infections and increased risk for recurrence because of reduction in GvT effects. The dosing schedule of alemtuzumab with optimal efficacy and minimal side-effects has been the subject of much empirical research.
In many transplant protocols, alemtuzumab is administered intravenously during conditioning similar to the original CAMPATH-1G though alemtuzumab has slower clearance than CAMPATH-1G and is therefore a more potent immunosuppressant [35]. It is practically always combined with single agent post-transplant prophylaxis consisting of a calcineurin inhibitor (cyclosporin A or tacrolimus). Post-transplant methotrexate is not usually administered in these protocols and this accounts for a low incidence of mucositis. Alemtuzumab clearance depends on the actual dose administered, the total number of CD52 binding sites, the hepatic function and also the monocyte–macrophage system. In practice though, most centers have used fixed dose-schedules of alemtuzumab. After the commonly used schedule of alemtuzumab 20 mg daily for five days (total 100 mg), a median absolute lymphocyte CD4+ level higher than 200×106/l was not reached until 9 months after SCT. The terminal half-life of alemtuzumab was 8 days and time to achieve levels below 0.1 μg/ml was estimated at 60 days. Concentrations as low as 0.1 μg/ml are sufficient for ADCC and therefore ongoing lymphocyte depletion might occur for approximately 2 months after SCT [36].
Based on concerns for impaired immune reconstitution and GvT effect, some investigators have attempted to lower the dose of alemtuzumab. Tholouli et al. reported 98 acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) patients who received either 50 or 100 mg alemtuzumab as part of RIC. Except for a trend for lower relapse with 50 mg, no differences were observed in terms of overall survival (OS), TRM and cytomegalovirus (CMV) reactivation [37]. Alemtuzumab doses as low as 10 mg may result in prolonged lymphopenia [38] and hence even lower doses have been explored. Bertz et al. reported that further de-escalation of in vivo alemtuzumab to 10 – 20 mg total dose given on day –1 was safe. There were no increases in severe acute and chronic GvHD or graft failure. There also did not seem to be an effect on relapse [39]. These data were confirmed by Spyridonidis et al. in a small study of 18 patients who received 10 mg alemtuzumab before sibling and matched unrelated SCT [40]. Chakraverty et al. also demonstrated that dose reduction of alemtuzumab to 30 mg given on day -1 effectively prevents GvHD and is associated with more rapid lymphocyte recovery after HLA-identical sibling SCT [41]. In their experience however, a dose of 20 mg or lower was associated with incomplete saturation of CD52 binding sites, more rapid clearance and greater risk of severe acute and chronic GvHD. In general, a dose of 30 mg or lower is associated with a significant fall in alemtuzumab level by day 28. Others reported concerns about increased incidence of Epstein-Barr virus (EBV)-driven post-transplant lymphoproliferative disease (PTLD) following a reduction in alemtuzumab total dose from 50 to 30 mg. Low-dose alemtuzumab may be insufficient to deplete B cells, allowing EBV-infected B cells to survive and potentially lead to PTLD [42]. In summary, the standard total dose of 100 mg seems more than is required in most transplant settings. Doses of less than 30 mg may be insufficient and predispose to increased risks of GVHD and of PTLD.
Biodistribution and clearance of alemtuzumab are further influenced by a high burden of CD52 antigen on active tumor, or soluble CD52 circulating in the bloodstream [43]. This has implications for transplant in highly CD52-positive neoplasms, particularly CLL. Presence of residual disease at the time of transplantation increases antigen concentration and shortens alemtuzumab plasma half-life. In this setting, the standard dose of alemtuzumab used in RIC might be insufficient to prevent graft rejection and GvHD [44,45]. Indeed, Delgado et al. reported a high incidence of graft failure (15%) after alemtuzumab-based RIC in CLL patients [46].
Subcutaneous (SQ) alemtuzumab has gained widespread usage in the treatment of CLL because it is associated with fewer infusion-related side effects and similar pharmacology [43]. Juliusson et al. administered SQ alemtuzumab at a dose of 30 mg × 3 to 26 patients and as a single dose of 30 mg to 14 patients. Immediate toxicity was minimal, and the lower dose of alemtuzumab was associated with low incidence of GvHD and excellent immune recovery [47].
Some groups, instead of administering alemtuzumab in vivo, have added it to the stem cell infusate at a usual dose of 20 mg (so-called ‘Campath in the bag’) [48,49]. This results in lysis of donor lymphocytes in the infusate. Since the product is infused without further manipulation, some of the alemtuzumab is also administered to the recipient and results in host immunosuppression [50]. This simple technique has been associated with a low incidence of both acute and chronic GvHD and low TRM [50–53]. Immune reconstitution remains an issue and some investigators have decreased the dose to 10 mg or 1 mg/1010 mononuclear cells with similar GvHD prophylaxis but fewer infectious complications [52,53]. The issue of graft rejection reported in the initial studies of Campath-1M, is no longer of much concern, because of the longer half-life of alemtuzumab, which persists for some time in the recipient, a higher stem cell dose and a more potent immunosuppresion of the recipient with fludarabine [52].
4. Clinical efficacy in allogeneic transplant for hematologic malignancies and related disorders
The most consistent benefit of alemtuzumab-containing conditioning regimens is the effective and dramatic reduction in acute and chronic GvHD and consequently low TRM [2,7,17]. While other RIC SCT report a 40% – 60% incidence of grade II – IV acute GvHD, most studies of alemtuzumab-containing regimens have observed an incidence of 10 to 20% acute GvHD, a low incidence of extensive chronic GvHD and a TRM of 10% – 20% [2,17,18]. The reduction in GvHD also applies to unrelated donor transplant [17,18]. Mead et al., in a large retrospective study report that alemtuzumab may overcome the adverse effectct of HLA mismatch and an 8 – 9/10 HLA-mismatched unrelated donor may be a reasonable option in this setting [54]. Alemtuzumab has also been administered after RIC haplo-identical SCT with very low rates of GvHD (16%) [55] but long-term outcomes remain disappointing [56]. Results from selected studies are reported in Tables 1 and 2.
Table 1.
Studies on Alemtuzumab-based RIC SCT in unselected diseases and AML/MDS
Study | Diagnosis | Nr | Disease Status | Age | Conditioning | Campath Dose | % Sib | TRM | Grade III – IV aGvHD | Extensive cGvHD | OS | DFS/PFS | DLI |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
[18] | All | 44 | 39% CR | 18 – 56 | Flu-Mel | 100 mg | 82% | 11% | No grade III – IV | Not extensive | 1y-73% | 1y-71% | Only for relapse |
[17] | All | 47 | 34% CR | 18 – 62 | Flu-Mel | 100 mg | 0% | 19.8% | 6.4% | Not extensive | 1y-75% | 1y-61.5% | 6/47 |
[2] | All | 78 | 20% CR | 44 | Flu-Mel | 100 mg | 100 % | 10.2% | 2.6% | 1/78 | 1y-72% | 1y-76% | 18/78 |
[50] | All | 73 | - | 19 – 61 | Cy-TBI | 20 mg in the bag | 100 % | 8% | No grade III – IV | 13/73 | 5y-48% | 5y-43% | 37/73 |
[54] | All | 157 | 79% S | 48 | Flu-Mel | 100 mg | 0% | 27% | 3% | 11.5% | 3y-52% | – | 44/157 |
[57] | AML/MDS | 62 | 56% CR | 53 | Flu-Bu | 100 mg | 39% | 15% | 9% | 2/62 | 2y-74% | 2y-62% | 26/62 |
[58] | AML/MDS | 76 | 55% CR | 52 | Flu-Mel | 50 – 100 mg | 46% | 19% | No grade III – IV | 2/76 | 3y-41% | 3y-37% | 9/76 |
[59] | AML/MDS | 95 | 50% CR | 54 | Flu-Mel | 100 mg | 50% | 24.6% | 8.6% | 16% both limited and extensive | 2y-40.5% | 2y-33% | No |
[62] | AML/MDS | 110 | 51% CR1 | 53 | Flu-Bu | 100 mg | 33% | 21% | 10.9% | 16% | 2y-59% | 2y-57% | 39/110 |
[63] | AML | 51 | 94% CR | 51 | Flu-Mel, Flu-Bu, Flu-Cy | 30 – 100 mg | 100 % | 12% | 14% grade II – IV | 4% | 5y-60% | – | – |
Age: median age or range, aGvHD: cumulative incidence of grade III-IV acute graft-versus-host disease, All: unselected haematological malignancies, AML: acute myelogenousleukemia, Bu: busulfan, Campath dose: cumulative dose of alemtuzumab, cGvHD: cumulative incidence of extensive chronic graft-versus-host disease, CR: complete remission, CR1: acute leukemia in first remission, Cy: cyclophosphamide, DFS: disease-free survival, Disease Status: disease status at the time of transplantation, DLI: donor lymphocyte infusion given for relapse or mixed chimerism, Flu: Fludarabine, Mel: melphalan, MDS: myelodysplastic syndrome, OS: overall survival, Nr: numbers of patients, PFS: progression-free survival, RIC: reduced-intensity conditioning, S: chemosensitive disease, SCT: allogeneic stem cell transplantation, % Sib: proportion of sibling donors, TBI: total body irradiation, TRM: treatment-related mortality 1 year after transplantation
Table 2.
Major studies on alemtuzumab-based RIC SCT in lymphoproliferative diseases
Study | Diagnosis | Nr | Disease status | Age | Conditioning | Campath Dose | % Sib | TRM | Grade III – IV aGvHD | Extensive cGvHD | OS | DFS/PFS | DLI |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
[67] | NHL | 88 | 88.6% S | 48 | Flu-Mel | 100 mg | 71% | 23% | 4.5% | 4.5% | 3y-55% | 3y-50% | 36/88 |
[68] | HG-NHL | 48 | 83% S | 46 | Flu-Mel | 20 – 100 mg | 62% | 29% | 4.2% | 13% | 4y-47% | 4y-48% | 12/48 |
[77] | NHL, HL | 67 | 66% S | 54 | Flu-Mel, Clo-Mel, Flu-Bu | 100 mg | 57% | 30% | 16.4% grade II – IV | 7/67 | 3y-47% | 3y-30% | 3/67 |
[69] | MCL | 70 | 81% S | 48 | Flu-Mel, BEAM, Flu-Bu | 30 – 100 mg | 60% | 18% | 10% | 34% | 5y-37% | 5y-14% | 27/70 |
[46] | CLL | 41 | 83% S | 54 | Flu-Mel | 40 – 100 mg | 58% | 26% | 10% | 4.9% | 2y-51% | 5y-45% | 18/41 |
[70] | FL | 82 | 90% S | 45 | Flu-Mel | 20 – 100 mg | 46% | 12% | 13% grade II – IV | 20% | 4y-76% | 4y-76% | 41/82 |
[79] | FL | 44 | 90.9% S | 48 | BEAM | 50 – 100 mg | 64% | 20% | No grade III – IV | 6.8% | 3y-69% | 3y-58% | 13/44 |
[78] | NHL, CLL, HL | 65 | 20% CR | 46 | BEAM | 50 – 100 mg | 87% | 13% | No grade III – IV | No extensive | 3y-63% | 3y-54% | 17/65 |
[73] | HL | 36 | 55.5% S | 36 | Flu-Mel | 50 – 100 mg | 100% | 7% | No grade III – IV | 2/36 | 4y-62% | 4y-39% | 19/36 |
Age: median age, aGvHD: cumulative incidence of grade III-IV acute graft-versus-host disease, BEAM: bis-chloroethylnitrosourea, Etoposide, Ara-C, Melphalan, Bu: busulfan, Camp: campath/alemtuzumab, Camp dose: cumulative dose of alemtuzumab, cGvHD: cumulative incidence of extensive chronic graft-versus-host disease, CLL: chronic lymphocytic leukemia, CR: complete remission, DFS: disease-free survival, Disease Status: disease status at the time of transplantation, DLI: donor lymphocyte infusion given for relapse or mixed chimerism, FL: follicular lymphoma, Flu: Fludarabine, HG-NHL: High-grade non-Hodgkin’s lymphoma, HL: Hodgkin’s lymphoma, MCL: mantle cell lymphoma, Mel: melphalan, NHL: non-Hodgkin’s lymphoma, Nr: numbers of patients, OS: overall survival, PFS: progression-free survival, RIC: reduced-intensity conditioning, S: chemosensitive disease, SCT: allogeneic stem cell transplantation, % Sib: proportion of sibling donor, TRM: treatment-related mortality 1 year after transplantation
Early on, it was observed that the efficacy of alemtuzumab in preventing GvHD was associated with a higher rate of mixed chimerism and a higher rate of disease recurrence [7]. The balance between increased risk of disease recurrence and decreased GvHD determines the overall utility of alemtuzumab-based conditioning in the various diseases that are discussed below.
AML and MDS can be cured with SCT but myeloablative conditioning is restricted to younger patients because of excess TRM and GvHD in older patients. Various RIC have been tested in older patients without a consensus on conditioning or GvHD prophylaxis. Studies using an alemtuzumab-based RIC SCT have TRM from 15% to 25% (Table 1) and have a very low incidence of severe acute and chronic GvHD [57–62]. We reported our own experience with 95 AML/MDS patients and compared them with 59 patients who received identical conditioning without alemtuzumab. Acute GvHD (23% grade II – IV) and especially chronic GvHD (16%) were effectively reduced by the addition of alemtuzumab. We found a non-significant increase in recurrence rates (24%) with alemtuzumab, and a non-significant decrease in TRM (25%). OS was nearly identical between both groups [59,60]. Another recent comparison between alemtuzumab and no alemtuzumab has been reported by Malladi et al. They found a trend toward less severe acute GvHD (14%) but a significant reduction in extensive chronic GvHD (4%) for alemtuzumab patients. There were no significant differences in non-relapse mortality and relapse risk between both groups [63]. Outcomes were significantly correlated with active disease at the time of transplant, adverse risk cytogenetics and increased intensity of post-transplant immunosuppression [64]. Collectively, alemtuzumab-based RIC is of interest in patients with AML and MDS due to its strong effect on extensive chronic GvHD. Relapse may be slightly increased, but this drawback is more than offset by a modest reduction in TRM and a major reduction in GvHD-related morbidity.
CD52 is also expressed on 66% – 78% of acute lymphoblastic leukemia (ALL) cells and alemtuzumab as part of the conditioning offers the dual benefit of reduced GvHD with a potential anti-leukemia effect. Primary results in a small cohort of 48 patients are encouraging [65].
Subgroups of patients with aggressive non-Hodgkin’s Lymphoma (NHL) are considered routinely for allogeneic transplant. This includes those who have refractory disease or patients relapsing after autologous transplantation. Myeloablative SCT in this setting is associated with very high TRM [66]. RIC may have advantages particularly in patients with comorbidities, but has considerable rates of disease recurrence. The incorporation of alemtuzumab in RIC result in significantly lower rates of acute (10% – 17% grade II – IV) and chronic GvHD (7% – 18%) but similar overall survival (Table 2), which is determined mainly by the disease status and patient’s condition [46,67–72]. Similar to aggressive NHL, SCT for Hodgkin’s lymphoma (HL) is mainly used for failure of autologous SCT and is also associated with very high TRM using standard conditioning. Alemtuzumab-based RIC has low TRM (16%) and has shown very promising results in this situation, particularly for those with chemosensitive relapse [73–75]. SCT may have a more important role in those lymphoproliferative disorders that have frequent bone marrow involvement including mantle cell lymphoma, follicular lymphoma and CLL. Follicular lymphoma has a very low rate of recurrence after myeloablative allogeneic transplant. This favorable outcome is variously attributed to GvT effects or to the combination of myeloablative conditioning with the infusion of a tumor-free graft [76]. RIC conditioning has less regimen-related toxicity, but increased complications due to GvHD, resulting in overall nearly similar TRM. Alemtuzumab-containing conditioning has shown a low incidence of GvHD, low TRM and excellent long term survival [17,67,70,77]. Some of the best results were reported by Faulkner et al. who used a bis-chloroethylnitrosourea, etoposide, Ara-C, melphalan (BEAM)-alemtuzumab conditioning with low rates of TRM (8%) and GvHD (17%) [78,79]. After the more commonly used fludarabine–melphalan conditioning, late recurrence is frequent but can often be successfully managed by donor lymphocyte infusions (DLI) or low intensity chemo-immunotherapy [70,77]. Similar observations apply to mantle cell lymphoma (MCL), where a high relapse rate was observed but DLI induced a significant number of sustained responses [69].
The role of alemtuzumab in conditioning for chronic leukemias or in myeloma is more controversial. In CLL, a higher rate of graft failure was observed with alemtuzumab-based RIC, and a higher TRM (26%) related to frequent and fatal infections [46]. Profound pre-existing immunosuppression in patients with advanced CLL may contribute to the higher complication rates [46]. In one series, patients receiving alemtuzumab-based conditioning had worse long-term survival. The small number of such patients and the non-random assignment of patients to particular conditioning regimens in this study precluded firm conclusions on the role of alemtuzumab in the poor outcomes [80]. In chronic myelogenous leukemia (CML), addition of alemtuzumab was associated with a higher relapse rate [81] but these recurrences can be effectively salvaged with DLI and/or tyrosine kinase inhibitors (TKI). Results in advanced-phase CML were disappointing [82]. In multiple myeloma (MM), a large retrospective study from Crawley et al. identifies alemtuzumab as a predictor for reduced chronic GvHD but also increased relapse rate [83]. Progression-free survival was reduced in patients receiving alemtuzumab-based conditioning, but OS was not.
In severe aplastic anemia, graft rejection and chronic GvHD are major obstacles to successful outcome after SCT. Alemtuzumab can effectively reduce GvHD with low rate of graft rejection [84]. Sickle cell anemia is another non-malignant disease where SCT may provide long-term survival. GvHD is particularly unwelcome in this situation and alemtuzumab has been successfully incorporated in RIC [85]. Finally, graft failure is a rare complication after SCT but carries a dismal prognosis. Risk factors for graft failure include HLA-mismatch, T-cell depletion, alloimmunization, RIC and poor graft quality [86]. Alemtuzumab associated with fludarabine induces substantial immunosuppression allowing second transplant with high engraftment rate and minimal toxicity [87].
Alemtuzumab may also have a role in the management of acute and chronic GvHD, though only small studies and preliminary results have been reported. Different schedules and doses have been used with a response observed in 55% – 78% of patients and complete response in 33% – 35%. The best responses were observed in gastrointestinal GVHD. As expected, infectious complications were common with invasive aspergillosis and CMV reactivation the most frequent. Lower doses of alemtuzumab (3 – 10 mg every 14 days) may be sufficient for GvHD therapy and have fewer infectious complications [19,88,89]. Reports on the use of alemtuzumab in chronic GvHD are even more anecdotal [90].
5. The role of salvage therapy and donor lymphocyte infusion to prevent or treat recurrence in alemtuzumab-based conditioning
T-cell depletion in general, including that mediated by alemtuzumab, has been recognized for many years as a risk factor for disease recurrence after SCT [5] which is usually managed with further chemotherapy and/or DLI. DLI can induce remissions through mediation of a GvT effect, but carries the risk of aplasia and GvHD, only partially prevented by dose reduction of DLI [91,92]. Responses to DLI have been reported in different diseases but while very good response rates are observed in indolent NHL and chronic-phase CML, efficacy is limited in acute leukemia or aggressive NHL [46,61,67–70,73]. Other therapeutic approaches such as reinduction chemotherapy in AML [93], TKIs’ in CML [82] or chemo-immunotherapy in NHL [77] are also often remarkably effective and can induce prolonged responses. The ability to effectively salvage patients after relapse distinguishes alemtuzumab-based conditioning from other RIC transplants and explains the observation that in many analyses alemtuzumab predisposes for recurrence, but not for decreased overall survival. The UK cooperative group in this regard has published interesting results with excellent long term survival outcomes, and has coined the term ‘current progression-free survival’ to describe lymphoma patients who experienced relapse after SCT but who, with DLI and small doses of rituximab obtained durable subsequent remissions [70]. In the calculation of ‘current progression-free survival’, requirement for DLI is not considered a censoring event.
Mixed T-cell chimerism is frequent after alemtuzumab-based RIC and has been associated with tolerance induction and with less GvHD. Some groups have used mixed chimerism as an indicator for imminent disease progression and have administered prophylactic DLI to such patients [17,62,75]. This has frequently resulted in full donor chimerism, and such patients have done well. Conversion to full donor chimerism has been postulated as an effective means to decrease relapse and to obtain prolonged disease control [70,75]. The studies were, however, not conducted in a prospective fashion and, selection and attrition bias may have played a major role in the outcome of those receiving prophylactic DLI [92]. Somewhat surprisingly in one study of Hodgkin’s lymphoma [75], recurrence rates were lower in those receiving prophylactic DLI for mixed chimerism than in those who were fully chimeric and who did not receive DLI [75]. Others, including our own group, have found that stable mixed chimerism is not a predictor for relapse and is possibly associated with better outcome because it predicts for less GvHD [62]. By contrast, a decline of more than 15% in T-cell chimerism between day 30 and day 180 predicted disease recurrence [94]. Close monitoring of full and T-cell chimerism is then mandatory to help guide therapeutic intervention after alemtuzumab-based RIC.
6. ATG versus Alemtuzumab
ATG is the other effective agent for in vivo T-cell depletion. Several formulations of ATG are available. They are polyclonal rabbit or horse immunoglobulins directed against multiple human lymphocyte cell-surface antigens. ATG binds to T-cells resulting in T-cell depletion via opsonisation and lysis following complement activation [95]. There may be considerable differences between the various ATG formulations (Thymoglobulin, ATG-Fresenius and ATGAM) but they are all polyclonal products directed against T-cells, in contrast to alemtuzumab which is a monoclonal antibody that depletes both T-and B-cells. Both ATG and alemtuzumab allow rapid engraftment and reduce GvHD effectively but the relative merits of these agents remain a matter of debate and ongoing study. In a comparative study in myeloma, alemtuzumab was associated with a lower rate of complete remission and a higher incidence of CMV reactivation than with ATG. No difference in relapse was found [96]. Juliusson et al. replaced prospectively ATG with alemtuzumab given subcutaneously for 3 days in their RIC protocol. They showed an attractive short-term toxicity profile for alemtuzumab but with slower lymphocyte recovery and with more mixed chimerism. The greater need for DLI and the higher incidence of opportunistic and fatal infections resulted in an impaired OS compared with ATG. They subsequently changed their protocol to one day alemtuzumab (30 mg) given on day –5 which translated into the same OS but with less toxicity than with ATG [47]. In a small retrospective study, Norlin et al. showed that alemtuzumab was associated with less overall acute GvHD but more chronic GvHD than ATG. Nevertheless, similar OS, relapse-free survival and infections rate were observed [97]. Finally, the International Bone Marrow Transplantation Registry (IBMTR) reported a large analysis comparing alemtuzumab, ATG and T-cell-repleted RIC transplants. Despite a significant reduction of both acute and chronic GvHD, T-cell depletion was associated with lower disease-free survival. The lowest OS was observed with ATG [98]. Possibly the most important advantage of alemtuzumab over ATG is the relatively low risk of EBV PTLD associated with alemtuzumab exposure [99] due to the effective B cell depletion by alemtuzumab. The respective effect on long term incidence of GvHD, relapse and survival of ATG versus alemtuzumab will require further prospective study.
7. Safety
Alemtuzumab is generally well tolerated with immediate toxicity mainly related to first-dose reactions, including fever, rigor and skin rash. Those reactions decrease with subsequent dosing by administering pre-treatment antihistamines and acetaminophen, and by slowing the rate of injection [29]. Intewrestingly, the first dose of alemtuzumab in transplant conditioning is usually administered as a full dose of 20 mg, with steroid prophylaxis, in contrast to the dose-escalation schemes often followed in CLL treatment. The subcutaneous route is associated with a better safety profile but rarely used in transplant [43]. A major concern with the use of alemtuzumab is the impaired immune reconstitution. Lymphopenia is often prolonged. CD4+ cell recovery is very slow, while CD8+ and particularly NK cells increase more rapidly [100,101]. This prolonged immunosuppression leads to an increased risk of opportunistic infections and requires close monitoring. CMV reactivation is very common and occurs early after SCT [102]. CMV reactivation has been observed in 30% to 85% of patients in different studies but CMV disease remains rare due to an effective pre-emptive strategy [2,17,46,57–59,62,67–70,73]. Because of a relative rapid CD8+ cell recovery, CMV-specific T-cells increase after episodes of CMV reactivation [103]. In combination with PCR-based detection of CMV DNA and prompt ganciclovir and/or valganciclovir therapy, most CMV reactivation remains self-limited [104]. At The University of Chicago, we use a very aggressive prophylactic strategy to prevent CMV reactivation. All high-risk patients receive ganciclovir (5 mg/kg intravenously twice daily from day –7 until day –2), acyclovir (500 mg/m2 every 8 hours intravenously from day –2 until engraftment) followed by high dose oral valacyclovir (2 g four times daily until day +210). With this well-tolerated strategy, CMV reactivation was decreased to 29% [105]. Beside CMV infection, we have also screened our patients for BK virus and did not find a higher incidence in patients receiving alemtuzumab [106]. Incorporation of alemtuzumab in RIC has also been associated with an increased incidence in EBV infection but low risk of PTLD due to an effective preemptive therapy with rituximab but also due to an effective depletion of B cells with alemtuzumab [42,107,108]. Finally, T-cell depletion with alemtuzumab is a recognized risk factor for adenovirus infections after SCT, particularly in children [109].
8. Conclusion
Alemtuzumab is a well-tolerated and effective drug when used for T-cell depletion before SCT. Its application in RIC results in a significant reduction of both acute and chronic GvHD and lower TRM. Graft rejection, a considerable problem in studies of earlier CAMPATH products, no longer represents much of an issue with alemtuzumab. In vivo administration may actually have graft-enhancing properties. Alemtuzumab has a prolonged half-life, but its pharmacokinetics is influenced by CD52 density on target cells, residual CD52 positive disease and circulating soluble CD52. The customary dosage of 100 mg of alemtuzumab given over 5 days prior to SCT induces profound and prolonged lymphopenia and may be excessive. Lower doses of alemtuzumab provide similar GvHD prophylaxis and may allow better immune reconstitution. However, low-dose alemtuzumab might be insufficient to prevent graft failure and GvHD in CLL patients. Further clinical trials are needed to address alemtuzumab dosage in different settings. Efficacy has been proven in various malignant and non-malignant diseases with GvHD in 10% – 20% and TRM as low as 10% – 20%. The incidence of recurrence and the overall outcome depends on the underlying disease. The long-term results are encouraging in patients with acute leukemia and lymphoma where survival parallels that achieved with other transplant approaches, but with less chronic GvHD. In multiple myeloma and particularly in CLL, high recurrence rates and high rates of opportunistic infections lead to caution in the routine use of alemtuzumab. Regardless of the underlying disease, recurrence is somewhat higher than with other approaches but may often be managed successfully with a variety of salvage approaches, including DLI. Strict surveillance for CMV accompanied by prophylaxis or early preemptive therapy can prevent severe CMV-related complications. Adenovirus infections may be particularly common in children. Other opportunistic infections are in our experience not more common than with conventional GvHD prophylaxis. On the contrary, because of the low incidence of GvHD, the overall incidence of opportunistic infections may actually be reduced. Alemtuzumab is also being investigated for treatment of refractory GvHD.
9. Expert Opinion
Alemtuzumab is a safe and effective drug that can reduce both GvHD and TRM after transplant. The side-effects are an increased risk of disease recurrence and of opportunistic infections, particularly CMV reactivation. The optimal dose of alemtuzumab, which minimizes immunosuppression but maintains effective GvHD prophylaxis, is still under investigation but is probably in the range of 30 – 50mg. Successful use of alemtuzumab containing conditioning regimens requires strict management of CMV viremia and rapid intervention to salvage patients after relapse. Early use of DLI or relatively non-toxic drugs (e.g. TKIs in CML, rituximab or nucleoside analogs in NHL) can be used either for early intervention or, when appropriate, as consolidation. Opportunistic infections may be prevented with adequate prophylaxis or pre-emptive therapy. The ex vivo expansion of infection-specific T-cells to support immune reconstitution after T-cell-depleted SCT is also under active investigation.
Drug Summary Box.
Drug Name : Alemtuzumab |
Indication under discussion : T-cell depletion to prevent both acute and chronic graft-versus-host disease |
Mechanism of action : CD52+ cells lysis (T and B cells) through CDC, ADCC and direct apoptosis |
Route of administration : Intravenous or subcutaneous |
Pivotal trials : Kottaridis et al. 2000 [18]; Chakraverty et al. 2002 [17], Perez-Simon et al. 2002 [2] and van Besien et al. 2005 [59]; These large trials described significant efficacy of alemtuzumab as GvHD prophylaxis after RIC SCT in different haematological diseases. |
Glossary
- AA
aplastic anemia
- ADCC
antibody-dependent cellular cytotoxicity
- ALL
acute lymphoblastic leukemia
- AML
acute myeloid leukemia
- ATG
anti-thymocyte globulin
- BEAM
bis-chloroethylnitrosourea, Etoposide, Ara-C, Melphalan
- CDC
complement-dependent cytotoxicity
- CLL
chronic lymphocytic leukemia
- CML
chronic myelogenous leukemia
- CMV
cytomegalovirus
- DLI
donor lymphocytes infusion
- EBV
Epstein-Barr virus
- FCGR
IgG Fc eceptor
- GvHD
graft-versus-host disease
- GvT
graft-versus-tumor
- HL
Hodgkin’s lymphoma
- HLA
human leucocyte antigen
- IBMTR
International Bone Marrow Transplantation Registry
- MCL
Mantle cell lymphoma
- MDS
myelodysplastic syndrome
- MM
multiple myeloma
- NHL
non-Hodgkin’s lymphoma
- OS
overall survival
- PTLD
post-transplant lymphoproliferative disease
- RIC
reduced-intensity conditioning
- SCT
allogeneic stem cell transplantation
- SQ
subcutaneous
- TKI
tyrosine kinase inhibitor
- T-PLL
T-prolymphocytic leukemia
- TRM
treatment-related mortality
Footnotes
Declaration of interest
The authors declare no conflict of interest and have received no payment in preparation of this manuscript. K Besien is supported by an NIH grant, number K24CA116471.
Bibliography
Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers
- 1.Gooley TA, Chien JW, Pergam SA, et al. Reduced mortality after allogeneic hematopoietic-cell transplantation. N Engl J Med; 363:2091–2101. doi: 10.1056/NEJMoa1004383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2••.Perez-Simon JA, Kottaridis PD, Martino R, et al. Nonmyeloablative transplantation with or without alemtuzumab: comparison between 2 prospective studies in patients with lymphoproliferative disorders. Blood. 2002;100:3121–3127. doi: 10.1182/blood-2002-03-0701. Evidence is presented that incorporation of alemtuzumab to RIC reduces significantly GvHD compared to RIC without alemtuzumab. [DOI] [PubMed] [Google Scholar]
- 3.Wolff D, Gerbitz A, Ayuk F, et al. Consensus conference on clinical practice in chronic graft-versus-host disease (GVHD): first-line and topical treatment of chronic GVHD. Biol Blood Marrow Transplant; 16:1611–1628. doi: 10.1016/j.bbmt.2010.06.015. [DOI] [PubMed] [Google Scholar]
- 4.Waldmann H, Hale G. CAMPATH: from concept to clinic. Philos Trans R Soc Lond B Biol Sci. 2005;360:1707–1711. doi: 10.1098/rstb.2005.1702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ho VT, Soiffer RJ. The history and future of T-cell depletion as graft-versus-host disease prophylaxis for allogeneic hematopoietic stem cell transplantation. Blood. 2001;98:3192–3204. doi: 10.1182/blood.v98.12.3192. [DOI] [PubMed] [Google Scholar]
- 6.Papadopoulos EB, Carabasi MH, Castro-Malaspina H, et al. T-cell-depleted allogeneic bone marrow transplantation as postremission therapy for acute myelogenous leukemia: freedom from relapse in the absence of graft-versus-host disease. Blood. 1998;91:1083–1090. [PubMed] [Google Scholar]
- 7.Hale G, Zhang MJ, Bunjes D, et al. Improving the outcome of bone marrow transplantation by using CD52 monoclonal antibodies to prevent graft-versus-host disease and graft rejection. Blood. 1998;92:4581–4590. [PubMed] [Google Scholar]
- 8.Martinez C, Urbano-Ispizua A, Rozman C, et al. Immune reconstitution following allogeneic peripheral blood progenitor cell transplantation: comparison of recipients of positive CD34+ selected grafts with recipients of unmanipulated grafts. Exp Hematol. 1999;27:561–568. doi: 10.1016/s0301-472x(98)00029-0. [DOI] [PubMed] [Google Scholar]
- 9.Gribben JG, Hallek M. Rediscovering alemtuzumab: current and emerging therapeutic roles. Br J Haematol. 2009;144:818–831. doi: 10.1111/j.1365-2141.2008.07557.x. [DOI] [PubMed] [Google Scholar]
- 10.Keating MJ, Cazin B, Coutre S, et al. Campath-1H treatment of T-cell prolymphocytic leukemia in patients for whom at least one prior chemotherapy regimen has failed. J Clin Oncol. 2002;20:205–213. doi: 10.1200/JCO.2002.20.1.205. [DOI] [PubMed] [Google Scholar]
- 11.Risitano AM, Selleri C, Serio B, et al. Alemtuzumab is safe and effective as immunosuppressive treatment for aplastic anaemia and single-lineage marrow failure: a pilot study and a survey from the EBMT WPSAA. Br J Haematol; 148:791–796. doi: 10.1111/j.1365-2141.2009.08027.x. [DOI] [PubMed] [Google Scholar]
- 12.Coles AJ, Compston DA, Selmaj KW, et al. Alemtuzumab vs. interferon beta-1a in early multiple sclerosis. N Engl J Med. 2008;359:1786–1801. doi: 10.1056/NEJMoa0802670. [DOI] [PubMed] [Google Scholar]
- 13.Tan HP, Donaldson J, Basu A, et al. Two hundred living donor kidney transplantations under alemtuzumab induction and tacrolimus monotherapy: 3-year follow-up. Am J Transplant. 2009;9:355–366. doi: 10.1111/j.1600-6143.2008.02492.x. [DOI] [PubMed] [Google Scholar]
- 14.Pearl JP, Parris J, Hale DA, et al. Immunocompetent T-cells with a memory-like phenotype are the dominant cell type following antibody-mediated T-cell depletion. Am J Transplant. 2005;5:465–474. doi: 10.1111/j.1600-6143.2005.00759.x. [DOI] [PubMed] [Google Scholar]
- 15.Bloom DD, Chang Z, Fechner JH, et al. CD4+ CD25+ FOXP3+ regulatory T cells increase de novo in kidney transplant patients after immunodepletion with Campath-1H. Am J Transplant. 2008;8:793–802. doi: 10.1111/j.1600-6143.2007.02134.x. [DOI] [PubMed] [Google Scholar]
- 16.Trzonkowski P, Zilvetti M, Chapman S, et al. Homeostatic repopulation by CD28− CD8+ T cells in alemtuzumab-depleted kidney transplant recipients treated with reduced immunosuppression. Am J Transplant. 2008;8:338–47. doi: 10.1111/j.1600-6143.2007.02078.x. [DOI] [PubMed] [Google Scholar]
- 17••.Chakraverty R, Peggs K, Chopra R, et al. Limiting transplantation-related mortality following unrelated donor stem cell transplantation by using a nonmyeloablative conditioning regimen. Blood. 2002;99:1071–1078. doi: 10.1182/blood.v99.3.1071. This research examines the efficacy of alemtuzumab in unrelated-donors SCT. [DOI] [PubMed] [Google Scholar]
- 18••.Kottaridis PD, Milligan DW, Chopra R, et al. In vivo CAMPATH-1H prevents graft-versus-host disease following nonmyeloablative stem cell transplantation. Blood. 2000;96:2419–2425. The first, to our knowledge, large trial describing in vivo alemtuzumab as an effective method to reduce GvHD. [PubMed] [Google Scholar]
- 19.Gomez-Almaguer D, Ruiz-Arguelles GJ, del Carmen Tarin-Arzaga L, et al. Alemtuzumab for the treatment of steroid-refractory acute graft-versus-host disease. Biol Blood Marrow Transplant. 2008;14:10–15. doi: 10.1016/j.bbmt.2007.08.052. [DOI] [PubMed] [Google Scholar]
- 20.Xia MQ, Hale G, Lifely MR, et al. Structure of the CAMPATH-1 antigen, a glycosylphosphatidylinositol-anchored glycoprotein which is an exceptionally good target for complement lysis. Biochem J. 1993;293:633–640. doi: 10.1042/bj2930633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Gilleece MH, Dexter TM. Effect of Campath-1H antibody on human hematopoietic progenitors in vitro. Blood. 1993;82:807–812. [PubMed] [Google Scholar]
- 22.Ginaldi L, De Martinis M, Matutes E, et al. Levels of expression of CD52 in normal and leukemic B and T cells: correlation with in vivo therapeutic responses to Campath-1H. Leuk Res. 1998;22:185–191. doi: 10.1016/s0145-2126(97)00158-6. [DOI] [PubMed] [Google Scholar]
- 23.Rodig SJ, Abramson JS, Pinkus GS, et al. Heterogeneous CD52 expression among hematologic neoplasms: implications for the use of alemtuzumab (CAMPATH-1H) Clin Cancer Res. 2006;12:7174–7179. doi: 10.1158/1078-0432.CCR-06-1275. [DOI] [PubMed] [Google Scholar]
- 24.Albitar M, Do KA, Johnson MM, et al. Free circulating soluble CD52 as a tumor marker in chronic lymphocytic leukemia and its implication in therapy with anti-CD52 antibodies. Cancer. 2004;101:999–1008. doi: 10.1002/cncr.20477. [DOI] [PubMed] [Google Scholar]
- 25.Matthews K, Lim Z, Afzali B, et al. Imbalance of effector and regulatory CD4 T cells is associated with graft-versus-host disease after hematopoietic stem cell transplantation using a reduced intensity conditioning regimen and alemtuzumab. Haematologica. 2009;94:956–966. doi: 10.3324/haematol.2008.003103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hu Y, Turner MJ, Shields J, et al. Investigation of the mechanism of action of alemtuzumab in a human CD52 transgenic mouse model. Immunology. 2009;128:260–270. doi: 10.1111/j.1365-2567.2009.03115.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Penack O, Fischer L, Stroux A, et al. Serotherapy with thymoglobulin and alemtuzumab differentially influences frequency and function of natural killer cells after allogeneic stem cell transplantation. Bone Marrow Transplant. 2008;41:377–383. doi: 10.1038/sj.bmt.1705911. [DOI] [PubMed] [Google Scholar]
- 28.Ratzinger G, Reagan JL, Heller G, et al. Differential CD52 expression by distinct myeloid dendritic cell subsets: implications for alemtuzumab activity at the level of antigen presentation in allogeneic graft-host interactions in transplantation. Blood. 2003;101:1422–1429. doi: 10.1182/blood-2002-04-1093. [DOI] [PubMed] [Google Scholar]
- 29.Ferrajoli A, O’Brien SM, Cortes JE, et al. Phase II study of alemtuzumab in chronic lymphoproliferative disorders. Cancer. 2003;98:773–778. doi: 10.1002/cncr.11551. [DOI] [PubMed] [Google Scholar]
- 30.Golay J, Manganini M, Rambaldi A, Introna M. Effect of alemtuzumab on neoplastic B cells. Haematologica. 2004;89:1476–1483. [PubMed] [Google Scholar]
- 31.Nuckel H, Frey UH, Roth A, et al. Alemtuzumab induces enhanced apoptosis in vitro in B-cells from patients with chronic lymphocytic leukemia by antibody-dependent cellular cytotoxicity. Eur J Pharmacol. 2005;514:217–224. doi: 10.1016/j.ejphar.2005.03.024. [DOI] [PubMed] [Google Scholar]
- 32.Stanglmaier M, Reis S, Hallek M. Rituximab and alemtuzumab induce a nonclassic, caspase-independent apoptotic pathway in B-lymphoid cell lines and in chronic lymphocytic leukemia cells. Ann Hematol. 2004;83:634–645. doi: 10.1007/s00277-004-0917-0. [DOI] [PubMed] [Google Scholar]
- 33.Zhang Z, Zhang M, Goldman CK, et al. Effective therapy for a murine model of adult T-cell leukemia with the humanized anti-CD52 monoclonal antibody, Campath-1H. Cancer Res. 2003;63:6453–6457. [PubMed] [Google Scholar]
- 34.Lin TS, Flinn IW, Modali R, et al. FCGR3A and FCGR2A polymorphisms may not correlate with response to alemtuzumab in chronic lymphocytic leukemia. Blood. 2005;105:289–291. doi: 10.1182/blood-2004-02-0651. [DOI] [PubMed] [Google Scholar]
- 35.Rebello P, Cwynarski K, Varughese M, et al. Pharmacokinetics of CAMPATH-1H in BMT patients. Cytotherapy. 2001;3:261–267. doi: 10.1080/146532401317070899. [DOI] [PubMed] [Google Scholar]
- 36•.Morris EC, Rebello P, Thomson KJ, et al. Pharmacokinetics of alemtuzumab used for in vivo and in vitro T-cell depletion in allogeneic transplantations: relevance for early adoptive immunotherapy and infectious complications. Blood. 2003;102:404–406. doi: 10.1182/blood-2002-09-2687. A comprehensive study on pharmacokinetics of alemtuzumab. [DOI] [PubMed] [Google Scholar]
- 37.Tholouli E, Liakopoulou E, Greenfield HM, et al. Outcomes following 50 mg versus 100 mg alemtuzumab in reduced-intensity conditioning stem cell transplants for acute myeloid leukaemia and poor risk myelodysplasia. Br J Haematol. 2008;142:318–320. doi: 10.1111/j.1365-2141.2008.07184.x. [DOI] [PubMed] [Google Scholar]
- 38.Brett S, Baxter G, Cooper H, et al. Repopulation of blood lymphocyte sub-populations in rheumatoid arthritis patients treated with the depleting humanized monoclonal antibody, CAMPATH-1H. Immunology. 1996;88:13–19. doi: 10.1046/j.1365-2567.1996.d01-650.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Bertz H, Spyridonidis A, Wasch R, et al. A novel GVHD-prophylaxis with low-dose alemtuzumab in allogeneic sibling or unrelated donor hematopoetic cell transplantation: the feasibility of deescalation. Biol Blood Marrow Transplant. 2009;15:1563–1570. doi: 10.1016/j.bbmt.2009.08.002. [DOI] [PubMed] [Google Scholar]
- 40.Spyridonidis A, Liga M, Triantafyllou E, et al. Pharmacokinetics and clinical activity of very low-dose alemtuzumab in transplantation for acute leukemia. Bone Marrow Transplant. doi: 10.1038/bmt.2010.308. published online 20 December 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chakraverty R, Orti G, Roughton M, et al. Impact of in vivo alemtuzumab dose before reduced intensity conditioning and HLA-identical sibling stem cell transplantation: pharmacokinetics, GVHD, and immune reconstitution. Blood; 116:3080–3088. doi: 10.1182/blood-2010-05-286856. [DOI] [PubMed] [Google Scholar]
- 42.Bokhari S, Das-Gupta E, Russell N, Byrne J. Post-transplant lymphoproliferative disease following reduced intensity conditioning transplants incorporating alemtuzumab. Bone Marrow Transplant. 2008;42:281–282. doi: 10.1038/bmt.2008.149. [DOI] [PubMed] [Google Scholar]
- 43.Hale G, Rebello P, Brettman LR, et al. Blood concentrations of alemtuzumab and antiglobulin responses in patients with chronic lymphocytic leukemia following intravenous or subcutaneous routes of administration. Blood. 2004;104:948–955. doi: 10.1182/blood-2004-02-0593. [DOI] [PubMed] [Google Scholar]
- 44.Oshima K, Kanda Y, Nakahara F, et al. Pharmacokinetics of alemtuzumab after haploidentical HLA-mismatched hematopoietic stem cell transplantation using in vivo alemtuzumab with or without CD52-positive malignancies. Am J Hematol. 2006;81:875–879. doi: 10.1002/ajh.20694. [DOI] [PubMed] [Google Scholar]
- 45.Mould DR, Baumann A, Kuhlmann J, et al. Population pharmacokinetics-pharmacodynamics of alemtuzumab (Campath) in patients with chronic lymphocytic leukaemia and its link to treatment response. Br J Clin Pharmacol. 2007;64:278–291. doi: 10.1111/j.1365-2125.2007.02914.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46•.Delgado J, Thomson K, Russell N, et al. Results of alemtuzumab-based reduced-intensity allogeneic transplantation for chronic lymphocytic leukemia: a British Society of Blood and Marrow Transplantation Study. Blood. 2006;107:1724–1730. doi: 10.1182/blood-2005-08-3372. This research examined the efficacy of alemtuzumab-based SCT in CLL. [DOI] [PubMed] [Google Scholar]
- 47.Juliusson G, Theorin N, Karlsson K, et al. Subcutaneous alemtuzumab vs ATG in adjusted conditioning for allogeneic transplantation: influence of Campath dose on lymphoid recovery, mixed chimerism and survival. Bone Marrow Transplant. 2006;37:503–510. doi: 10.1038/sj.bmt.1705263. [DOI] [PubMed] [Google Scholar]
- 48.Novitzky N, Thomas V, Hale G, Waldmann H. Ex vivo depletion of T cells from bone marrow grafts with CAMPATH-1 in acute leukemia: graft-versus-host disease and graft-versus-leukemia effect. Transplantation. 1999;67:620–626. doi: 10.1097/00007890-199902270-00022. [DOI] [PubMed] [Google Scholar]
- 49.Jacobs P, Wood L. Immunohematopoietic stem cell transplantation: introduction and 35 years of development in South Africa–the historical and scientific perspective. Bone Marrow Transplant. 2008;42 (Suppl 1):S125–S132. doi: 10.1038/bmt.2008.140. [DOI] [PubMed] [Google Scholar]
- 50.Barge RM, Starrenburg CW, Falkenburg JH, et al. Long-term follow-up of myeloablative allogeneic stem cell transplantation using Campath “in the bag” as T-cell depletion: the Leiden experience. Bone Marrow Transplant. 2006;37:1129–1134. doi: 10.1038/sj.bmt.1705385. [DOI] [PubMed] [Google Scholar]
- 51.von dem Borne PA, Beaumont F, Starrenburg CW, et al. Outcomes after myeloablative unrelated donor stem cell transplantation using both in vitro and in vivo T-cell depletion with alemtuzumab. Haematologica. 2006;91:1559–1562. [PubMed] [Google Scholar]
- 52.Novitzky N, Thomas V, du Toit C, McDonald A. Reduced-intensity conditioning for severe aplasia using fludarabine and CY followed by infusion of ex vivo T-cell-depleted grafts leads to excellent engraftment and absence of GVHD. Bone Marrow Transplant. 2009;43:779–785. doi: 10.1038/bmt.2008.390. [DOI] [PubMed] [Google Scholar]
- 53.Chakrabarti S, MacDonald D, Hale G, et al. T-cell depletion with Campath-1H “in the bag” for matched related allogeneic peripheral blood stem cell transplantation is associated with reduced graft-versus-host disease, rapid immune constitution and improved survival. Br J Haematol. 2003;121:109–118. doi: 10.1046/j.1365-2141.2003.04228.x. [DOI] [PubMed] [Google Scholar]
- 54.Mead AJ, Thomson KJ, Morris EC, et al. HLA-mismatched unrelated donors are a viable alternate graft source for allogeneic transplantation following alemtuzumab-based reduced-intensity conditioning. Blood; 115:5147–5153. doi: 10.1182/blood-2010-01-265413. [DOI] [PubMed] [Google Scholar]
- 55.Rizzieri DA, Koh LP, Long GD, et al. Partially matched, nonmyeloablative allogeneic transplantation: clinical outcomes and immune reconstitution. J Clin Oncol. 2007;25:690–697. doi: 10.1200/JCO.2006.07.0953. [DOI] [PubMed] [Google Scholar]
- 56.Rizzieri DA, Dev P, Long GD, et al. Response and toxicity of donor lymphocyte infusions following T-cell depleted non-myeloablative allogeneic hematopoietic SCT from 3–6/6 HLA matched donors. Bone Marrow Transplant. 2009;43:327–333. doi: 10.1038/bmt.2008.321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Ho AY, Pagliuca A, Kenyon M, et al. Reduced-intensity allogeneic hematopoietic stem cell transplantation for myelodysplastic syndrome and acute myeloid leukemia with multilineage dysplasia using fludarabine, busulphan, and alemtuzumab (FBC) conditioning. Blood. 2004;104:1616–1623. doi: 10.1182/blood-2003-12-4207. [DOI] [PubMed] [Google Scholar]
- 58•.Tauro S, Craddock C, Peggs K, et al. Allogeneic stem-cell transplantation using a reduced-intensity conditioning regimen has the capacity to produce durable remissions and long-term disease-free survival in patients with high-risk acute myeloid leukemia and myelodysplasia. J Clin Oncol. 2005;23:9387–9393. doi: 10.1200/JCO.2005.02.0057. This study examined the efficacy of alemtuzumab-based SCT in AML and MDS. [DOI] [PubMed] [Google Scholar]
- 59••.van Besien K, Artz A, Smith S, et al. Fludarabine, melphalan, and alemtuzumab conditioning in adults with standard-risk advanced acute myeloid leukemia and myelodysplastic syndrome. J Clin Oncol. 2005;23:5728–5738. doi: 10.1200/JCO.2005.15.602. The first large series, to our knowledge, describing efficacy of alemtuzumab-based SCT in AML and MDS. [DOI] [PubMed] [Google Scholar]
- 60•.van Besien K, Kunavakkam R, Rondon G, et al. Fludarabine-melphalan conditioning for AML and MDS: alemtuzumab reduces acute and chronic GVHD without affecting long-term outcomes. Biol Blood Marrow Transplant. 2009;15:610–617. doi: 10.1016/j.bbmt.2009.01.021. A comparative study of alemtuzumab-based and no alemtuzumabb-based SCT in AML and MDS patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Lim ZY, Ho AY, Ingram W, et al. Outcomes of alemtuzumab-based reduced intensity conditioning stem cell transplantation using unrelated donors for myelodysplastic syndromes. Br J Haematol. 2006;135:201–209. doi: 10.1111/j.1365-2141.2006.06272.x. [DOI] [PubMed] [Google Scholar]
- 62.Lim ZY, Pearce L, Ho AY, et al. Delayed attainment of full donor chimaerism following alemtuzumab-based reduced-intensity conditioning haematopoeitic stem cell transplantation for acute myeloid leukaemia and myelodysplastic syndromes is associated with improved outcomes. Br J Haematol. 2007;138:517–526. doi: 10.1111/j.1365-2141.2007.06676.x. [DOI] [PubMed] [Google Scholar]
- 63.Malladi RK, Peniket AJ, Littlewood TJ, et al. Alemtuzumab markedly reduces chronic GVHD without affecting overall survival in reduced-intensity conditioning sibling allo-SCT for adults with AML. Bone Marrow Transplant. 2009;43:709–715. doi: 10.1038/bmt.2008.375. [DOI] [PubMed] [Google Scholar]
- 64.Craddock C, Nagra S, Peniket A, et al. Factors predicting long-term survival after T-cell depleted reduced intensity allogeneic stem cell transplantation for acute myeloid leukemia. Haematologica; 95:989–995. doi: 10.3324/haematol.2009.013920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Patel B, Kirkland KE, Szydlo R, et al. Favorable outcomes with alemtuzumab-conditioned unrelated donor stem cell transplantation in adults with high-risk Philadelphia chromosome-negative acute lymphoblastic leukemia in first complete remission. Haematologica. 2009;94:1399–1406. doi: 10.3324/haematol.2009.008649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Freytes CO, Loberiza FR, Rizzo JD, et al. Myeloablative allogeneic hematopoietic stem cell transplantation in patients who experience relapse after autologous stem cell transplantation for lymphoma: a report of the International Bone Marrow Transplant Registry. Blood. 2004;104:3797–3803. doi: 10.1182/blood-2004-01-0231. [DOI] [PubMed] [Google Scholar]
- 67•.Morris E, Thomson K, Craddock C, et al. Outcomes after alemtuzumab-containing reduced-intensity allogeneic transplantation regimen for relapsed and refractory non-Hodgkin lymphoma. Blood. 2004;104:3865–3871. doi: 10.1182/blood-2004-03-1105. This paper describes the efficacy of alemtuzumab-based SCT in a cohort of NHL patients. [DOI] [PubMed] [Google Scholar]
- 68.Thomson KJ, Morris EC, Bloor A, et al. Favorable long-term survival after reduced-intensity allogeneic transplantation for multiple-relapse aggressive non-Hodgkin’s lymphoma. J Clin Oncol. 2009;27:426–432. doi: 10.1200/JCO.2008.17.3328. [DOI] [PubMed] [Google Scholar]
- 69.Cook G, Smith GM, Kirkland K, et al. Outcome following Reduced-Intensity Allogeneic Stem Cell Transplantation (RIC AlloSCT) for relapsed and refractory mantle cell lymphoma (MCL): a study of the British society for blood and marrow transplantation. Biol Blood Marrow Transplant; 16:1419–1427. doi: 10.1016/j.bbmt.2010.04.006. [DOI] [PubMed] [Google Scholar]
- 70.Thomson KJ, Morris EC, Milligan D, et al. T-cell-depleted reduced-intensity transplantation followed by donor leukocyte infusions to promote graft-versus-lymphoma activity results in excellent long-term survival in patients with multiply relapsed follicular lymphoma. J Clin Oncol; 28:3695–3700. doi: 10.1200/JCO.2009.26.9100. [DOI] [PubMed] [Google Scholar]
- 71.Morris E, Mackinnon S. Outcome following alemtuzumab (CAMPATH-1H)-containing reduced intensity allogeneic transplant regimen for relapsed and refractory non-Hodgkin’s lymphoma (NHL) Transfus Apher Sci. 2005;32:73–83. doi: 10.1016/j.transci.2004.10.008. [DOI] [PubMed] [Google Scholar]
- 72.Robinson SP, Goldstone AH, Mackinnon S, et al. Chemoresistant or aggressive lymphoma predicts for a poor outcome following reduced-intensity allogeneic progenitor cell transplantation: an analysis from the Lymphoma Working Party of the European Group for Blood and Bone Marrow Transplantation. Blood. 2002;100:4310–4316. doi: 10.1182/blood-2001-11-0107. [DOI] [PubMed] [Google Scholar]
- 73.Peggs KS, Sureda A, Qian W, et al. Reduced-intensity conditioning for allogeneic haematopoietic stem cell transplantation in relapsed and refractory Hodgkin lymphoma: impact of alemtuzumab and donor lymphocyte infusions on long-term outcomes. Br J Haematol. 2007;139:70–80. doi: 10.1111/j.1365-2141.2007.06759.x. [DOI] [PubMed] [Google Scholar]
- 74.Thomson KJ, Peggs KS, Smith P, et al. Superiority of reduced-intensity allogeneic transplantation over conventional treatment for relapse of Hodgkin’s lymphoma following autologous stem cell transplantation. Bone Marrow Transplant. 2008;41:765–770. doi: 10.1038/sj.bmt.1705977. [DOI] [PubMed] [Google Scholar]
- 75.Peggs KS, Kayani I, Edwards N, et al. Donor lymphocyte infusions modulate relapse risk in mixed chimeras and induce durable salvage in relapsed patients after T-cell-depleted allogeneic transplantation for Hodgkin’s lymphoma. J Clin Oncol; 29:971–978. doi: 10.1200/JCO.2010.32.1711. [DOI] [PubMed] [Google Scholar]
- 76.Hari P, Carreras J, Zhang MJ, Gale RP, et al. Allogeneic transplants in follicular lymphoma: higher risk of disease progression after reduced-intensity compared to myeloablative conditioning. Biol Blood Marrow Transplant. 2008;14:236–245. doi: 10.1016/j.bbmt.2007.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kenkre VP, Horowitz S, Artz AS, et al. T-cell-depleted allogeneic transplant without donor leukocyte infusions results in excellent long-term survival in patients with multiply relapsed lymphoma. Predictors for survival after transplant relapse. Leuk Lymphoma. 2011;52:214–222. doi: 10.3109/10428194.2010.538777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Faulkner RD, Craddock C, Byrne JL, et al. BEAM-alemtuzumab reduced-intensity allogeneic stem cell transplantation for lymphoproliferative diseases: GVHD, toxicity, and survival in 65 patients. Blood. 2004;103:428–434. doi: 10.1182/blood-2003-05-1406. [DOI] [PubMed] [Google Scholar]
- 79.Ingram W, Devereux S, Das-Gupta EP, et al. Outcome of BEAM-autologous and BEAM-alemtuzumab allogeneic transplantation in relapsed advanced stage follicular lymphoma. Br J Haematol. 2008;141:235–243. doi: 10.1111/j.1365-2141.2008.07067.x. [DOI] [PubMed] [Google Scholar]
- 80.Dreger P, Dohner H, Ritgen M, et al. Allogeneic stem cell transplantation provides durable disease control in poor-risk chronic lymphocytic leukemia: long-term clinical and MRD results of the German CLL Study Group CLL3X trial. Blood. 2010;116:2438–2447. doi: 10.1182/blood-2010-03-275420. [DOI] [PubMed] [Google Scholar]
- 81.Crawley C, Szydlo R, Lalancette M, et al. Outcomes of reduced-intensity transplantation for chronic myeloid leukemia: an analysis of prognostic factors from the Chronic Leukemia Working Party of the EBMT. Blood. 2005;106:2969–2976. doi: 10.1182/blood-2004-09-3544. [DOI] [PubMed] [Google Scholar]
- 82.Poire X, Artz A, Larson RA, et al. Allogeneic stem cell transplantation with alemtuzumab-based conditioning for patients with advanced chronic myelogenous leukemia. Leuk Lymphoma. 2009;50:85–91. doi: 10.1080/10428190802626624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Crawley C, Lalancette M, Szydlo R, et al. Outcomes for reduced-intensity allogeneic transplantation for multiple myeloma: an analysis of prognostic factors from the Chronic Leukaemia Working Party of the EBMT. Blood. 2005;105:4532–4539. doi: 10.1182/blood-2004-06-2387. [DOI] [PubMed] [Google Scholar]
- 84.Siegal D, Xu W, Sutherland R, et al. Graft-versus-host disease following marrow transplantation for aplastic anemia: different impact of two GVHD prevention strategies. Bone Marrow Transplant. 2008;42:51–56. doi: 10.1038/bmt.2008.88. [DOI] [PubMed] [Google Scholar]
- 85.Hsieh MM, Kang EM, Fitzhugh CD, et al. Allogeneic hematopoietic stem-cell transplantation for sickle cell disease. N Engl J Med. 2009;361:2309–2317. doi: 10.1056/NEJMoa0904971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Chewning JH, Castro-Malaspina H, Jakubowski A, et al. Fludarabine-based conditioning secures engraftment of second hematopoietic stem cell allografts (HSCT) in the treatment of initial graft failure. Biol Blood Marrow Transplant. 2007;13:1313–1323. doi: 10.1016/j.bbmt.2007.07.006. [DOI] [PubMed] [Google Scholar]
- 87.Bolanos-Meade J, Luznik L, Muth M, et al. Salvage transplantation for allograft failure using fludarabine and alemtuzumab as conditioning regimen. Bone Marrow Transplant. 2009;43:477–480. doi: 10.1038/bmt.2008.353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Martinez C, Solano C, Ferra C, et al. Alemtuzumab as treatment of steroid-refractory acute graft-versus-host disease: results of a Phase II study. Biol Blood Marrow Transplant. 2009;15:639–642. doi: 10.1016/j.bbmt.2009.01.014. [DOI] [PubMed] [Google Scholar]
- 89.Schnitzler M, Hasskarl J, Egger M, et al. Successful treatment of severe acute intestinal graft-versus-host resistant to systemic and topical steroids with alemtuzumab. Biol Blood Marrow Transplant. 2009;15:910–918. doi: 10.1016/j.bbmt.2009.04.002. [DOI] [PubMed] [Google Scholar]
- 90.Ruiz-Arguelles GJ, Gil-Beristain J, Magana M, Ruiz-Delgado GJ. Alemtuzumab-induced resolution of refractory cutaneous chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2008;14:7–9. doi: 10.1016/j.bbmt.2007.09.013. [DOI] [PubMed] [Google Scholar]
- 91.Mackinnon S, Papadopoulos EB, Carabasi MH, et al. Adoptive immunotherapy evaluating escalating doses of donor leukocytes for relapse of chronic myeloid leukemia after bone marrow transplantation: separation of graft-versus-leukemia responses from graft-versus-host disease. Blood. 1995;86:1261–1268. [PubMed] [Google Scholar]
- 92•.Peggs KS, Thomson K, Hart DP, et al. Dose-escalated donor lymphocyte infusions following reduced intensity transplantation: toxicity, chimerism, and disease responses. Blood. 2004;103:1548–1556. doi: 10.1182/blood-2003-05-1513. This paper describes the successful application of DLI after alemtuzumab-based SCT. [DOI] [PubMed] [Google Scholar]
- 93.Pollyea DA, Artz AS, Stock W, et al. Outcomes of patients with AML and MDS who relapse or progress after reduced intensity allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. 2007;40:1027–1032. doi: 10.1038/sj.bmt.1705852. [DOI] [PubMed] [Google Scholar]
- 94.van Besien K, Dew A, Lin S, et al. Patterns and kinetics of T-cell chimerism after allo transplant with alemtuzumab-based conditioning: mixed chimerism protects from GVHD, but does not portend disease recurrence. Leuk Lymphoma. 2009;50:1809–1817. doi: 10.3109/10428190903200790. [DOI] [PubMed] [Google Scholar]
- 95.Bonnefoy-Berard N, Vincent C, Revillard JP. Antibodies against functional leukocyte surface molecules in polyclonal antilymphocyte and antithymocyte globulins. Transplantation. 1991;51:669–673. doi: 10.1097/00007890-199103000-00024. [DOI] [PubMed] [Google Scholar]
- 96.Kroger N, Shaw B, Iacobelli S, et al. Comparison between antithymocyte globulin and alemtuzumab and the possible impact of KIR-ligand mismatch after dose-reduced conditioning and unrelated stem cell transplantation in patients with multiple myeloma. Br J Haematol. 2005;129:631–643. doi: 10.1111/j.1365-2141.2005.05513.x. [DOI] [PubMed] [Google Scholar]
- 97.Norlin AC, Remberger M. A comparison of Campath and Thymoglobulin as part of the conditioning before allogeneic hematopoietic stem cell transplantation. Eur J Haematol; 86:57–66. doi: 10.1111/j.1600-0609.2010.01537.x. [DOI] [PubMed] [Google Scholar]
- 98.Soiffer R, LeRademacher J, Ho V, et al. Impact of in vivo T-cell depletion on outcome of reduced intensity conditioning (RIC) hematopoetic cell transplantation (HCT) for hematological malignancies. Blood (ASH Annual Meeting Abstracts) 2010;116 abstract 2305. [Google Scholar]
- 99.Landgren O, Gilbert ES, Rizzo JD, et al. Risk factors for lymphoproliferative disorders after allogeneic hematopoietic cell transplantation. Blood. 2009;113:4992–5001. doi: 10.1182/blood-2008-09-178046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Lowdell MW, Craston R, Ray N, et al. The effect of T cell depletion with Campath-1M on immune reconstitution after chemotherapy and allogeneic bone marrow transplant as treatment for leukaemia. Bone Marrow Transplant. 1998;21:679–686. doi: 10.1038/sj.bmt.1701153. [DOI] [PubMed] [Google Scholar]
- 101.Schmidt-Hieber M, Schwarck S, Stroux A, et al. Immune reconstitution and cytomegalovirus infection after allogeneic stem cell transplantation: the important impact of in vivo T cell depletion. Int J Hematol; 91:877–885. doi: 10.1007/s12185-010-0597-6. [DOI] [PubMed] [Google Scholar]
- 102.Chakrabarti S, Mackinnon S, Chopra R, et al. High incidence of cytomegalovirus infection after nonmyeloablative stem cell transplantation: potential role of Campath-1H in delaying immune reconstitution. Blood. 2002;99:4357–4363. doi: 10.1182/blood.v99.12.4357. [DOI] [PubMed] [Google Scholar]
- 103.Lamba R, Carrum G, Myers GD, et al. Cytomegalovirus (CMV) infections and CMV-specific cellular immune reconstitution following reduced intensity conditioning allogeneic stem cell transplantation with Alemtuzumab. Bone Marrow Transplant. 2005;36:797–802. doi: 10.1038/sj.bmt.1705121. [DOI] [PubMed] [Google Scholar]
- 104.Lim ZY, Cook G, Johnson PR, et al. Results of a phase I/II British Society of Bone Marrow Transplantation study on PCR-based pre-emptive therapy with valganciclovir or ganciclovir for active CMV infection following alemtuzumab-based reduced intensity allogeneic stem cell transplantation. Leuk Res. 2009;33:244–249. doi: 10.1016/j.leukres.2008.07.016. [DOI] [PubMed] [Google Scholar]
- 105•.Kline J, Pollyea DA, Stock W, et al. Pre-transplant ganciclovir and post transplant high-dose valacyclovir reduce CMV infections after alemtuzumab-based conditioning. Bone Marrow Transplant. 2006;37:307–310. doi: 10.1038/sj.bmt.1705249. This paper describes effective CMV prophylaxis in alemtuzumab-based SCT. [DOI] [PubMed] [Google Scholar]
- 106.O’Donnell PH, Swanson K, Josephson MA, et al. BK virus infection is associated with hematuria and renal impairment in recipients of allogeneic hematopoetic stem cell transplants. Biol Blood Marrow Transplant. 2009;15:1038–1048. e1. doi: 10.1016/j.bbmt.2009.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Carpenter B, Haque T, Dimopoulou M, et al. Incidence and dynamics of Epstein-Barr virus reactivation after alemtuzumab-based conditioning for allogeneic hematopoietic stem-cell transplantation. Transplantation; 90:564–570. doi: 10.1097/TP.0b013e3181e7a3bf. [DOI] [PubMed] [Google Scholar]
- 108.Chakrabarti S, Milligan DW, Pillay D, et al. Reconstitution of the Epstein-Barr virus-specific cytotoxic T-lymphocyte response following T-cell-depleted myeloablative and nonmyeloablative allogeneic stem cell transplantation. Blood. 2003;102:839–842. doi: 10.1182/blood.V102.3.839. [DOI] [PubMed] [Google Scholar]
- 109.Chakrabarti S, Mautner V, Osman H, et al. Adenovirus infections following allogeneic stem cell transplantation: incidence and outcome in relation to graft manipulation, immunosuppression, and immune recovery. Blood. 2002;100:1619–1627. doi: 10.1182/blood-2002-02-0377. [DOI] [PubMed] [Google Scholar]