Although sickle cell disease (SCD) has a variable clinical course, many patients develop end-organ complications that are associated with significant morbidity and early mortality. Myeloablative allogeneic HSCT (allo-HSCT) is curative but has been historically performed only in children younger than 16 years of age. Modest modifications in the conditioning regimen and supportive care have improved outcome such that the majority of children with a suitable HLA-matched sibling donor can expect a cure from this approach. However, adult patients have been excluded from myeloablative allo-HSCT because of anticipated excess toxicity resulting from accumulated disease burden. Efforts to use nonmyeloablative transplantation strategies in adults logically followed but were initially met with largely disappointing results. Recent results, however, indicate that nonmyeloablative allo-HSCT in adult patients with SCD allows for stable mixed hematopoietic chimerism with associated full-donor erythroid engraftment and normalization of blood counts, and persistence in some without continued immunosuppression suggests immunologic tolerance. The attainment of tolerance should allow extension of these potentially curative approaches to alternative donor sources. Efforts to build on these experiences should increase the use of allo-HSCT in patients with SCD while minimizing morbidity and mortality.

Sickle cell disease (SCD) results from a single nucleotide mutation, which changes the glutamate for valine in the 6th position of the β-globin protein. This change results in a propensity for the hemoglobin protein to polymerize when deoxygenated and causes the characteristic sickle-shaped red cells. The disorder is characterized by anemia, ongoing hemolysis, along with acute and chronic complications affecting multiple organs. Although transfusions can prevent further neurologic events in patients at risk, iron overload is common, resulting in significant end-organ toxicity. The implementation of newborn screening, penicillin prophylaxis, and prompt evaluation and treatment for fever have improved the outlook for children with SCD. Specific treatment has remained limited and includes erythrocyte transfusions and more recently hydroxyurea.1-3  Hydroxyurea results in a significant reduction in the number of painful crises per year and a decreased frequency of acute chest syndrome1 ; it has become the treatment of choice for many patients with SCD. Unfortunately, hydroxyurea is not curative and does not appear to reverse established end-organ damage. The medical costs of this disease are also enormous, with estimates of $40 000 per patient per year (year 2000 figures) for chronic transfusion therapy and chelation alone, but they do not include the impact on quality of life of those with the disease.4  It is astounding that a single nucleotide substitution can lead to a multiorgan disease that dramatically reduces the quality of life and shortens the lifespan of those affected. Unfortunately, these existing treatments only ameliorate the manifestation of SCD, leaving an increasing number of adults with accumulating end-organ damage. Although longstanding efforts to deliver the correct β-globin gene through gene therapy approaches are progressing well, currently allo-HSCT remains the only immediate cure. Because patients with SCD are living longer and with chronic organ insufficiencies from sickle-related organ damage or from transfusional iron overload, improving HSCT for children and adults with SCD has emerged as an area of research interest with immediate potential clinical benefit.

Although the curative potential of HSCT has been well established in several nonmalignant disorders, there has always been difficulty in determining which patients with SCD warrant the potential risks of this technique. Early efforts to develop criteria to distinguish the more severe patients proved difficult in a disorder with chronic, yet mostly manageable complications, punctuated by rarer, more severe, even life-threatening complications. There remains a temptation to intervene early when patient status would improve the odds against complications from the conditioning regimen. Alternatively, intervening later when chronic, irreversible disease complications clearly establish the severity of the patient's phenotype limits its application because the conditioning regimen is less likely to be well tolerated. Transcranial Doppler (TCD) examination has proven effective in predicting the risk of stroke and has been used to predict the severity of disease. Although abnormal TCD examinations predict subsequent development of stroke, and prompt red cell transfusion therapy dramatically reduces that risk,5  data are beginning to show that TCDs can remain abnormal in a small proportion of patients.6  Furthermore, among children with a first stroke on chronic transfusion therapy, a second event of overt stroke, silent stroke, or CNS vasculopathy can occur.7  These red cell transfusions, even when extensively matched, can lead to red cell8,9  or HLA10  allo-antibodies and iron overload in a significant proportion of patients. Unfortunately, there are currently no reliable predictors as to who will have persistently abnormal TCD or who will develop a second stroke or red cell alloimmunization.

In the early 1990s, hydroxyurea was shown to decrease the frequency and severity of vaso-occlusive crises and acute chest syndromes. HSCT were thus performed in severely affected pediatric patients who would not be expected to benefit from hydroxyurea (stroke, red cell alloimmunizations, or avascular necrosis affecting multiple joints) or in those who did not benefit from hydroxyurea (recurrent vaso-occlusive crises or acute chest syndrome on hydroxyurea).11,12  The authors of recent studies have regrettably shown that despite more widespread use of hydroxyurea, the average patient with SCD only lives ∼ 40 years.13,14  Unlike in pediatric patients, in whom the prediction of who will have severe disease is not certain, the trajectory of adult patients having a shortened lifespan and a large proportion of adult patients developing irreversible organ damage are certain. This dramatically reduced lifespan or irreversible end-organ damage (elevated tricuspid regurgitant jet velocity,15  renal insufficiency,16,17  or hepatopathy,18,19  all with associated increased mortality) should provide physicians motivation to offer HSCT to those with matched siblings at younger ages and to develop nonmyeloablative regimens that are appropriate for adults with a high disease burden. The responsibility is on the SCD transplant community to develop HSCT regimens that are minimally toxic and that have a realistic chance for success to extend their lives. Table 1 summarizes the historical indications for HSCT, hydroxyurea, and the criteria that we have developed, after incorporating recent clinical trials and their impact on mortality.

Allo-HSCT remains the only curative strategy for patients with SCD. The first successful HSCT in a patient with SCD was reported in 1984 in a pediatric patient with coexisting acute myeloid leukemia.20  Traditional myeloablative conditioning regimens that use BM as the HSC source and myeloablative doses of busulfan in combination with highly immunosuppressive doses of cyclophosphamide were used with transplants first reported in Europe,21,22  and then in the United States.12  Antithymocyte globulin was later added more consistently to decrease the risk of graft rejection in this population of patients who have been previously transfused and are frequently alloimmunized.11  In one early trial, neurologic complications, including seizures and fatal intracranial hemorrhage, occurred in 7 of 21 patients with SCD who underwent myeloablative conditioning.23  The incidence of neurologic complications subsequently decreased by maintaining a platelet threshold of > 50 000/μL and a hemoglobin level of 9-11 g/dL, adding phenytoin prophylaxis, and preventing significant hypertension and hypomagnesemia.12 

In one series, patients were divided into 2 groups by criteria reflective of their access to care. The first group consisted of patients who underwent transplantation according to traditional severity criteria and were permanent residents of a European country. The second group of patients underwent transplantation because they would subsequently be returning to their country of origin in Africa, where access to chronic SCD care may be limited. Although patients in the first group were older (median age 8.6 vs 2 years; P = .0016), the first group had a greater rate of graft rejection (25% vs 7%, P < .001), and all of the patients who developed severe GVHD were in group 1.21  These data suggest that transplantation for patients with SCD may be performed more safely in pediatric patients who have not experienced end-organ damage or other markers of disease severity.

The largest group to date was reported in 2007.11  Overall survival and event-free survival were 93% and 86%, respectively. A major finding of this study was that after the addition of antithymocyte globulin in 1992 to the conditioning regimen, the rejection rate decreased from 22.6% to 3%, with an event-free survival rate of 95% among patients who underwent transplantation after January 2000. These results establish that HLA-matched transplants that use myeloablative conditioning should be the standard of care for eligible pediatric patients with SCD. To date, ∼ 300-400 affected subjects mostly younger than the age of 16 years have undergone fully myeloablative allo-HSCT.11,12,24-27 

The results of these studies are summarized in Table 2. However, the number of patients who have undergone transplantation is small compared with the estimated 70 000 patients with SCD in the United States alone, indicating that this curative therapy is underused. Furthermore, a 15% to 20% GVHD rate with cyclosporine-based postgraft immunosuppression in the myeloablative setting has been reported, with some deaths directly attributable to GVHD. What is remarkable is that despite myeloablative dosing in the conditioning regimens, a mixture of both donor and recipient hematopoietic cells, termed mixed donor chimerism, is consistently observed in approximately 10% to 20% of these children.25  Interestingly, this mixed chimeric state is sufficient to direct BM to produce donor-type hemoglobin and red cells, revert the SCD phenotype, and minimize the risk of GVHD. This observation has both confirmed the therapeutic efficacy and secured the goal of mixed chimerism for the hemoglobinopathies.28 

However, many adults with SCD have accumulated end-organ damage, and presumed excessive toxicity from myeloablative conditioning has prohibited this potentially curative approach. Older adults, especially those older than 35 years of age, are often left with no therapeutic options when they become refractory to hydroxyurea or develop severe allo-immunization precluding red cell transfusions. Nonmyeloablative approaches in which the conditioning regimens were intentionally modified to allow application in sicker patients were thus developed, with potential application to severely affected adults. As seen in Table 3, nonablative HSCTs have been performed in substantially fewer patients, and until recently, the results were not consistently successful. Compared with myeloablative HSCT, mixed chimerism is much more common, yet GVHD is still observed with calcineurin inhibitor–based immunosuppression. The various degrees of sickle-free survival directly result from the various approaches used. Thus, HSCT in adults with SCD remains an area of active research interest, where there are many patients in need, and where there is the potential for immediate clinical benefit.

As depicted in Tables 2 and 3, myeloablative doses of busulfan and high-dose cyclophosphamide have produced the most consistent results in children with SCD. However, the persistent finding of graft rejection and GVHD indicates that further improvements must be made. Reduced doses of busulfan (from 14 mg/kg to 6.4 mg/kg) appeared to allow engraftment,29  whereas a similar reduction in melphalan dosing did not.30  Since then, a bewildering number of conditioning regimens have been developed, mostly in the setting of malignant disease, in an attempt to bring transplantation procedures to a greater number of patients and potentially to those with SCD. To allow comparisons across studies, a recent categorization of conditioning regimens was proposed that used the terms myeloablative (MA), reduced-intensity conditioning, and nonmyeloablative (NMA).31  In the proposed classification, MA regimens were defined as those that cause irreversible pancytopenia and thus require stem cell support. NMA regimens were defined as regimens that produce minimal cytopenia with no need for stem cell support. Those regimens remaining, which fit neither classification, were termed reduced-intensity conditioning. Although this classification should prove useful in comparative analyses, additional conceptual factors could be considered in comparing and even constructing regimens that are disease-dependent. The emphasis currently is placed on lowering toxicity, yet further contemplation on mechanism of engraftment and desired use of the allograft should direct further research in the area. Indeed, not all conditioning regimens are equivalent, and the era of one-size-fits-all is certainly behind us. We propose a further division of the classification of conditioning based on the mechanism of engraftment: immune ablation or myelosuppression with tolerance induction.

In patients with malignancies, myeloablative conditioning regimens designed to both eradicate host hematopoiesis (including malignant cells) and to ensure complete donor immune reconstitution have historically been used with busulfan and cyclophosphamide or cyclophosphamide and total body irradiation (TBI). A graft-versus-malignancy effect has long been recognized and adds to the curative potential of such approaches, securing complete donor immune reconstitution as a necessary goal. Similarly, nonmyeloablative transplantation strategies in malignant disease share the goal of complete eradication of host hematopoiesis as well as replacement of the immune system with that of donor. A graft-versus-malignancy effect can then be harnessed to achieve long-term disease-free survival. With these goals in mind, we and others have developed NMA conditioning regimens that target the host immune system, allowing engraftment to proceed through a donor allo-immune response. The use of strategies such as high-dose cyclophosphamide and fludarabine proved highly successful in attaining engraftment without myeloablation in virtually all patients. Early engraftment profiles after these highly immunosuppressive agents showed high degrees of donor T-cell chimerism and were contrasted by lower levels of donor myeloid chimerism. However, this mixed chimeric state was temporary, and in most cases, alloreactive donor T cells appeared to eradicate recipient stem cells, converting patients from mixed- to full-donor chimerism. In situations in which less than complete donor engraftment was obtained, especially in the presence of residual malignant disease, donor lymphocytes were often used to tip the balance in favor of donor.32,33 

Regimens that rely on donor lymphocyte infusions to establish full donor engraftment are thus of the immune ablative classification. Of note, this immune effect proved successful in eradicating malignant disease in a sizable fraction of recipients, yet was frequently associated with the occurrence of GVHD. Furthermore, although GVHD correlates with graft-versus-tumor effects, it provides no benefit to patients with nonmalignant hematologic disorders and is associated with considerable morbidity and mortality.32,33  In patients with nonmalignant hematologic disorders, complete eradication of host hematopoiesis is usually not necessary, and we submit that the conditioning regimen should be tailored specifically for the targeted patient population, especially when there is evidence that mixed donor chimerism (MDC) could revert the disorder. Indeed, reflexive application of regimens developed for malignant disorders would likely lead to greater than expected transplantation-related morbidity in patients with SCD. We thus favor developing specific regimens, reflecting different patient ages, disease indications, disease burden, cell sources, and degree of HLA matching. In disorders such as SCD and the thalassemias, where low levels of donor chimerism are sufficient to reverse the disease, regimens intentionally aimed at a mixed chimeric state warrant investigation.

Given the limitations of immune ablative regimens for nonmalignant disease, we sought to develop a transplantation regimen for adults with SCD for whom engraftment is not dependent on alloreactivity. Early attempts by others using regimens previously attempted in malignant disorders proved unsuccessful at achieving reliable long-term donor engraftment after nonmyeloablative conditioning in patients with SCD,34  dampening enthusiasm for this approach. We therefore went back to the murine model to tailor a regimen for SCD. We specifically sought to develop a regimen that would allow sufficient myelosuppression to achieve the modest levels of donor myeloid chimerism required for disease reversion while also achieving immunologic tolerance through pharmacologic manipulation. This approach is similar to that sought in solid organ transplantation, whereby myelosuppression represents the equivalent of the organ transplant and the pharmacologic immunomodulatory approach the equivalent of tolerance induction. Several regimens tested in patients with SCD could fit this classification. Indeed, the work of Ianonne et al,34  in which they used 200 cGy of TBI, fludarabine, and immunosuppression with cyclosporine, showed that patients had temporary donor engraftment. However, when cyclosporine was tapered as per protocol, donor grafts decreased to undetectable levels, suggesting that immune tolerance had not been achieved.

Realizing the important finding of this study, we too chose a low-dose radiation approach, but used sirolimus (rapamycin) instead of cyclosporine, on the basis of a novel mechanism of tolerance induction. Unlike calcineurin inhibitors (eg, cyclosporine and tacrolimus), sirolimus does not block T-cell activation (signal 1) but binds to the mammalian target of rapamycin blocking proliferation (signal 2). Signal 1 in the absence of signal 2 renders T cells anergic, promoting T-cell tolerance.35  In a murine peripheral blood stem cell low-dose TBI transplantation model comparing a 30-day course of CSA to sirolimus, long-term, high level chimerism was attained only in sirolimus-treated mice, and mixed lymphocyte reactions demonstrated tolerance to donor cells. Correction of disease was also demonstrated in sickle cell transgenic knockout mice by use of the sirolimus-based regimen.36 

Given the almost exclusive use of G-CSF–mobilized peripheral blood as the source of HSCs for adult transplantation applications along with the 50% probability for heterozygosity for the sickle mutation among sibling donors with sickle cell trait, and the known adverse effects of G-CSF mobilization in SCD37-40  along with potential problems encountered with cryopreservation of SCT products,41-43  we initiated a prospective, controlled clinical trial of peripheral blood hematopoietic stem cell mobilization and processing in patients with sickle cell trait with age- and race-matched controls. Symptoms were similar between the 2 arms, and there was no evidence for G-CSF–induced sickling in subjects with sickle cell trait. HSC mobilization, collection, and cryopreservation also were similar, and a superior cryopreservation method was developed that constitutes our new standard.44 

We then considered how adult patients with accumulated organ damage might tolerate any given conditioning regimen. Those with renal insufficiency may experience worsening renal function with calcineurin inhibitors; those with a history of cerebrovascular or Moya-Moya disease may have severe neurologic complications from posterior reversible encephalopathy syndrome attributable to calcineurin inhibitor use; and those with severe sickle hepatopathy may be at significant risk for veno-occlusive disease from traditional alkylating agents. Thus, we built on the results of our previous work and on the previous nonmyeloablative experiences for adults34,45  and devised a regimen on the basis of 300 cGy of TBI. As additional GVHD prophylaxis, we chose alemtuzumab over antithymocyte globulin for better infusional toxicity profile in patients with SCD and superior prophylaxis against GVHD.46,47  Within days of administration, alemtuzumab depletes majority of circulating host and infused donor lymphocytes. Alemtuzumab remains detectable for several weeks and further deletes alloreactive T cells during subsequent donor engraftment and initial immune reconstitution. Both components are preferable over more technically challenging ex vivo T-cell depletion procedures to allow engraftment and minimize early GVHD. We hypothesized that long-term administration of sirolimus (for 6-12 months) could then further induce sufficient number of regulatory T cells to promote tolerance in a mixed chimeric setting during the remainder of donor immune reconstitution to reduce delayed GVHD. Thus low-dose TBI, alemtuzumab, and sirolimus theoretically provide the necessary environment for stable mixed chimerism and minimal GVHD.

Ten adult patients with severe disease initially underwent transplantation, and their outcomes were reported recently.48  All patients tolerated the conditioning regimen, with infrequent serious adverse events found. All 10 patients are alive at a median follow-up period of 30 months and all but one have long-term donor engraftment. No patients have experienced acute or chronic GVHD. The kinetics of donor myeloid chimerism were rapid and reached nearly 100% early in the majority of patients, whereas the kinetics of donor T-cell engraftment was less rapid and reached a plateau after 12 months. No patient reached 100% donor chimerism in both compartments, and immunosuppression was continued on the basis of our initial withdrawal plan requiring full donor lymphoid chimerism in the absence of GVHD, but a modification of this plan to allow tapering with lymphoid chimerism levels of 50% or greater has allowed the withdrawal of immunosuppression in 4 patients to date. These 4 patients continue to demonstrate stable mixed chimerism in the absence of immunosuppression. The mixed chimeric state allowed complete replacement by donor-type hemoglobin to levels that gradually improved to the normal range post-HSCT. Importantly, the replacement by donor type red cells allowed complete withdrawal of chronic narcotic therapy. The use of nonmyeloablative regimens in this and other studies will also have the benefit of preserving fertility. Because the duration of immunosuppression may be longer in our study than in children who underwent myeloablative conditioning, we are following our patients closely for long-term effects of sirolimus, the effect of our conditioning regimen on pulmonary function and endocrine organs, and the development of secondary malignancies.49 

MDC has frequently been described not only in patients with SCD50,51  but also in those with thalassemia after HSCT, and most patients with MDC are free of their underlying hemoglobin disorders, making MDC a minimal necessary goal. Certainly, the notion that less-than-complete donor engraftment may revert several disorders such as the severe combined immunodeficiencies has fostered work in genetic modification of autologous HSCs that has recently proven effective and continues to support this type of strategy for eventual application in SCD.52-54  However, the minimum percentage of donor cells that defined MDC differed in SCD and thalassemia patients undergoing allo-HSCT, and the cell populations (total leukocytes, mononuclear cells, or lineage specific cells) that were assayed for chimerism also varied; thus, the threshold percentage of donor cells sufficient to ameliorate the hemoglobin disorders has not yet been firmly established. This minimum, however, has been suggested to be as low as 10% in patients with SCD.25  This minimum has been corroborated in thalassemic mice, in whom 10% to 20% is sufficient,55  and 10% to 20% in thalassemic patients after myeloablative HSCT.56 

Others have questioned whether MDC can in fact be stable.57,58  Andreani et al59  recently reported their experience in myeloablative HSCT in patients with thalassemia major. They demonstrated that the risk of graft rejection is the greatest in the first 2 months after HSCT when MDC levels decrease to < 75%. However, this risk dramatically decreases after 1-2 years, even when the MDC levels decrease < 75%,60  indicating the first several months after transplantation are the most critical, when either rejection may occur or recipient hematopoiesis may directly compete with the donor hematopoietic contribution. After this period, patients with MDC appeared stable, with long-term reversion of their disease state.

There are several potential factors that seem to be associated with MDC. Less-intensive conditioning regimens in general are associated with a greater proportion of MDC. Interestingly, the addition of ATG to busulfan and cyclophosphamide increased the percent of French children with MDC (50% to 95% of donor cells) from 5% to 35% undergoing myeloablative HSCT but reduced the graft rejection rate.11  Similarly, the addition of antilymphocyte globulin also increased the proportion of Italian children with MDC undergoing myeloablative HSCT for thalassemia.61  The low-dose TBI, graded doses of alemtuzumab, large doses of G-CSF–mobilized peripheral blood HSCs, and sirolimus were proposed to induce stable MDC in the National Institutes of Health cohort of adults undergoing nonmyeloablative HSCT.48  These results together suggest that in vivo T-cell depletion may be a central component in inducing MDC. No doubt the mechanism(s) of MDC and the characteristics of tolerance between donor and recipient cells need to be elucidated. HSCTs in hemoglobin disorders are then poised to lead the field of tolerance induction because there are many eligible patients, there are shared interests for HSCT between the community and transplant physicians, and most importantly, tolerance without GVHD is the ultimate goal. The stable MDC observed, even in patients weaned from immunosuppression, provides indirect evidence for graft-specific tolerance, opening the possibility of expanding to alternative donor sources.

Although the results of HSCT for SCD in the HLA-matched sibling setting have been encouraging, many barriers to transplantation exist. To explore how much risk they would be willing to accept to cure their children of SCD, parents were provided a questionnaire specifically designed to quantitate this risk.62  Fifty-four percent of parents were willing to accept some short-term mortality risk, but only 13% could accept the estimated risks at the time of a 15% mortality risk with an additional 15% risk of developing GVHD. Another large barrier exists with regard to pursuing HLA typing. In another series, among 4848 patients with SCD who were younger than 16 years of age, 315 (6.5%) met entry criteria for transplantation.63  However, only 128 (41% of eligible or 2.6% of total) underwent HLA typing. Reasons for not performing HLA typing included lack of a sibling donor (24%), lack of financial or psychosocial support (10.5%), parenteral refusal (9.5%), and physician refusal (4%). Further studies in the new era of HSCT for patients with SCD with the improved overall survival and decreased risk of GVHD seen in more recent years to further explore parental and physician opinions and provide education are thus indicated.

However, the main limitation to HSCT for patients with SCD has been a scarcity of HLA-matched sibling donors. From our own experience, where 112 patients had SCD severe enough to be eligible for our transplantation protocol, only 24 patients (21%) had a histocompatible sibling donor. Furthermore, this percentage reflected a numerator that included patients referred solely because of an already-established matched sibling donor. In the aforementioned study, where 315 patients with SCD were found to be eligible for transplantation, only 44 (14% of eligible patients) had an HLA-matched sibling.63  Therefore, there is an urgent need to increase the donor pool so that HSCT can be made available to the majority of patients with severe SCD.

Cord blood (CB) transplantation (CBT), which was first successfully reported in a patient with SCD in 199664  (Table 4), offers the opportunity to obtain the donor source at a much faster rate compared with matched unrelated donor grafts and theoretically allows for a greater degree of mismatching. The experience of CBT has, however, been limited in patients with SCD, with a total of 33 cases reported, two-thirds of whom were in the related donor setting.64-71  The majority of related CBT have been 6/6 HLA-matched. Conditioning has predominantly consisted of busulfan and cyclophosphamide with or without ATG and calcineurin inhibitor–based GVHD prophylaxis. There has only been one reported death, and event-free survival in these small studies has ranged from 66% to 100%, with only a total of 3 patients developing grade 1-2 acute GVHD, and one patient developing limited chronic GVHD. Although these results on the whole are remarkable, the chances of finding a related HLA-identical CB unit are no better than that of finding an HLA-matched sibling.68  In addition, the relatively low cell dose attainable with CB restricts broad application.

There has been one report of double umbilical cord transplantation, which has been used in an attempt to augment the total cell dose, in a young adult patient with SCD and Hodgkin lymphoma.71  However, double umbilical cord transplants may not provide sufficient cell dose to overcome the graft rejection barrier in nonablative studies, compared with the large doses of CD34 cells that can be collected from G-CSF–mobilized peripheral blood. Certainly, in the nonmyeloablative setting, cell dose may be even more critical, and our success in the HLA-matched sibling setting may relate in part to the high doses of CD34+ cells (> 20 × 106 CD34+ cells per kilogram of recipient body weight) obtained from our donors. As more related CBT are performed in patients with SCD, we may find that the CD34 dose collected may not be sufficient to overcome the host versus graft barrier, even in the HLA-identical setting. Given the nonacute need for transplantation in this disorder, a greater dose of cells could easily be collected from the cord donor at a later date, potentially improving outcome in the nonmyeloablative setting.

In unrelated CBT, conditioning regimens have varied immensely, and the results have been much less successful (Table 4). The largest study consisting of 7 patients reported one death, an event-free survival of 43%, and 5 patients developed acute GVHD (2 with grade 3-4 acute GVHD) and one patient chronic extensive GVHD.69  These discouraging results compared with related CBT are likely because none of the transplantations were 6/6 HLA-matched. In addition, because of the African ancestry of most Americans with SCD, the chance of finding a 6/6 HLA-matched umbilical cord unit is dismal. Further, to our knowledge, there are no reports of matched unrelated donor HSCT for patients with SCD, again likely because of the difficulty finding a suitable matched unrelated donor. Although the National Marrow Donor Program (NMDP) has registered > 4 million volunteer donors, only a small minority have been underrepresented minority donors.72  The number of African-American volunteer donors has, however, increased from 59 104 to 345 236 between December 1993 and April 2001, respectively. Between 1989 and 2001, the NMDP performed donor searches for 77 patients with SCD.72  Presumably, the majority of patients were children or young adults; 34% had 1-2 possible HLA-matched donors, and 26% of patients had at least 3 potential HLA-matched donors. A total of 46% of patients had 1-2 possible CB units available, and 71% of patients had at least 3 potential CB units available. Others have theorized that 54% of patients weighing 40 kg would match at least 4 HLA CB unit loci at a cell dose of at least 5 × 107 nucleated cells/kg.73  These results reflect HLA-typing at the antigen level only and are thus an overestimate of the probability of finding a donor.

In 2007, we initiated a donor search through the NMDP and Bone Marrow Donors Worldwide for 10 adult patients with severe SCD who lacked HLA-matched sibling donors to evaluate the feasibility of matched unrelated donor or CB transplantation for adult patients with SCD. Although potential donors were identified by serotyping in the majority, we found that only one patient had a greater than 1% chance of having a 6/6 HLA-matched donor according to Haplogic algorithm.74  Further, Dew and colleagues75  performed unrelated donor searches for 51 African-Americans with malignant or nonmalignant hematologic diseases and who were a median age of 38 years. Only 3 patients (6%) were found to have an HLA-matched donor by molecular typing at all loci. However, 50 of the patients (98%) had at least one and frequently > 1 (median 14, range 1-158) 4/6 HLA-matched CB units available with a median cell dose of 2.85 × 107 total nucleated cells per kilogram body weight. However, in our donor search, only a median of one suitable CB unit was available per patient (≥ 2.5 × 107 total nucleated cells per kilogram body weight, at least 4/6 HLA-matched, and ABO-matched). Greater degrees of matching reduced this median to zero. The discrepancy between the 2 searches likely exists because of our exclusion of major ABO mismatched cords because of the possible risk for the development of pure red cell aplasia posttransplantation in the nonmyeloablative setting because of persistent recipient anti-donor red cell antibodies should stable mixed chimerism ensue.

Therefore, although some patients have been successfully treated with CBT, alternative donor sources are limited principally by availability, GVHD, and graft rejection. HLA-identical related CBT, which may have a lower risk of graft rejection and GVHD, will be severely limited by the majority of patients not having a matched related donor and the limited cell count collected. Matched unrelated donor and matched unrelated CBT will likely not be applicable to sufficient number of patients with SCD because of a lack of suitable donors in the national and worldwide registries. Mismatched unrelated CBT, although more feasible for pediatric patients on the basis of the availability of 4/6 HLA-matched cord blood units, appears to be associated with a greater risk of graft rejection and GVHD on the basis of the limited data. Further, CBT for adults will further be limited by the necessary total nucleated cell count per kilogram of body weight for engraftment to occur and the difficulty in achieving this goal in adult patients.

In contrast, haploidentical donors would greatly increase the donor pool because the majority of patients will have a suitable parent, child, or haploidentical sibling donor. Haploidentical HSCT is invariably associated with a greater risk of graft rejection and GVHD compared with the matched related or matched unrelated donor setting. The authors of one study reported that in patients with hematologic malignancies, HLA-mismatched compared with HLA-matched family members led to an incidence of acute grade 3-4 GVHD of 31% of patients compared with 9%, respectively (P < .0001), and engraftment failure occurred in 6.3% versus 2.4%, respectively (P = .01).76  There was no significant difference between the incidence of chronic GVHD between groups (60% vs 47%, respectively, P = .11). In another study also in patients with hematologic malignancies, the authors reported that graft failure occurred in 12.3% of haploidentical relatives compared with 2% of HLA-matched siblings (P < .0001).77  Furthermore, Anasetti et al78  demonstrated that in recipients of haploidentical grafts, the incidence of severe GVHD was associated with the amount of HLA incompatibility, with a relative risk of 1.95 for each incompatible locus (95% confidence interval 1.52-2.5, P < .0001). Others have reported an incidence of grade 2-4 acute GVHD ranging from 12% to 54%, grade 3-4 acute GVHD ranging from 6% to 13%, and chronic extensive GVHD ranging from 13% to 48% for patients with hematologic malignancies undergoing haploidentical HSCT.79-83 

Unlike hematologic malignancies, where survival is expected to be poor and the graft-versus leukemia (GVL) effect is beneficial, SCD is in general a chronic illness, and substituting one chronic illness with another in the form of GVHD is highly undesirable. Perhaps the risk of GVHD will be lower for patients with SCD versus patients with hematologic malignancies because of the absence of irradiation and chemotherapy before transplantation. Instead, patients with SCD may be at an increased risk for graft failure because of the strong hematopoietic drive and high incidence of alloimmunization. Indeed, one case report demonstrated failure to engraft in a 14-year-old boy with severe SCD after receiving a BM transplant from his mother (Table 5).84  Because the patient was only conditioned with fludarabine and 200 cGy of TBI, a more intensive regimen is likely necessary to overcome the engraftment barrier. However, the authors of another report cited 3 patients with paroxysmal nocturnal hemoglobinuria, one of whom also had SCD, who underwent haploidentical HSCT and were conditioned with fludarabine, 200 cGy of TBI, and pre- and posttransplant cyclophosphamide (Table 5).85  Two of the patients, including the patient with SCD, engrafted with no evidence of GVHD, and both patients were free of disease at 1 and 4 years after transplantation. The development of a safe and effective low-intensity regimen that can be applied to the majority of patients with severe SCD with the use of haploidentical donors is necessary, and further research in this area is currently ongoing.

For those lacking a suitable allogeneic HSC donor, autologous HSC gene transfer with the use of viral vectors designed to permanently integrate a therapeutic gene into the target cell chromosomal DNA remains a rational alternative. Proof of concept through clinical successes in the immunodeficiencies and storage disorders has recently renewed enthusiasm for this approach,86-90  although vector-mediated insertional mutagenesis has also proven to be more than a theoretical risk in several patients. The globin disorders have long been held as a therapeutic target but have proven much more difficult because of the requirement for regulated, lineage-specific, high-level globin expression. The discovery of the human β-globin locus control region (LCR), which functions as a tissue-specific enhancer, led to the evolution of a new generation of globin retroviral vectors incorporating 4 or 5 hypersensitive LCR elements able to direct erythroid specific globin expression; however, these vectors were highly unstable and proved unreliable. The stable incorporation of these large LCR fragments became feasible with the development of vector systems based on HIV1, allowing for the first-time regulated human β-globin expression sufficient to revert the phenotype in a β-thalassemia mouse model,91  with confirmation soon after for both thalassemia and SCD.92-94  Recently, promising results were reported in a single patient with severe βE0 thalassemia after autologous HSC gene transfer with the use of an HIV1-based vector encoding a mutant human β globin.95  At ∼ 1 year after transplantation, hemoglobin levels increased to ∼ 10 g/dL, allowing therapeutic phlebotomy, with an estimated 3 g/dL contribution by the therapeutic vector. This increase was, however, accompanied by an increase in the contribution by a clone bearing an integration in the HMGA2 gene, a gene that when overexpressed results in myeloproliferation.96  The significance of this clonal expansion will require long-term follow-up, and efforts to improve on the safety of integrating vectors remain a priority.

HSCT for patients with SCD, although highly effective, is underused. The cumulative data on myeloablative HSCT in children and nonmyeloablative HSCT in adults indicate that there are shared interests in the sickle cell community to use HSCT as an immediately available curative therapy for patients who are at high risk for SCD-related morbidity and mortality. In these high-risk children and adults, enrollment in well-designed clinical studies can be justified, and SCD-specific transplantation regimens can emerge. We need successive regimens to be tested that use G-CSF–mobilized versus steady-state BM-derived HSCs and promote MDC as a minimum without exchanging for GVHD or graft failure. Future well-designed studies are poised to lead the field in immune tolerance and provide insight to differentiate graft-versus-host and graft-versus-malignancy effects. Indeed, achieving tolerance should allow extension of these potentially curative approaches to alternative donor sources. These efforts no doubt will continue to benefit children and now adults with chronic organ damage and in the future may allow application earlier in the disease. HSCT by the use of regimens developed in hematologic malignancies have brought cures to patients with SCD; it is time to build on these experiences with SCD-specific issues in mind to increase the use of HSCT while minimizing morbidity and mortality.

This work is supported by the intramural research program of the National Institutes of Health in the National Heart, Lung, and Blood Institute and the National Institute of Diabetes and Digestive and Kidney Diseases.

National Institutes of Health

Contribution: M.M.H., C.D.F., and J.F.T. reviewed the literature, analyzed the data, wrote the manuscript, and approved the final submission.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: John Tisdale, MD, 9000 Rockville Pike, Bldg 10 9N 112, Bethesda, MD 20892; e-mail: johntis@mail.nih.gov.

1
Charache
 
S
Terrin
 
ML
Moore
 
RD
et al. 
Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia [see comments].
N Engl J Med
1995
, vol. 
332
 
20
(pg. 
1317
-
1322
)
2
Ferster
 
A
Vermylen
 
C
Cornu
 
G
et al. 
Hydroxyurea for treatment of severe sickle cell anemia: a pediatric clinical trial.
Blood
1996
, vol. 
88
 
6
(pg. 
1960
-
1964
)
3
Rodgers
 
GP
Dover
 
GJ
Noguchi
 
CT
Schechter
 
AN
Nienhuis
 
AW
Hematologic responses of patients with sickle cell disease to treatment with hydroxyurea.
N Engl J Med
1990
, vol. 
322
 
15
(pg. 
1037
-
1045
)
4
Wayne
 
AS
Schoenike
 
SE
Pegelow
 
CH
Financial analysis of chronic transfusion for stroke prevention in sickle cell disease.
Blood
2000
, vol. 
96
 
7
(pg. 
2369
-
2372
)
5
Bernaudin
 
F
Verlhac
 
S
Arnaud
 
C
et al. 
Impact of early transcranial Doppler screening and intensive therapy on cerebral vasculopathy outcome in a newborn sickle cell anemia cohort.
Blood
2011
, vol. 
117
 
4
(pg. 
1130
-
1140
)
6
Kwiatkowski
 
JL
Yim
 
E
Miller
 
S
Adams
 
RJ
Effect of transfusion therapy on transcranial Doppler ultrasonography velocities in children with sickle cell disease.
Pediatr Blood Cancer
2011
, vol. 
56
 
5
(pg. 
777
-
782
)
7
Hulbert
 
ML
McKinstry
 
RC
Lacey
 
JL
et al. 
Silent cerebral infarcts occur despite regular blood transfusion therapy after first strokes in children with sickle cell disease.
Blood
1
20
, vol. 
117
 
3
(pg. 
772
-
779
)
8
Godfrey
 
GJ
Lockwood
 
W
Kong
 
M
Bertolone
 
S
Raj
 
A
Antibody development in pediatric sickle cell patients undergoing erythrocytapheresis.
Pediatr Blood Cancer
2010
, vol. 
55
 
6
(pg. 
1134
-
1137
)
9
Lasalle-Williams
 
M
Nuss
 
R
Le
 
T
et al. 
Extended red blood cell antigen matching for transfusions in sickle cell disease: a review of a 14-year experience from a single center [published online ahead of print February 18, 2011].
Transfusion
 
10
McPherson
 
ME
Anderson
 
AR
Castillejo
 
MI
et al. 
HLA alloimmunization is associated with RBC antibodies in multiply transfused patients with sickle cell disease.
Pediatr Blood Cancer
2010
, vol. 
54
 
4
(pg. 
552
-
558
)
11
Bernaudin
 
F
Socie
 
G
Kuentz
 
M
et al. 
Long-term results of related, myeloablative stem cell transplantation to cure sickle cell disease.
Blood
2007
, vol. 
110
 
7
(pg. 
2749
-
2756
)
12
Walters
 
MC
Patience
 
M
Leisenring
 
W
et al. 
Bone marrow transplantation for sickle cell disease.
N Engl J Med
1996
, vol. 
335
 
6
(pg. 
369
-
376
)
13
Fitzhugh
 
CD
Lauder
 
N
Jonassaint
 
JC
et al. 
Cardiopulmonary complications leading to premature deaths in adult patients with sickle cell disease.
Am J Hematol
2010
, vol. 
85
 
1
(pg. 
36
-
40
)
14
Powars
 
DR
Chan
 
LS
Hiti
 
A
Ramicone
 
E
Johnson
 
C
Outcome of sickle cell anemia: a 4-decade observational study of 1056 patients.
Medicine (Baltimore)
2005
, vol. 
84
 
6
(pg. 
363
-
376
)
15
Gladwin
 
Mt
Sachdev
 
V
Jison
 
ML
et al. 
Pulmonary hypertension as a risk factor for death in patients with sickle cell disease.
N Engl J Med
2004
, vol. 
350
 
9
(pg. 
886
-
895
)
16
Powars
 
DR
Elliott-Mills
 
DD
Chan
 
L
et al. 
Chronic renal failure in sickle cell disease: risk factors, clinical course, and mortality.
Ann Intern Med
1991
, vol. 
115
 
8
(pg. 
614
-
620
)
17
Bakir
 
AA
Hathiwala
 
SC
Ainis
 
H
et al. 
Prognosis of the nephrotic syndrome in sickle glomerulopathy. A retrospective study.
Am J Nephrol
1987
, vol. 
7
 
2
(pg. 
110
-
115
)
18
Darbari
 
DS
Kple-Faget
 
P
Kwagyan
 
J
Rana
 
S
Gordeuk
 
VR
Castro
 
O
Circumstances of death in adult sickle cell disease patients.
Am J Hematol
2006
, vol. 
81
 
11
(pg. 
858
-
863
)
19
Ahn
 
H
Li
 
CS
Wang
 
W
Sickle cell hepatopathy: clinical presentation, treatment, and outcome in pediatric and adult patients.
Pediatr Blood Cancer
2005
, vol. 
45
 
2
(pg. 
184
-
190
)
20
Johnson
 
FL
Look
 
AT
Gockerman
 
J
Ruggiero
 
MR
Dalla-Pozza
 
L
Billings
 
FT
Bone-marrow transplantation in a patient with sickle-cell anemia.
N Engl J Med
1984
, vol. 
311
 
12
(pg. 
780
-
783
)
21
Vermylen
 
C
Cornu
 
G
Ferster
 
A
Ninane
 
J
Sariban
 
E
Bone marrow transplantation in sickle cell disease: the Belgian experience.
Bone Marrow Transplant
1993
, vol. 
12
 
suppl 1
(pg. 
116
-
117
)
22
Bernaudin
 
F
Souillet
 
G
Vannier
 
JP
et al. 
Bone marrow transplantation (BMT) in 14 children with severe sickle cell disease (SCD): the French experience. GEGMO.
Bone Marrow Transplant
1993
, vol. 
12
 
suppl 1
(pg. 
118
-
121
)
23
Walters
 
MC
Sullivan
 
KM
Bernaudin
 
F
et al. 
Neurologic complications after allogeneic marrow transplantation for sickle cell anemia [see comments].
Blood
1995
, vol. 
85
 
4
(pg. 
879
-
884
)
24
Vermylen
 
C
Cornu
 
G
Ferster
 
A
et al. 
Haematopoietic stem cell transplantation for sickle cell anaemia: the first 50 patients transplanted in Belgium.
Bone Marrow Transplant
1998
, vol. 
22
 
1
(pg. 
1
-
6
)
25
Walters
 
MC
Patience
 
M
Leisenring
 
W
et al. 
Stable mixed hematopoietic chimerism after bone marrow transplantation for sickle cell anemia.
Biol Blood Marrow Transplant
2001
, vol. 
7
 
12
(pg. 
665
-
673
)
26
Brachet
 
C
Azzi
 
N
Demulder
 
A
et al. 
Hydroxyurea treatment for sickle cell disease: impact on haematopoietic stem cell transplantation's outcome.
Bone Marrow Transplant
2004
, vol. 
33
 
8
(pg. 
799
-
803
)
27
Panepinto
 
JA
Walters
 
MC
Carreras
 
J
et al. 
Matched-related donor transplantation for sickle cell disease: report from the Center for International Blood and Transplant Research.
Br J Haematol
2007
, vol. 
137
 
5
(pg. 
479
-
485
)
28
Wu
 
CJ
Gladwin
 
M
Tisdale
 
J
et al. 
Mixed haematopoietic chimerism for sickle cell disease prevents intravascular haemolysis.
Br J Haematol
2007
, vol. 
139
 
3
(pg. 
504
-
507
)
29
Jacobsohn
 
DA
Duerst
 
R
Tse
 
W
Kletzel
 
M
Reduced intensity haemopoietic stem-cell transplantation for treatment of non-malignant diseases in children.
Lancet
2004
, vol. 
364
 
9429
(pg. 
156
-
162
)
30
Shenoy
 
S
Has stem cell transplantation come of age in the treatment of sickle cell disease?
Bone Marrow Transplant
2007
, vol. 
40
 
9
(pg. 
813
-
821
)
31
Bacigalupo
 
A
Ballen
 
K
Rizzo
 
D
et al. 
Defining the intensity of conditioning regimens: working definitions.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
12
(pg. 
1628
-
1633
)
32
Childs
 
R
Chernoff
 
A
Contentin
 
N
et al. 
Regression of metastatic renal-cell carcinoma after nonmyeloablative allogeneic peripheral-blood stem-cell transplantation.
N Engl J Med
2000
, vol. 
343
 
11
(pg. 
750
-
758
)
33
Srinivasan
 
R
Takahashi
 
Y
McCoy
 
JP
et al. 
Overcoming graft rejection in heavily transfused and allo-immunised patients with bone marrow failure syndromes using fludarabine-based haematopoietic cell transplantation.
Br J Haematol
2006
, vol. 
133
 
3
(pg. 
305
-
314
)
34
Iannone
 
R
Casella
 
JF
Fuchs
 
EJ
et al. 
Results of minimally toxic nonmyeloablative transplantation in patients with sickle cell anemia and beta-thalassemia.
Biol Blood Marrow Transplant
2003
, vol. 
9
 
8
(pg. 
519
-
528
)
35
Powell
 
JD
Lerner
 
CG
Schwartz
 
RH
Inhibition of cell cycle progression by rapamycin induces T cell clonal anergy even in the presence of costimulation.
J Immunol
1999
, vol. 
162
 
5
(pg. 
2775
-
2784
)
36
Powell
 
JD
Fitzhugh
 
C
Kang
 
EM
Hsieh
 
M
Schwartz
 
RH
Tisdale
 
JF
Low-dose radiation plus rapamycin promotes long-term bone marrow chimerism.
Transplantation
2005
, vol. 
80
 
11
(pg. 
1541
-
1545
)
37
Abboud
 
M
Laver
 
J
Blau
 
CA
Granulocytosis causing sickle-cell crisis [letter].
Lancet
1998
, vol. 
351
 
9107
pg. 
959
 
38
Adler
 
BK
Salzman
 
DE
Carabasi
 
MH
Vaughan
 
WP
Reddy
 
VV
Prchal
 
JT
Fatal sickle cell crisis after granulocyte colony-stimulating factor administration.
Blood
2001
, vol. 
97
 
10
(pg. 
3313
-
3314
)
39
Grigg
 
AP
Granulocyte colony-stimulating factor-induced sickle cell crisis and multiorgan dysfunction in a patient with compound heterozygous sickle cell/beta+ thalassemia.
Blood
2001
, vol. 
97
 
12
(pg. 
3998
-
3999
)
40
Fitzhugh
 
CD
Hsieh
 
MM
Bolan
 
CD
Saenz
 
C
Tisdale
 
JF
Granulocyte colony-stimulating factor (G-CSF) administration in individuals with sickle cell disease: time for a moratorium?
Cytotherapy
2009
, vol. 
11
 
4
(pg. 
464
-
471
)
41
Meryman
 
HT
Hornblower
 
M
Freezing and deglycerolizing sickle-trait red blood cells.
Transfusion
1976
, vol. 
16
 
6
(pg. 
627
-
632
)
42
Bodensteiner
 
D
White cell reduction in blood from donors with sickle cell trait [letter].
Transfusion
1994
, vol. 
34
 
1
pg. 
84
 
43
Kelleher
 
JF
Luban
 
NL
Transfusion of frozen erythrocytes from a donor with sickle trait.
Transfusion
1984
, vol. 
24
 
2
(pg. 
167
-
168
)
44
Kang
 
EM
Areman
 
EM
David-Ocampo
 
V
et al. 
Mobilization, collection, and processing of peripheral blood stem cells in individuals with sickle cell trait.
Blood
2002
, vol. 
99
 
3
(pg. 
850
-
855
)
45
Horan
 
JT
Liesveld
 
JL
Fenton
 
P
Blumberg
 
N
Walters
 
MC
Hematopoietic stem cell transplantation for multiply transfused patients with sickle cell disease and thalassemia after low-dose total body irradiation, fludarabine, and rabbit anti-thymocyte globulin.
Bone Marrow Transplant
2005
, vol. 
35
 
2
(pg. 
171
-
177
)
46
Kottaridis
 
PD
Milligan
 
DW
Chopra
 
R
et al. 
In vivo CAMPATH-1H prevents graft-versus-host disease following nonmyeloablative stem cell transplantation.
Blood
2000
, vol. 
96
 
7
(pg. 
2419
-
2425
)
47
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
, vol. 
99
 
3
(pg. 
1071
-
1078
)
48
Hsieh
 
MM
Kang
 
EM
Fitzhugh
 
CD
et al. 
Allogeneic hematopoietic stem-cell transplantation for sickle cell disease.
N Engl J Med
2009
, vol. 
361
 
24
(pg. 
2309
-
2317
)
49
Fitzhugh
 
CD
Perl
 
S
Hsieh
 
MM
Late effects of myeloablative bone marrow transplantation (BMT) in sickle cell disease (SCD).
Blood
2008
, vol. 
111
 
3
(pg. 
1742
-
1743
author reply 1744
50
Krishnamurti
 
L
Kharbanda
 
S
Biernacki
 
MA
et al. 
Stable long-term donor engraftment following reduced-intensity hematopoietic cell transplantation for sickle cell disease.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
11
(pg. 
1270
-
1278
)
51
Horwitz
 
ME
Spasojevic
 
I
Morris
 
A
et al. 
Fludarabine-based nonmyeloablative stem cell transplantation for sickle cell disease with and without renal failure: clinical outcome and pharmacokinetics.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
12
(pg. 
1422
-
1426
)
52
Aiuti
 
A
Cattaneo
 
F
Galimberti
 
S
et al. 
Gene therapy for immunodeficiency due to adenosine deaminase deficiency.
N Engl J Med
2009
, vol. 
360
 
5
(pg. 
447
-
458
)
53
Hacein-Bey-Abina
 
S
Hauer
 
J
Lim
 
A
et al. 
Efficacy of gene therapy for X-linked severe combined immunodeficiency.
N Engl J Med
2010
, vol. 
363
 
4
(pg. 
355
-
364
)
54
Stein
 
S
Ott
 
MG
Schultze-Strasser
 
S
et al. 
Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease.
Nat Med
2010
, vol. 
16
 
2
(pg. 
198
-
204
)
55
Persons
 
DA
Allay
 
ER
Sabatino
 
DE
Kelly
 
P
Bodine
 
DM
Nienhuis
 
AW
Functional requirements for phenotypic correction of murine beta-thalassemia: implications for human gene therapy.
Blood
2001
, vol. 
97
 
10
(pg. 
3275
-
3282
)
56
Andreani
 
M
Testi
 
M
Gaziev
 
J
et al. 
Quantitatively different red cells/nucleated cell chimerism in patients with long-term persistant hematopoietic mixed chimerism after bone marrow transplantation for thalassemia major or sickle cell disease.
Haematologica
2011
, vol. 
96
 
1
(pg. 
128
-
133
)
57
Shenoy
 
S
Grossman
 
WJ
DiPersio
 
J
et al. 
A novel reduced-intensity stem cell transplant regimen for nonmalignant disorders.
Bone Marrow Transplant
2005
, vol. 
35
 
4
(pg. 
345
-
352
)
58
van Besien
 
K
Bartholomew
 
A
Stock
 
W
et al. 
Fludarabine-based conditioning for allogeneic transplantation in adults with sickle cell disease.
Bone Marrow Transplant
2000
, vol. 
26
 
4
(pg. 
445
-
449
)
59
Andreani
 
M
Testi
 
M
Battarra
 
M
et al. 
Relationship between mixed chimerism and rejection after bone marrow transplantation in thalassaemia.
Blood Transfus
2008
, vol. 
6
 
3
(pg. 
143
-
149
)
60
Andreani
 
M
Nesci
 
S
Lucarelli
 
G
et al. 
Long-term survival of ex-thalassemic patients with persistent mixed chimerism after bone marrow transplantation.
Bone Marrow Transplant
2000
, vol. 
25
 
4
(pg. 
401
-
404
)
61
Nesci
 
S
Manna
 
M
Andreani
 
M
Fattorini
 
P
Graziosi
 
G
Lucarelli
 
G
Mixed chimerism in thalassemic patients after bone marrow transplantation.
Bone Marrow Transplant
1992
, vol. 
10
 
2
(pg. 
143
-
146
)
62
Kodish
 
E
Lantos
 
J
Stocking
 
C
Singer
 
PA
Siegler
 
M
Johnson
 
FL
Bone marrow transplantation for sickle cell disease. A study of parents' decisions.
N Engl J Med
1991
, vol. 
325
 
19
(pg. 
1349
-
1353
)
63
Walters
 
MC
Patience
 
M
Leisenring
 
W
et al. 
Barriers to bone marrow transplantation for sickle cell anemia.
Biol Blood Marrow Transplant
1996
, vol. 
2
 
2
(pg. 
100
-
104
)
64
Brichard
 
B
Vermylen
 
C
Ninane
 
J
Cornu
 
G
Persistence of fetal hemoglobin production after successful transplantation of cord blood stem cells in a patient with sickle cell anemia [see comments].
J Pediatr
1996
, vol. 
128
 
2
(pg. 
241
-
243
)
65
Miniero
 
R
Rocha
 
V
Saracco
 
P
et al. 
Cord blood transplantation (CBT) in hemoglobinopathies. Eurocord.
Bone Marrow Transplant
1998
, vol. 
22
 
suppl 1
(pg. 
S78
-
S79
)
66
Gore
 
L
Lane
 
PA
Quinones
 
RR
Giller
 
RH
Successful cord blood transplantation for sickle cell anemia from a sibling who is human leukocyte antigen-identical: implications for comprehensive care.
J Pediatr Hematol Oncol
2000
, vol. 
22
 
5
(pg. 
437
-
440
)
67
Locatelli
 
F
Rocha
 
V
Reed
 
W
et al. 
Related umbilical cord blood transplantation in patients with thalassemia and sickle cell disease.
Blood
2003
, vol. 
101
 
6
(pg. 
2137
-
2143
)
68
Walters
 
MC
Quirolo
 
L
Trachtenberg
 
ET
et al. 
Sibling donor cord blood transplantation for thalassemia major: Experience of the Sibling Donor Cord Blood Program.
Ann NY Acad Sci
2005
, vol. 
1054
 (pg. 
206
-
213
)
69
Adamkiewicz
 
TV
Szabolcs
 
P
Haight
 
A
et al. 
Unrelated cord blood transplantation in children with sickle cell disease: review of four-center experience.
Pediatr Transplant
2007
9
, vol. 
11
 
6
(pg. 
641
-
644
)
70
Mazur
 
M
Kurtzberg
 
J
Halperin
 
E
Ciocci
 
G
Szabolcs
 
P
Transplantation of a child with sickle cell anemia with an unrelated cord blood unit after reduced intensity conditioning.
J Pediatr Hematol Oncol
2006
, vol. 
28
 
12
(pg. 
840
-
844
)
71
Sauter
 
C
Rausen
 
AR
Barker
 
JN
Successful unrelated donor cord blood transplantation for adult sickle cell disease and Hodgkin lymphoma.
Bone Marrow Transplant
2010
, vol. 
45
 
7
pg. 
1252
 
72
Krishnamurti
 
L
Abel
 
S
Maiers
 
M
Flesch
 
S
Availability of unrelated donors for hematopoietic stem cell transplantation for hemoglobinopathies.
Bone Marrow Transplant
2003
, vol. 
31
 
7
(pg. 
547
-
550
)
73
Adamkiewicz
 
TV
Boyer
 
MW
Bray
 
R
Haight
 
A
Yeager
 
AM
Identification of unrelated cord blood units for hematopoietic stem cell transplantation in children with sickle cell disease.
J Pediatr Hematol Oncol
2006
, vol. 
28
 
1
(pg. 
29
-
32
)
74
Hsieh
 
M
Wilder
 
J
Fitzhugh
 
C
Link
 
B
Tisdale
 
JF
Results of alternative donor search in adult patients with severe sickle cell disease (SCD) eligible for hematopoietic stem cell transplantation (HSCT).
Paper presented at Annual Meeting of the American Society of Hematology Annual Meetingm
December, 2007
Atlanta, GA
75
Dew
 
A
Collins
 
D
Artz
 
A
et al. 
Paucity of HLA-identical unrelated donors for African-Americans with hematologic malignancies: the need for new donor options.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
8
(pg. 
938
-
941
)
76
Kanda
 
Y
Chiba
 
S
Hirai
 
H
et al. 
Allogeneic hematopoietic stem cell transplantation from family members other than HLA-identical siblings over the last decade (1991-2000).
Blood
2003
, vol. 
102
 
4
(pg. 
1541
-
1547
)
77
Anasetti
 
C
Amos
 
D
Beatty
 
PG
et al. 
Effect of HLA compatibility on engraftment of bone marrow transplants in patients with leukemia or lymphoma.
N Engl J Med
1989
, vol. 
320
 
4
(pg. 
197
-
204
)
78
Anasetti
 
C
Beatty
 
PG
Storb
 
R
et al. 
Effect of HLA incompatibility on graft-versus-host disease, relapse, and survival after marrow transplantation for patients with leukemia or lymphoma.
Hum Immunol
1990
, vol. 
29
 
2
(pg. 
79
-
91
)
79
Wang
 
HX
Yan
 
HM
Duan
 
LN
et al. 
Haploidentical hematopoietic stem cell transplantation in child hematologic malignancies with G-CSF–mobilized marrow grafts without T-cell depletion: a single-center report of 45 cases.
Pediatr Hematol Oncol
2009
, vol. 
26
 
3
(pg. 
119
-
128
)
80
Dodero
 
A
Carniti
 
C
Raganato
 
A
et al. 
Haploidentical stem cell transplantation after a reduced-intensity conditioning regimen for the treatment of advanced hematologic malignancies: posttransplantation CD8-depleted donor lymphocyte infusions contribute to improve T-cell recovery.
Blood
2009
, vol. 
113
 
19
(pg. 
4771
-
4779
)
81
Huang
 
X
Liu
 
D
Liu
 
K
et al. 
Haploidentical hematopoietic stem cell transplantation without in vitro T cell depletion for treatment of hematologic malignancies in children.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
1 suppl
(pg. 
91
-
94
)
82
Huang
 
XJ
Liu
 
DH
Liu
 
KY
et al. 
Haploidentical hematopoietic stem cell transplantation without in vitro T-cell depletion for the treatment of hematological malignancies.
Bone Marrow Transplant
2006
, vol. 
38
 
4
(pg. 
291
-
297
)
83
Luznik
 
L
O'Donnell
 
PV
Symons
 
HJ
et al. 
HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
6
(pg. 
641
-
650
)
84
Raj
 
A
Bertolone
 
S
Cheerva
 
A
Successful treatment of refractory autoimmune hemolytic anemia with monthly rituximab following nonmyeloablative stem cell transplantation for sickle cell disease.
J Pediatr Hematol Oncol
2004
, vol. 
26
 
5
(pg. 
312
-
314
)
85
Brodsky
 
RA
Luznik
 
L
Bolanos-Meade
 
J
Leffell
 
MS
Jones
 
RJ
Fuchs
 
EJ
Reduced intensity HLA-haploidentical BMT with post transplantation cyclophosphamide in nonmalignant hematologic diseases.
Bone Marrow Transplant
2008
, vol. 
42
 
8
(pg. 
523
-
527
)
86
Cavazzana-Calvo
 
M
Hacein-Bey
 
S
de Saint Basile
 
G
et al. 
Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease [see comments].
Science
2000
, vol. 
288
 
5466
(pg. 
669
-
672
)
87
Aiuti
 
A
Vai
 
S
Mortellaro
 
A
et al. 
Immune reconstitution in ADA-SCID after PBL gene therapy and discontinuation of enzyme replacement.
Nat Med
2002
, vol. 
8
 
5
(pg. 
423
-
425
)
88
Aiuti
 
A
Slavin
 
S
Aker
 
M
et al. 
Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning.
Science
2002
, vol. 
296
 
5577
(pg. 
2410
-
2413
)
89
Ott
 
MG
Schmidt
 
M
Schwarzwaelder
 
K
et al. 
Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1.
Nat Med
2006
, vol. 
12
 
4
(pg. 
401
-
409
)
90
Aiuti
 
A
Cassani
 
B
Andolfi
 
G
et al. 
Multilineage hematopoietic reconstitution without clonal selection in ADA-SCID patients treated with stem cell gene therapy.
J Clin Invest
2007
, vol. 
117
 
8
(pg. 
2233
-
2240
)
91
May
 
C
Rivella
 
S
Callegari
 
J
et al. 
Therapeutic haemoglobin synthesis in beta-thalassaemic mice expressing lentivirus-encoded human beta-globin.
Nature
2000
, vol. 
406
 
6791
(pg. 
82
-
86
)
92
Persons
 
DA
Allay
 
ER
Sawai
 
N
et al. 
Successful treatment of murine beta-thalassemia using in vivo selection of genetically modified, drug-resistant hematopoietic stem cells.
Blood
2003
, vol. 
102
 
2
(pg. 
506
-
513
)
93
Persons
 
DA
Hargrove
 
PW
Allay
 
ER
Hanawa
 
H
Nienhuis
 
AW
The degree of phenotypic correction of murine beta -thalassemia intermedia following lentiviral-mediated transfer of a human gamma-globin gene is influenced by chromosomal position effects and vector copy number.
Blood
2003
, vol. 
101
 
6
(pg. 
2175
-
2183
)
94
Pawliuk
 
R
Westerman
 
KA
Fabry
 
ME
et al. 
Correction of sickle cell disease in transgenic mouse models by gene therapy.
Science
2001
, vol. 
294
 
5550
(pg. 
2368
-
2371
)
95
Imren
 
S
Payen
 
E
Westerman
 
KA
et al. 
Permanent and panerythroid correction of murine beta thalassemia by multiple lentiviral integration in hematopoietic stem cells.
Proc Natl Acad Sci U S A
2002
, vol. 
99
 
22
(pg. 
14380
-
14385
)
96
Ikeda
 
K
Mason
 
PJ
Bessler
 
M
3′UTR-truncated Hmga2 cDNA causes MPN-like hematopoiesis by conferring a clonal growth advantage at the level of HSC in mice.
Blood
2011
, vol. 
117
 
22
(pg. 
5860
-
5869
)
Sign in via your Institution