Next Article in Journal
Usefulness of the Urine Methylation Test (Bladder EpiCheck®) in Follow-Up Patients with Non-Muscle Invasive Bladder Cancer and Cytological Diagnosis of Atypical Urothelial Cells—An Institutional Study
Next Article in Special Issue
Debating the Future of Sickle Cell Disease Curative Therapy: Haploidentical Hematopoietic Stem Cell Transplantation vs. Gene Therapy
Previous Article in Journal
Impact of Cardiopulmonary Resuscitation of Donors on Days Alive and Out of Hospital after Orthotopic Heart Transplantation
Previous Article in Special Issue
Long-Term Health Effects of Curative Therapies on Heart, Lungs, and Kidneys for Individuals with Sickle Cell Disease Compared to Those with Hematologic Malignancies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Across the Myeloablative Spectrum: Hematopoietic Cell Transplant Conditioning Regimens for Pediatric Patients with Sickle Cell Disease

by
Emily Limerick
1,* and
Allistair Abraham
2
1
Cellular and Molecular Therapeutics Branch, National Health, Lung, and Blood Institute, Bethesda, MD 20892, USA
2
Division of Blood and Marrow Transplantation, Children’s National Hospital, Washington, DC 20010, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(13), 3856; https://doi.org/10.3390/jcm11133856
Submission received: 7 May 2022 / Revised: 18 June 2022 / Accepted: 22 June 2022 / Published: 3 July 2022
(This article belongs to the Special Issue The Future of Curative Therapies for Sickle Cell Disease)

Abstract

:
One out of every five hundred African American children in the United States has sickle cell disease (SCD). While multiple disease-modifying therapies are available, hematopoietic cell transplantation (HCT) remains the only curative option for children with SCD. HLA-matched sibling HCT has demonstrated excellent efficacy, but its availability remains limited; alternative donor strategies are increasingly explored. While Busulfan-Cyclophosphamide has become the most widespread conditioning regimen employed in HCT for pediatric SCD, many other regimens have been examined. This review explores different conditioning regimens across the intensity spectrum: from myeloablative to non-myeloablative. We describe survival and organ function outcomes in pediatric SCD patients who have received HCT and discuss the strengths and weaknesses of the various conditioning intensities. Finally, we posit novel directions in allogeneic HCT for SCD.

1. Introduction

Sickle cell disease (SCD) affects over 100,000 people in the US alone. The sequelae may include painful vaso-occlusive crises, chronic pain, acute chest episodes, chronic kidney disease, and cardiomyopathy. SCD is associated with early mortality as well as significant direct and indirect economic costs [1,2,3,4,5]. Improvements in supportive care and disease-modifying therapy have provided new treatment opportunities, but hematopoietic cell transplant (HCT) remains the only potentially curative strategy. As the use of HCT expands, conditioning regimens to meet the needs of SCD patients across the age and morbidity spectrum are necessary. Here, we focus on conditioning strategies employed in pediatric SCD patients.
Operational definitions of conditioning intensity are based on the expected duration of pancytopenia and the need for stem cell support for hematopoietic recovery. The intensity of conditioning regimens can vary significantly. While the conventional conditioning for most young patients with SCD is myeloablative, attempts have been made to limit early transplant toxicity by modifying the intensity of the conditioning regimen. Myeloablative conditioning (MAC) refers to administering total body irradiation (TBI) and/or alkylating agents at doses that will not allow autologous hematologic recovery. While complete myeloablation is likely impossible [6], a practical definition is a combination of agents expected to produce profound pancytopenia and myeloablation within 1–3 weeks of administration; pancytopenia is long-lasting, usually irreversible, and in most cases fatal unless hematopoiesis is restored by hematopoietic stem cell infusion [7].
Non-myeloablative (NMA) conditioning expands HCT availability to older populations and those with significant co-morbidities by minimizing cytopenia and early toxicity through immunoablation; the combination of substantial immunosuppression and larger CD34+ stem cell doses allows donor engraftment [8]. An NMA regimen can therefore be defined as a regimen that causes minimal cytopenia and does not require stem cell support for marrow recovery [7]. Reduced intensity conditioning (RIC) is an intermediate category that differs from MAC and NMA. RIC regimens are associated with cytopenia and require stem cell support. Still, TBI and alkylating agent doses are reduced by at least 30%, so these regimens are defined only by their inability to meet the MAC or NMA definition. For some conditioning regimens, the classification may not be straightforward [7]. For example, some authors believe non-myeloablative regimens may include alkylating agents at a low dose, while others classify such regimens as either myeloablative or reduced intensity. Here, we review myeloablative, non-myeloablative, and reduced intensity conditioning regimens employed in the pediatric SCD setting. Table 1 is adapted from CIBMTR conference proceedings [9] and outlines the conditioning intensity of the regimens most commonly used in this patient population.

2. Myeloablative Conditioning Regimens

The first HCT in a patient with SCD was for the treatment of acute myelogenous leukemia in a pediatric patient [10]. Her myeloablative conditioning, which included cyclophosphamide and TBI, was given for her malignancy, but the matched sibling donor transplant demonstrated the potential for cure of SCD. Subsequently, five children with severe SCD received HLA-matched sibling transplants after conditioning with busulfan and cyclophosphamide with complete cessation of vaso-occlusive episodes and hemolysis [11]. Since those first reported HCT cases, busulfan (BU) and cyclophosphamide (Cy) have been the mainstay of conditioning for HLA-matched HCT for pediatric SCD [12,13,14]. Indeed, a 2017 CIBMTR, European Group for Blood and Marrow Transplantation (EBMT), and Eurocord database investigation reported that 87% of MAC conditioning for pediatric patients was with this regimen [15].
Outcomes for pediatric regimens employing a Bu/Cy conditioning have been excellent: overall survival (OS) rates range from 90 to 100% and event-free survival (EFS) from 77 to 100% [13,16,17,18,19,20,21,22,23,24,25]. On the other hand, the same reports describe grade II-IV aGVHD in 11–39% of patients. The BU/Cy regimen is associated with cGVHD rates ranging from 0 to 21%, though GVHD prevention strategies vary [13,16,17,18,19,20,21,22,23,24,25].
Despite its demonstrated efficacy, this regimen causes significant toxicity, including invasive infections, severe pancytopenia requiring aggressive transfusion support, mucositis, and hepatic sinusoidal obstructive syndrome [26]. Additionally, infertility is a potential risk of BU/Cy, and surveys have indicated that it is a significant concern for patients and families considering HCT [27,28]. Indeed, of the various classes of chemotherapy agents, alkylating agents, such as busulfan, pose the highest risk to fertility and are associated with premature ovarian insufficiency [29]. Further, alkylators are also associated with therapy-related malignancies, specifically acute leukemia and myelodysplasia [30], as well as delayed lung toxicity; BU, in particular, is known for its potential pulmonary effects [31].
BU/Cy dosing strategies have varied to mitigate drug risks and benefits. Early strategies included BU and Cy at different doses, including 200 mg/kg or 260 mg/kg Cy and 14, 16, or >16 mg/kg of BU [32]. Higher BU doses are associated with hepatic sinusoidal obstructive syndrome, seizures, interstitial pneumonitis, and mortality; lower BU is associated with higher rates of graft rejection [33]. Children treated with high-dose BU achieve lower plasma concentrations, measured by area under the plasma–concentration time curve (AUC) or steady-state concentration (Css), than adults [34]. (The Css is a counterpart of AUC and is calculated as the AUC divided by dose frequency.) SCD, on the other hand, does not independently impact BU clearance compared to children without SCD [35]. Targeted busulfan dosing to achieve a total Css of 600–700 ng/mL (14.4–16.8 mg × h/L AUC per dose) has been associated with robust and sustained engraftment in pediatric HCT for SCD. Further, targeting AUC > 900 µMol × min (Css > 615 ng/mL; >14.8 mg × h/L AUC per dose) may contribute to sustained engraftment [24,36,37]. Although better outcomes have been reported with targeted BU therapy, the use of a BU-based conditioning regimen is established only in the setting of MSD transplant. Less than 20% of SCD patients needing an HCT have a matched sibling available. Therefore, alternative donor approaches are critical [38,39].
Studies in leukemia and thalassemia have demonstrated that substituting fludarabine for cyclophosphamide in MAC improves safety and retains efficacy [40,41]. Therefore, to establish less toxic conditioning strategies in SCD conditioning, fludarabine (flu) has been added to BU/cy to reduce busulfan and cyclophosphamide doses [20]; it has also been proposed as an alternative to cyclophosphamide [42,43,44,45]. Fludarabine is highly immunosuppressive and relatively safe [45]. BU/Flu has been employed in 7.5% of pediatric SCD MAC conditioning regimens [15]. BU/Flu has been employed in the HLA-matched setting (MSD and MUD) [42,44,45], as well as haploidentical HCT [43]. Outcomes of Bu/Flu conditioning vary. While overall survival exceeds 90%, Krishnamurti et al.’s multicenter pilot study of adolescents and young adults reports 60% 3-year EFS among MUD transplants compared to 88% for MSD [42]. Exclusively pediatric cohorts have reported 2-year OS and EFS of 100% with the BU/Flu regimen in the MSD setting [45] and 75% EFS in the MUD context [44]. Pawlowska describes early results in a small cohort of adolescents and young adults; they achieved 100% OS and EFS with up to 11 months of follow-up [43]. GVHD rates are similarly variable: no acute or chronic GVHD was reported in the MUD patients. While no haplo patients developed aGVHD, 75% developed cGVHD, primarily mild and limited. aGVHD was noted in 18% of MSD and 27% developed cGVHD [42,43,44,45].
Treosulfan (treo) is a structural analog of BU with significant myeloablative potential, immunosuppressive activity, and minimal extramedullary effects [46]. It has been employed in HCT for malignant and non-malignant diseases with low toxicity and demonstrated efficacy [47,48]. Therefore, another attempt to reduce toxicity in patients with SCD within the myeloablative framework includes conditioning with flu, thiotepa (thio), and treo [19,49,50,51,52]. An analysis of unrelated donor HCT for SCD reported that 3-year OS in patients treated with flu/thio/treo conditioning was higher than in patients treated with other regimens. However, the 3-year GVHD-free, relapse-free survival (GRFS) was not significantly different [19]. Others have reported excellent outcomes even in the alternative donor setting with 100% primary engraftment and OS and EFS 90% and higher [49,50,52]. Of note, Marzollo and colleagues describe a cohort of 11 children transplanted after flu/thio/treo. Donor sources included MSD, haplo, MUD, and MMUD. All patients engrafted, and there were no instances of secondary graft failure. They detail stable mixed chimerism in a large proportion of patients [50]. While treo has been approved for use in Canada and Europe, the FDA has not yet approved its use in the USA [53].

3. Reduced Intensity and Non-Myeloablative Conditioning Regimens

Despite these efforts to reduce toxicity, MAC remains a significant risk and is often prohibitively damaging, particularly in older adults—registry data have demonstrated that 5-year survival is 80% in patients >16 years compared to 95% in those <16 years—and those with significant comorbidities [15]. Thus, reduced intensity and non-myeloablative conditioning regimens have been employed with various transplant types, including matched sibling and matched unrelated donor, mismatch unrelated, and haploidentical HCT. Understanding that complete donor chimerism is unnecessary for SCD symptoms to resolve has allowed expansion of alternative donor strategies coupled with less intense conditioning regimens. Mixed chimerism occurs after allogeneic HCT when the lymphohematopoietic system comprises both donor and recipient-derived blood cells [54]. Stable mixed chimerism was also described in Strocchio’s analysis; they hypothesize that the tolerance achieved in these patients may indeed also be associated with lower GVHD rates [51], a concept previously demonstrated in thalassemia patients [55]. Younger age, use of ATG, and cord blood vs. bone marrow sources have been associated with mixed chimerism [56]. Minimum donor myeloid chimerism of 20–25% has been reported to reverse the SCD phenotype [57,58,59]. As alternative donor approaches are necessary to improve the less than 10% availability of matched sibling transplants, the role of RIC and NMA conditioning has increased [60].

3.1. Reduced Intensity Conditioning

Fludarabine has been employed as part of reduced intensity conditioning regimens with varying success [17,61,62,63,64,65,66,67]. Two of three haploidentical patients conditioned with an RIC regimen of fludarabine, thiotepa, and busulfan with anti-thymocyte globulin (ATG) and muromonomab-CD3 experienced rejection [17]. A small cohort of children with SCD received a mismatch unrelated donor transplant after RIC with flu, melphalan (mel), alem, and an infusion of marrow-derived mesenchymal stem cells to facilitate engraftment; all patients either died or had autologous recovery [67]. The Sickle Cell Unrelated Donor Transplant (SCURT) trial enrolled 29 patients conditioned with flu, mel, alem [65]. Although the 1-year EFS was 76%, high rates of GVHD and GVHD-related deaths marred the study. Further, the cord blood arm of SCURT stopped early due to high rates of graft rejection [62]. Subsequent work, however, has demonstrated that adding hydroxyurea and thiotepa to the fludarabine/melphalan regimen can improve engraftment after cord blood transplants [61]. American Society of Hematology (ASH) guidelines for stem cell transplantation in SCD recommend using myeloablative conditioning over RIC with flu/mel in pediatric patients with a matched sibling donor available [68].
A small pediatric cohort transplanted with a reduced intensity bu/flu/ATG/total lymphocyte irradiation regimen experienced 86% EFS [69]. Similar RIC regimens in the MSD, unrelated donor and haplo settings have been associated with >90% overall and event-free survival [63,64,66,70]. The ideal RIC regimen that maximizes engraftment and minimizes toxicity is unknown [71].

3.2. Non-Myeloablative Conditioning

The NIH platform is a non-myeloablative regimen used in MSD HCT and is aimed at tolerance induction; it includes low-dose TBI and alemtuzumab and has demonstrated efficacy in children and adults [8,72,73,74]. Early non-myeloablative conditioning strategies in the haploidentical setting employed fludarabine and low-dose TBI with meager results: most patients’ engraftment was only transient [75,76]. The Sickle transplant Using a Nonmyeloablative approach (SUN) multicenter clinical trial (NCT03587272) employs the NIH regimen and is ongoing [77]. Interim results report EFS of 87.5% and 100% OS. Likewise, Johns Hopkins researchers have described their experience in young adults with non-myeloablative haploidentical HCT using ATG, flu, cy, TBI, and subsequently an RIC version that added thiotepa [78]. Their cohort, whose median age was 22, experienced 100% OS and 93% EFS. The excellent results of these low-intensity approaches prompted Nickel et al. to suggest reserving myeloablation for a second transplant in the minority of patients who experience graft failure [77]. The ASH SCD stem cell transplant guidelines suggest NMA conditioning over RIC, specifically flu/mel-containing regimens, for adults with SCD who have a transplant indication [79].

3.3. T-Cell Depletion Strategies

Authors have hypothesized that exposure to minor histocompatibility antigens through multiple blood transfusions may increase the likelihood of immunologic rejection in SCD patients. ATG, Alemtuzumab, and other T-cell depletion strategies have been employed to mitigate that risk [14,76]. Indeed, in 87 young SCD patients who received an MAC regimen, the addition of ATG was associated with a reduction of the rejection rate from 23% to 3% [13]. The timing of t-cell depletion in an RIC regimen, early vs. late, may impact engraftment. Specifically, ‘early’ alemtuzumab may be associated with improved engraftment, though may carry the risk of increased acute and chronic GVHD [80].

4. NMA vs. RIC vs. MAC Pro/Cons

Direct comparison of the outcomes of different conditioning regimens is limited. A transplant database analysis reported that the OS and EFS were highest after NMA conditioning with no significant differences between MAC and RIC [81]. Acute and chronic GVHD rates were higher with MAC and RIC than NMA regimens. However, in analyses limited to MSD transplants, though some have reported that graft failure was higher after RIC [81], multiple reports conclude that conditioning regimen intensity is not associated with overall survival [15,81]. Mixed chimerism (MC) has been reported after NMA, RIC, and MAC, though the incidence varies with different MC definitions. Although it is more common with RIC and NMA preparations, a considerable number (29%) of patients with MC have been described after MAC as well [82]. CD34+ stem cell boosts have been successfully employed in the setting of falling chimerism and return of SCD after RIC and NMA conditioning [83,84]. Finally, although the risk of developing a malignancy post-transplant was seven times higher with NMA versus RIC, this did not reach statistical significance; there were no cases of malignant neoplasm after myeloablative conditioning [81].
Additional limitations of direct comparisons include differences in follow-up duration. For example, the median follow-up duration for the predominantly MAC pediatric MSD studies described in a 2017 international registry study was 56.4 months [15]. A 2019 registry study describes median follow-up of surviving patients: MSD, whose predominant conditioning was myeloablative, had 36 months of follow-up, while haploidentical, the majority of whom had NMA conditioning, had 25 months of follow-up [81]. Further, age is a significant variable that differs between donor type and conditioning regimen. MSD patients, for whom MAC predominates, tend to be younger than the predominantly NMA haploidentical recipients [81]. Data have shown that a younger age of transplant is associated with improved outcomes [15]. Table 2 summarizes the available data comparing relative outcomes of different regimen intensities.

5. Symptomatic/Phenotypic Improvements after HCT

While all regimen intensities have been associated with reversal of the SCD phenotype and resolution of SCD symptoms [17,18,85,86,87], their late effects on other organ systems are of tremendous import.
Reproductive: Most girls require hormone replacement after myeloablative HCT [16]. Girls who were significantly younger at the time of transplant were more likely to have spontaneous puberty. Of 20 women >25 years old, 4 women had six spontaneous pregnancies; they were all transplanted at 5–7 years of age [16]. Others have also reported spontaneous puberty and pregnancies in some girls after myeloablative conditioning with busulfan, including a healthy pregnancy after spontaneous resolution of post-HCT primary ovarian insufficiency [18]. Fertility preservation may be offered before HCT, but potential risks include painful crises and even respiratory failure [88,89]. Mishkin et al. also argue that the psychiatric burden of SCD, HCT toxicities, and resultant infertility may be cumulative [90]. Reproductive system effects have been reported after RIC as well. Three adolescent males all developed oligospermia or azoospermia after RIC [91]. Even in the RIC setting, amenorrhea is noted in patients who are post-pubertal at the time of transplant and spontaneous menarche in patients who are younger in age at the time of transplant [69]. Spontaneous pregnancies have also been reported in men and women after non-myeloablative HCT for SCD in adolescents and adults [8].
Spleen and kidney: Improvement in splenic function has been described with the disappearance of Howell–Jolly bodies post-HCT [13]. Stable renal function after NMA HCT has also been reported [8,92].

6. Toward the Future

Excellent results have been achieved with matched sibling donor transplants. Clinical trials are ongoing to determine the best approaches to optimize efficacy and minimize toxicity, especially using alternative donor strategies, which are necessary to expand transplant availability. Pre-transplant immunosuppression has been employed to suppress endogenous hematopoiesis, overcome the engraftment barrier, and improve graft failure rates in the haploidentical setting [43,93]. Strategies include pentostatin and cyclophosphamide (NCT# 03077542), dexamethasone and fludarabine [43], and hydroxyurea and azathioprine [94]. Granulocyte stimulants like gCSF are also being employed safely in the post-HCT to enhance neutrophil recovery [95]. Finally, new conditioning strategies that avoid genotoxicity altogether represent the next wave of HCT development. Anti-CD117 and anti-CD45 antibodies have been conjugated with immunotoxins and deployed in experimental models to effectively condition mice without concomitant cytopenias [96,97]. Similarly, chimeric antigen receptor t-cells targeted against CD117 have also been employed in murine models to condition effectively without chemotherapy or radiation [98].

7. Conclusions

Myeloablation with Bu-Cy has established efficacy and has become the standard of care in the pediatric MSD setting, but at the cost of significant toxicity and limited tolerability for only younger patients with fewer comorbidities. Reduced intensity and nonmyeloablative strategies may rival myeloablative ones in terms of event-free and overall survival, though clinical trials to assess the effectiveness of specific regimens and conditioning intensity are required. In the meantime, choosing a regimen based on the available donor strategy—particularly for SCD patients requiring an alternative option—demonstrated outcomes, and anticipated toxicity, typically in the clinical trial setting.

Author Contributions

E.L. and A.A. conceptualized the review, reviewed the literature, drafted the article and performed critical revisions. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding

Conflicts of Interest

The authors declare no conflict of interest

References

  1. Lee, S.; Vania, D.K.; Bhor, M.; Revicki, D.; Abogunrin, S.; Sarri, G. Patient-Reported Outcomes and Economic Burden of Adults with Sickle Cell Disease in the United States: A Systematic Review. Int. J. Gen. Med. 2020, 13, 361–377. [Google Scholar] [CrossRef] [PubMed]
  2. Holdford, D.; Vendetti, N.; Sop, D.M.; Johnson, S.; Smith, W.R. Indirect Economic Burden of Sickle Cell Disease. Value Health 2021, 24, 1095–1101. [Google Scholar] [CrossRef] [PubMed]
  3. Gladwin, M.T.; Barst, R.J.; Gibbs, J.S.R.; Hildesheim, M.; Sachdev, V.; Nouraie, M.; Hassell, K.L.; Little, J.A.; Schraufnagel, D.E.; Krishnamurti, L.; et al. Risk factors for death in 632 patients with sickle cell disease in the United States and United Kingdom. PLoS ONE 2014, 9, e99489. [Google Scholar] [CrossRef] [Green Version]
  4. Maitra, P.; Caughey, M.; Robinson, L.; Desai, P.C.; Jones, S.; Nouraie, M.; Gladwin, M.T.; Hinderliter, A.; Cai, J.; Ataga, K.I. Risk factors for mortality in adult patients with sickle cell disease: A meta-analysis of studies in North America and Europe. Haematologica 2017, 102, 626–636. [Google Scholar] [CrossRef] [PubMed]
  5. Payne, A.B.; Mehal, J.M.; Chapman, C.; Haberling, D.L.; Richardson, L.C.; Bean, C.J.; Hooper, W.C. Trends in Sickle Cell Disease-Related Mortality in the United States, 1979 to 2017. Ann. Emerg. Med. 2020, 76, S28–S36. [Google Scholar] [CrossRef]
  6. Fox, B.W.; Lajtha, L.G. Radiation damage and repair phenomena. Br. Med. Bull. 1973, 29, 16–22. [Google Scholar] [CrossRef] [Green Version]
  7. Bacigalupo, A.; Ballen, K.; Rizzo, D.; Giralt, S.; Lazarus, H.; Ho, V.; Apperley, J.; Slavin, S.; Pasquini, M.; Sandmaier, B.M.; et al. Defining the intensity of conditioning regimens: Working definitions. Biol. Blood Marrow Transpl. 2009, 15, 1628–1633. [Google Scholar] [CrossRef] [Green Version]
  8. Hsieh, M.M.; Fitzhugh, C.D.; Weitzel, R.P.; Link, M.E.; Coles, W.A.; Zhao, X.; Rodgers, G.P.; Powell, J.D.; Tisdale, J.F. Nonmyeloablative HLA-matched sibling allogeneic hematopoietic stem cell transplantation for severe sickle cell phenotype. JAMA 2014, 312, 48–56. [Google Scholar] [CrossRef]
  9. Chhabra, S. Conditioning Intensity. 2021. Available online: https://www.cibmtr.org/Meetings/Materials/CRPDMC/Docments/2021/09%20-Conditioning%20Intensity.pdf (accessed on 6 January 2022).
  10. Johnson, F.L.; Look, A.T.; Gockerman, J.; Ruggiero, M.R.; Dalla-Pozza, L.; Billings, F.T., 3rd. Bone-marrow transplantation in a patient with sickle-cell anemia. N. Engl. J. Med. 1984, 311, 780–783. [Google Scholar] [CrossRef]
  11. Vermylen, C.; Fernandez Robles, E.; Ninane, J.; Cornu, G. Bone marrow transplantation in five children with sickle cell anaemia. Lancet 1988, 1, 1427–1428. [Google Scholar] [CrossRef]
  12. Vermylen, C.; Cornu, G.; Ferster, A.; Brichard, B.; Ninane, J.; Ferrant, A.; Zenebergh, A.; Maes, P.; Dhooge, C.; Benoit, Y.; et al. Haematopoietic stem cell transplantation for sickle cell anaemia: The first 50 patients transplanted in Belgium. Bone Marrow Transpl. 1998, 22, 1701291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Bernaudin, F.; Socie, G.; Kuentz, M.; Chevret, S.; Duval, M.; Bertrand, Y.; Vannier, J.-P.; Yakouben, K.; Thuret, I.; Bordigoni, P.; et al. Long-term results of related myeloablative stem-cell transplantation to cure sickle cell disease. Blood 2007, 110, 2749–2756. [Google Scholar] [CrossRef] [PubMed]
  14. Walters, M.C.; Storb, R.; Patience, M.; Leisenring, W.; Taylor, T.; Sanders, J.E.; Buchanan, G.E.; Rogers, Z.R.; Dinndorf, P.; Davies, S.C.; et al. Impact of bone marrow transplantation for symptomatic sickle cell disease: An interim report. Multicenter investigation of bone marrow transplantation for sickle cell disease. Blood 2000, 95, 1918–1924. [Google Scholar] [PubMed]
  15. Gluckman, E.; Cappelli, B.; Bernaudin, F.; Labopin, M.; Volt, F.; Carreras, J.; Simões, B.P.; Ferster, A.; Dupont, S.; de la Fuente, J.; et al. Sickle cell disease: An international survey of results of HLA-identical sibling hematopoietic stem cell transplantation. Blood 2017, 129, 1548–1556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Bernaudin, F.; Dalle, J.-H.; Bories, D.; De Latour, R.P.; Robin, M.; Bertrand, Y.; Pondarre, C.; Vannier, J.-P.; Neven, B.; Kuentz, M.; et al. Long-term event-free survival, chimerism and fertility outcomes in 234 patients with sickle-cell anemia younger than 30 years after myeloablative conditioning and matched-sibling transplantation in France. Haematologica 2020, 105, 91–101. [Google Scholar] [CrossRef]
  17. Dallas, M.H.; Triplett, B.; Shook, D.R.; Hartford, C.; Srinivasan, A.; Laver, J.; Ware, R.; Leung, W. Long-term outcome and evaluation of organ function in pediatric patients undergoing haploidentical and matched related hematopoietic cell transplantation for sickle cell disease. Biol. Blood Marrow Transpl. 2013, 19, 820–830. [Google Scholar] [CrossRef] [Green Version]
  18. Dedeken, L.; Lê, P.Q.; Azzi, N.; Brachet, C.; Heijmans, C.; Huybrechts, S.; Devalck, C.; Rozen, L.; Ngalula, M.; Ferster, A. Haematopoietic stem cell transplantation for severe sickle cell disease in childhood: A single centre experience of 50 patients. Br. J. Haematol. 2014, 165, 402–408. [Google Scholar] [CrossRef]
  19. Gluckman, E.; Fuente, J.; Cappelli, B.; Scigliuolo, G.; Volt, F.; Tozatto-Maio, K.; Rocha, V.; Tommaso, M.; O’Boyle, F.; Smiers, F.; et al. The role of HLA matching in unrelated donor hematopoietic stem cell transplantation for sickle cell disease in Europe. Bone Marrow Transpl. 2020, 55, 1946–1954. [Google Scholar] [CrossRef]
  20. Horan, J.T.; Haight, A.; Dioguardi, J.L.; Brown, C.; Grizzle, A.; Shelman, C.; Kanter, J.; Hale, G.; Nieder, M.; Benton, M.; et al. Using fludarabine to reduce exposure to alkylating agents in children with sickle cell disease receiving busulfan, cyclophosphamide, and antithymocyte globulin transplant conditioning: Results of a dose de-escalation trial. Biol. Blood Marrow Transpl. 2015, 21, 900–905. [Google Scholar] [CrossRef] [Green Version]
  21. Isgrò, A.; Paciaroni, K.; Gaziev, J.; Sodani, P.; Gallucci, C.; Marziali, M.; De Angelis, G.; Alfieri, C.; Ribersani, M.; Roveda, A.; et al. Haematopoietic stem cell transplantation in Nigerian sickle cell anaemia children patients. Niger. Med. J. 2015, 56, 175–179. [Google Scholar] [CrossRef] [Green Version]
  22. Kogel, F.; Hakimeh, D.; Sodani, P.; Lang, P.; Kühl, J.; Hundsdoerfer, P.; Künkele, A.; Eggert, A.; Oevermann, L.; Schulte, J.H. Allogeneic hematopoietic stem cell transplantation from sibling and unrelated donors in pediatric patients with sickle cell disease-A single center experience. Pediatr. Transpl. 2021, 25, e13892. [Google Scholar] [CrossRef] [PubMed]
  23. Locatelli, F.; Rocha, V.; Reed, W.; Bernaudin, F.; Ertem, M.; Grafakos, S.; Brichard, B.; Li, X.; Nagler, A.; Giorgiani, G.; et al. Related umbilical cord blood transplantation in patients with thalassemia and sickle cell disease. Blood 2003, 101, 2137–2143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Maheshwari, S.; Kassim, A.; Yeh, R.F.; Domm, J.; Calder, C.; Evans, M.J.; Manes, B.; Bruce, K.; Brown, V.; Ho, R.; et al. Targeted Busulfan therapy with a steady-state concentration of 600–700 ng/mL in patients with sickle cell disease receiving HLA-identical sibling bone marrow transplant. Bone Marrow Transpl. 2014, 49, 366–369. [Google Scholar] [CrossRef] [PubMed]
  25. Majumdar, S.; Robertson, Z.; Robinson, A.; Starnes, S.; Iyer, R.; Megason, G. Outcome of hematopoietic cell transplantation in children with sickle cell disease, a single center’s experience. Bone Marrow Transpl. 2010, 45, 895–900. [Google Scholar] [CrossRef] [Green Version]
  26. Diaconescu, R.; Flowers, C.R.; Storer, B.; Sorror, M.L.; Maris, M.B.; Maloney, D.G.; Sandmaier, B.M.; Storb, R. Morbidity and mortality with nonmyeloablative compared with myeloablative conditioning before hematopoietic cell transplantation from HLA-matched related donors. Blood 2004, 104, 1550–1558. [Google Scholar] [CrossRef]
  27. Brachet, C.; Heinrichs, C.; Tenoutasse, S.; Devalck, C.; Azzi, N.; Ferster, A. Children with sickle cell disease: Growth and gonadal function after hematopoietic stem cell transplantation. J. Pediatr. Hematol. Oncol. 2007, 29, 445–450. [Google Scholar] [CrossRef]
  28. Thompson, A.L.; Ma, A.B.; Twohy, E.; Pa, J.D.; Sande, J.; Hsu, L.L.; Kamani, N.; Meier, E.R. An educational symposium for patients with sickle cell disease and their families: Results from surveys of knowledge and factors influencing decisions about hematopoietic stem cell transplant. Pediatr. Blood Cancer 2013, 60, 1946–1951. [Google Scholar] [CrossRef]
  29. Meirow, D.; Biederman, H.; Anderson, R.A.; Wallace, W.H. Toxicity of chemotherapy and radiation on female reproduction. Clin. Obstet. Gynecol. 2010, 53, 727–739. [Google Scholar] [CrossRef]
  30. Bomken, S.; Skinner, R. Secondary Malignant Neoplasms Following Haematopoietic Stem Cell Transplantation in Childhood. Children 2015, 2, 146–173. [Google Scholar] [CrossRef]
  31. Matijasic, N.; Bonevski, A.; Tokic Pivac, V.; Pavic, I. Busulfan-Induced Lung Injury in Pediatric Oncology Patients-Review of the Literature with an Illustrative Case. Pediatr. Allergy Immunol. Pulmonol. 2019, 32, 86–91. [Google Scholar] [CrossRef]
  32. Bernaudin, F.; Souillet, G.; Vannier, J.P.; Plouvier, E.; Lemerle, S.; Michel, G.; Bordigoni, P.; Lutz, P.; Kuentz, M. Bone marrow transplantation (BMT) in 14 children with severe sickle cell disease (SCD): The French experience. GEGMO. Bone Marrow Transpl. 1993, 12 (Suppl. S1), 118–121. [Google Scholar]
  33. Bolinger, A.M.; Zangwill, A.B.; Slattery, J.T.; Glidden, D.; DeSantes, K.; Heyn, L.; Risler, L.J.; Bostrom, B.; Cowan, M.J. An evaluation of engraftment, toxicity and busulfan concentration in children receiving bone marrow transplantation for leukemia or genetic disease. Bone Marrow Transpl. 2000, 25, 925–930. [Google Scholar] [CrossRef] [PubMed]
  34. Grochow, L.B.; Krivit, W.; Whitley, C.B.; Blazar, B. Busulfan disposition in children. Blood 1990, 75, 1723–1727. [Google Scholar] [CrossRef] [PubMed]
  35. Remy, A.; Théorêt, Y.; Ansari, M.; Bittencourt, H.; Ducruet, T.; Nava, T.; Pastore, Y.; Rezgui, M.A.; Krajinovic, M.; Kleiber, N. Is Busulfan Clearance Different in Patients with Sickle Cell Disease? Let’s Clear Up That Case with Some Controls. J. Pediatr. Hematol. Oncol. 2021, 43, e867–e872. [Google Scholar] [CrossRef]
  36. McPherson, M.E.; Hutcherson, D.; Olson, E.; Haight, A.E.; Horan, J.; Chiang, K.Y. Safety and efficacy of targeted busulfan therapy in children undergoing myeloablative matched sibling donor BMT for sickle cell disease. Bone Marrow Transpl. 2011, 46, 27–33. [Google Scholar] [CrossRef] [Green Version]
  37. Gaziev, J.; Isgrò, A.; Mozzi, A.F.; Petain, A.; Nguyen, L.; Ialongo, C.; Dinallo, V.; Sodani, P.; Marziali, M.; Andreani, M.; et al. New insights into the pharmacokinetics of intravenous busulfan in children with sickle cell anemia undergoing bone marrow transplantation. Pediatr. Blood Cancer 2015, 62, 680–686. [Google Scholar] [CrossRef]
  38. Walters, M.C.; Patience, M.; Leisenring, W.; Eckman, J.R.; Buchanan, G.R.; Rogers, Z.R.; E Olivieri, N.; Vichinsky, E.; Davies, S.C.; Mentzer, W.C.; et al. Barriers to bone marrow transplantation for sickle cell anemia. Biol. Blood Marrow Transpl. 1996, 2, 100–104. [Google Scholar]
  39. Alfraih, F.; Aljurf, M.; Fitzhugh, C.D.; Kassim, A.A. Alternative donor allogeneic hematopoietic cell transplantation for hemoglobinopathies. Semin. Hematol. 2016, 53, 120–128. [Google Scholar] [CrossRef]
  40. Andersson, B.S.; de Lima, M.; Thall, P.F.; Wang, X.; Couriel, D.; Korbling, M.; Roberson, S.; Giralt, S.; Pierre, B.; Russell, J.A.; et al. Once daily i.v. busulfan and fludarabine (i.v. Bu-Flu) compares favorably with i.v. busulfan and cyclophosphamide (i.v. BuCy2) as pretransplant conditioning therapy in AML/MDS. Biol. Blood Marrow Transpl. 2008, 14, 672–684. [Google Scholar] [CrossRef] [Green Version]
  41. La Nasa, G.; Littera, R.; Locatelli, F.; Lai, S.; Alba, F.; Caocci, G.; Lisini, D.; Nesci, S.; Vacca, A.; Piras, E.; et al. The human leucocyte antigen-G 14-basepair polymorphism correlates with graft-versus-host disease in unrelated bone marrow transplantation for thalassaemia. Br. J. Haematol. 2007, 139, 284–288. [Google Scholar] [CrossRef]
  42. Krishnamurti, L.; Neuberg, D.S.; Sullivan, K.M.; Kamani, N.R.; Abraham, A.; Campigotto, F.; Zhang, W.; Dahdoul, T.; De Castro, L.; Parikh, S.; et al. Bone marrow transplantation for adolescents and young adults with sickle cell disease: Results of a prospective multicenter pilot study. Am. J. Hematol. 2019, 94, 446–454. [Google Scholar] [CrossRef] [Green Version]
  43. Pawlowska, A.B.; Cheng, J.C.; Karras, N.A.; Sun, W.; Wang, L.D.; Bell, A.D.; Gutierrez, L.; Rosenthal, J. HLA Haploidentical Stem Cell Transplant with Pretransplant Immunosuppression for Patients with Sickle Cell Disease. Biol. Blood Marrow Transpl. 2018, 24, 185–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Rangarajan, H.G.; Abu-Arja, R.; Pai, V.; Guilcher, G.M.T.; Soni, S. Outcomes of Unrelated Donor Stem Cell Transplantion with Post-Transplant Cyclophosphamide for Graft-versus-Host Disease Prophylaxis in Patients with Severe Sickle Cell Disease. Biol. Blood Marrow Transpl. 2018, 24, 413–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Bhatia, M.; Jin, Z.; Baker, C.; Geyer, M.B.; Radhakrishnan, K.; Morris, E.; Satwani, P.; George, D.; Garvin, J.; Del Toro, G.; et al. Reduced toxicity, myeloablative conditioning with BU, fludarabine, alemtuzumab and SCT from sibling donors in children with sickle cell disease. Bone Marrow Transpl. 2014, 49, 913–920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Sjoo, F.; Hassan, Z.; Abedi-Valugerdi, M.; Griskevicius, L.; Nilsson, C.; Remberger, M.; Aschan, J.; Concha, H.; Gaughan, U.; Hassan, M. Myeloablative and immunosuppressive properties of treosulfan in mice. Exp. Hematol. 2006, 34, 115–121. [Google Scholar] [CrossRef] [Green Version]
  47. Slatter, M.A.; Boztug, H.; Potschger, U.; Sykora, K.-W.; Lankester, A.; Yaniv, I.; Sedlacek, P.; Glogova, E.; Veys, P.; Gennery, A.R.; et al. Treosulfan-based conditioning regimens for allogeneic haematopoietic stem cell transplantation in children with non-malignant diseases. Bone Marrow Transpl. 2015, 50, 1536–1541. [Google Scholar] [CrossRef] [Green Version]
  48. Boztug, H.; Sykora, K.W.; Slatter, M.; Zecca, M.; Veys, P.; Lankester, A.; Cant, A.; Skinner, R.; Wachowiak, J.; Glogova, E.; et al. European Society for Blood and Marrow Transplantation Analysis of Treosulfan Conditioning Before Hematopoietic Stem Cell Transplantation in Children and Adolescents with Hematological Malignancies. Pediatr. Blood Cancer 2016, 63, 139–148. [Google Scholar] [CrossRef] [Green Version]
  49. Foell, J.; Schulte, J.H.; Pfirstinger, B.; Troeger, A.; Wolff, D.; Edinger, M.; Hofmann, P.; Aslanidis, C.; Lang, P.; Holler, E.; et al. Haploidentical CD3 or alpha/beta T-cell depleted HSCT in advanced stage sickle cell disease. Bone Marrow Transpl. 2019, 54, 1859–1867. [Google Scholar] [CrossRef]
  50. Marzollo, A.; Calore, E.; Tumino, M.; Pillon, M.; Gazzola, M.V.; Destro, R.; Colombatti, R.; Marson, P.; Tison, T.; Colpo, A.; et al. Treosulfan-Based Conditioning Regimen in Sibling and Alternative Donor Hematopoietic Stem Cell Transplantation for Children with Sickle Cell Disease. Mediterr. J. Hematol. Infect. Dis. 2017, 9, e2017014. [Google Scholar] [CrossRef] [Green Version]
  51. Strocchio, L.; Zecca, M.; Comoli, P.; Mina, T.; Giorgiani, G.; Giraldi, E.; Vinti, L.; Merli, P.; Regazzi, M.; Locatelli, F. Treosulfan-based conditioning regimen for allogeneic haematopoietic stem cell transplantation in children with sickle cell disease. Br. J. Haematol. 2015, 169, 726–736. [Google Scholar] [CrossRef]
  52. Wiebking, V.; Hutker, S.; Schmid, I.; Immler, S.; Feuchtinger, T.; Albert, M.H. Reduced toxicity, myeloablative HLA-haploidentical hematopoietic stem cell transplantation with post-transplantation cyclophosphamide for sickle cell disease. Ann. Hematol. 2017, 96, 1373–1377. [Google Scholar] [CrossRef] [PubMed]
  53. Terry, M. FDA Hands Out Surprising Rejection of Meduxus and Medac’s Treosulfan. BioSpace, 3 August 2021. [Google Scholar]
  54. Sachs, D.H.; Kawai, T.; Sykes, M. Induction of tolerance through mixed chimerism. Cold Spring Harb. Perspect Med. 2014, 4, a015529. [Google Scholar] [CrossRef] [PubMed]
  55. Lisini, D.; Zecca, M.; Giorgiani, G.; Montagna, D.; Cristantielli, R.; Labirio, M.; Grignani, P.; Previderè, C.; Di Cesare-Merlone, A.; Amendola, G.; et al. Donor/recipient mixed chimerism does not predict graft failure in children with beta-thalassemia given an allogeneic cord blood transplant from an HLA-identical sibling. Haematologica 2008, 93, 1859–1867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Guilcher, G.M.T.; Truong, T.H.; Saraf, S.L.; Joseph, J.J.; Rondelli, D.; Hsieh, M.M. Curative therapies: Allogeneic hematopoietic cell transplantation from matched related donors using myeloablative, reduced intensity, and nonmyeloablative conditioning in sickle cell disease. Semin. Hematol. 2018, 55, 87–93. [Google Scholar] [CrossRef]
  57. Fitzhugh, C.D.; Cordes, S.; Taylor, T.; Coles, W.; Roskom, K.; Link, M.; Hsieh, M.M.; Tisdale, J.F. At least 20% donor myeloid chimerism is necessary to reverse the sickle phenotype after allogeneic HSCT. Blood 2017, 130, 1946–1948. [Google Scholar] [CrossRef] [Green Version]
  58. Abraham, A.; Hsieh, M.; Eapen, M.; Fitzhugh, C.; Carreras, J.; Keesler, D.; Guilcher, G.; Kamani, N.; Walters, M.C.; Boelens, J.J.; et al. Relationship between Mixed Donor-Recipient Chimerism and Disease Recurrence after Hematopoietic Cell Transplantation for Sickle Cell Disease. Biol. Blood Marrow Transpl. 2017, 23, 2178–2183. [Google Scholar] [CrossRef] [Green Version]
  59. Walters, M.C.; Patience, M.; Leisenring, W.; Rogers, Z.R.; Aquino, V.M.; Buchanan, G.R.; Roberts, I.A.; Yeager, A.M.; Hsu, L.; Adamkiewicz, T.; et al. Stable mixed hematopoietic chimerism after bone marrow transplantation for sickle cell anemia. Biol. Blood Marrow Transpl. 2001, 7, 665–673. [Google Scholar] [CrossRef] [Green Version]
  60. Hansbury, E.N.; Schultz, W.H.; Ware, R.E.; Aygun, B. Bone marrow transplant options and preferences in a sickle cell anemia cohort on chronic transfusions. Pediatr. Blood Cancer 2012, 58, 611–615. [Google Scholar] [CrossRef]
  61. Abraham, A.; Cluster, A.; Jacobsohn, D.; Delgado, D.; Hulbert, M.L.; Kukadiya, D.; Murray, L.; Shenoy, S. Unrelated Umbilical Cord Blood Transplantation for Sickle Cell Disease Following Reduced-Intensity Conditioning: Results of a Phase I Trial. Biol. Blood Marrow Transpl. 2017, 23, 1587–1592. [Google Scholar] [CrossRef] [Green Version]
  62. Kamani, N.R.; Walters, M.C.; Carter, S.; Aquino, V.; Brochstein, J.A.; Chaudhury, S.; Eapen, M.; Freed, B.M.; Grimley, M.; Levine, J.E.; et al. Unrelated donor cord blood transplantation for children with severe sickle cell disease: Results of one cohort from the phase II study from the Blood and Marrow Transplant Clinical Trials Network (BMT CTN). Biol. Blood Marrow Transpl. 2012, 18, 1265–1272. [Google Scholar] [CrossRef] [Green Version]
  63. King, A.A.; Kamani, N.; Bunin, N.; Sahdev, I.; Brochstein, J.; Hayashi, R.J.; Grimley, M.; Abraham, A.; Dioguardi, J.; Chan, K.W.; et al. Successful matched sibling donor marrow transplantation following reduced intensity conditioning in children with hemoglobinopathies. Am. J. Hematol. 2015, 90, 1093–1098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Ngwube, A.; Shah, N.; Godder, K.; Jacobsohn, D.; Hulbert, M.L.; Shenoy, S. Abatacept is effective as GVHD prophylaxis in unrelated donor stem cell transplantation for children with severe sickle cell disease. Blood Adv. 2020, 4, 3894–3899. [Google Scholar] [CrossRef] [PubMed]
  65. Shenoy, S.; Eapen, M.; Panepinto, J.A.; Logan, B.R.; Wu, J.; Abraham, A.; Brochstein, J.; Chaudhury, S.; Godder, K.; Haight, A.E.; et al. A trial of unrelated donor marrow transplantation for children with severe sickle cell disease. Blood 2016, 128, 2561–2567. [Google Scholar] [CrossRef] [PubMed]
  66. Gilman, A.L.; Eckrich, M.J.; Epstein, S.; Barnhart, C.; Cannon, M.; Fukes, T.; Hyland, M.; Shah, K.; Grochowski, D.; Champion, E.; et al. Alternative donor hematopoietic stem cell transplantation for sickle cell disease. Blood Adv. 2017, 1, 1215–1223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Kharbanda, S.; Smith, A.R.; Hutchinson, S.K.; McKenna, D.H.; Ball, J.B.; Lamb, L.S., Jr.; Agarwal, R.; Weinberg, K.I.; Wagner, J.E., Jr. Unrelated donor allogeneic hematopoietic stem cell transplantation for patients with hemoglobinopathies using a reduced-intensity conditioning regimen and third-party mesenchymal stromal cells. Biol. Blood Marrow Transpl. 2014, 20, 581–586. [Google Scholar] [CrossRef] [Green Version]
  68. Kanter, J.; Liem, R.I.; Bernaudin, F.; Bolaños-Meade, J.; Fitzhugh, C.D.; Hankins, J.S.; Murad, M.H.; Panepinto, J.A.; Rondelli, D.; Shenoy, S.; et al. American Society of Hematology 2021 guidelines for sickle cell disease: Stem cell transplantation. Blood Adv. 2021, 5, 3668–3689. [Google Scholar] [CrossRef]
  69. Krishnamurti, L.; Kharbanda, S.; Biernacki, M.A.; Zhang, W.; Baker, K.S.; Wagner, J.E.; Wu, C.J. Stable long-term donor engraftment following reduced-intensity hematopoietic cell transplantation for sickle cell disease. Biol. Blood Marrow Transpl. 2008, 14, 1270–1278. [Google Scholar] [CrossRef] [Green Version]
  70. Matthes-Martin, S.; Lawitschka, A.; Fritsch, G.; Lion, T.; Grimm, B.; Breuer, S.; Boztug, H.; Karlhuber, S.; Holter, W.; Peters, C.; et al. Stem cell transplantation after reduced-intensity conditioning for sickle cell disease. Eur. J. Haematol. 2013, 90, 308–312. [Google Scholar] [CrossRef]
  71. Kassim, A.A.; Sharma, D. Hematopoietic stem cell transplantation for sickle cell disease: The changing landscape. Hematol. Oncol. Stem Cell Ther. 2017, 10, 259–266. [Google Scholar] [CrossRef]
  72. Alzahrani, M.; Damlaj, M.; Jeffries, N.; Alahmari, B.; Singh, A.; Rondelli, D.; Tisdale, J.F.; Saraf, S.L.; Hsieh, M.M. Non-myeloablative human leukocyte antigen-matched related donor transplantation in sickle cell disease: Outcomes from three independent centres. Br. J. Haematol. 2021, 192, 761–768. [Google Scholar] [CrossRef]
  73. Guilcher, G.M.T.; Monagel, D.A.; Nettel-Aguirre, A.; Truong, T.H.; Desai, S.J.; Bruce, A.; Shah, R.M.; Leaker, M.T.; Lewis, V.A. Nonmyeloablative Matched Sibling Donor Hematopoietic Cell Transplantation in Children and Adolescents with Sickle Cell Disease. Biol. Blood Marrow Transpl. 2019, 25, 1179–1186. [Google Scholar] [CrossRef] [PubMed]
  74. Saraf, S.L.; Oh, A.L.; Patel, P.R.; Jalundhwala, Y.; Sweiss, K.; Koshy, M.; Campbell-Lee, S.; Gowhari, M.; Hassan, J.; Peace, D.; et al. Nonmyeloablative Stem Cell Transplantation with Alemtuzumab/Low-Dose Irradiation to Cure and Improve the Quality of Life of Adults with Sickle Cell Disease. Biol. Blood Marrow Transpl. 2016, 22, 441–448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Iannone, R.; Casella, J.F.; Fuchs, E.J.; Chen, A.R.; Jones, R.J.; Woolfrey, A.; Amylon, M.; Sullivan, K.M.; Storb, R.F.; Walters, M.C. Results of minimally toxic nonmyeloablative transplantation in patients with sickle cell anemia and beta-thalassemia. Biol. Blood Marrow Transpl. 2003, 9, 519–528. [Google Scholar] [CrossRef] [Green Version]
  76. Horan, J.T.; Liesveld, J.L.; Fenton, P.; Blumberg, N.; Walters, M.C. 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 Transpl. 2005, 35, 171–177. [Google Scholar] [CrossRef]
  77. Nickel, R.S.C.K.; Hardy, S.J.; Ragarajan, H.G.; Chaudhury, S.; Kent, M.; Bhatia, M.; Zack, J.; Manj, V.; Guilcher, G.M.T.; Abraham, A. Nonmyeloablative HLA-Identical Sibling Donor Transplantation for Children and Young Adults with Sickle Cell Disease: Interim Results of the SUN Multicenter Phase II Trial. Blood 2021, 138, 1799. [Google Scholar] [CrossRef]
  78. de la Fuente, J.; Dhedin, N.; Koyama, T.; Bernaudin, F.; Kuentz, M.; Karnik, L.; Socié, G.; Culos, K.A.; Brodsky, R.A.; DeBaun, M.R.; et al. Haploidentical Bone Marrow Transplantation with Post-Transplantation Cyclophosphamide Plus Thiotepa Improves Donor Engraftment in Patients with Sickle Cell Anemia: Results of an International Learning Collaborative. Biol. Blood Marrow Transpl. 2019, 25, 1197–1209. [Google Scholar] [CrossRef]
  79. Ataga, K.I.; Kutlar, A.; Kanter, J.; Liles, D.; Cancado, R.; Friedrisch, J.; Guthrie, T.H.; Knight-Madden, J.; Alvarez, O.A.; Gordeuk, V.R.; et al. Crizanlizumab for the Prevention of Pain Crises in Sickle Cell Disease. N. Engl. J. Med. 2017, 376, 429–439. [Google Scholar] [CrossRef]
  80. Sahdev, I.; Brochstein, J.; Werther, N.; Stiles, J. Timing of Alemtuzumab with Respect to Day of Bone Marrow Infusion and its Effects Upon Engraftment and Graft-Versus-Host Disease in Patients with Sickle Cell Disease: A Single-Institutional Study. J. Pediatr. Hematol. Oncol. 2020, 42, e718–e722. [Google Scholar] [CrossRef]
  81. Eapen, M.; Brazauskas, R.; Walters, M.C.; Bernaudin, F.; Bo-Subait, K.; Fitzhugh, C.D.; Hankins, J.S.; Kanter, J.; Meerpohl, J.J.; Bolaños-Meade, J.; et al. Effect of donor type and conditioning regimen intensity on allogeneic transplantation outcomes in patients with sickle cell disease: A retrospective multicentre, cohort study. Lancet Haematol. 2019, 6, e585–e596. [Google Scholar] [CrossRef] [Green Version]
  82. Magnani, A.; Pondarre, C.; Bouazza, N.; Magalon, J.; Miccio, A.; Six, E.; Roudaut, C.; Arnaud, C.; Kamdem, A.; Touzot, F.; et al. Extensive multilineage analysis in patients with mixed chimerism after allogeneic transplantation for sickle cell disease: Insight into hematopoiesis and engraftment thresholds for gene therapy. Haematologica 2020, 105, 1240–1247. [Google Scholar] [CrossRef]
  83. Ngwube, A.; Franay, C.; Shah, N. Post-transplant CD34+ selected stem cell boost as an intervention for declining mixed chimerism following reduced intensity conditioning allogeneic stem cell transplant in children and young adults with sickle cell disease: A case series. Pediatr. Hematol. Oncol. 2022, 2013369. [Google Scholar] [CrossRef] [PubMed]
  84. Shin, S.H.; Park, S.S.; Park, S.; Jeon, Y.-W.; Yoon, J.-H.; Yahng, S.-A.; Cho, B.-S.; Kim, Y.-J.; Lee, S.; Kim, H.-J.; et al. Non-myeloablative matched sibling stem cell transplantation with the optional reinforced stem cell infusion for patients with hemoglobinopathies. Eur. J. Haematol. 2020, 105, 387–398. [Google Scholar] [CrossRef] [PubMed]
  85. Hernigou, P.; Bernaudin, F.; Reinert, P.; Kuentz, M.; Vernant, J.P. Bone-marrow transplantation in sickle-cell disease. Effect on osteonecrosis: A case report with a four-year follow-up. J. Bone Joint Surg. Am. 1997, 79, 1726–1730. [Google Scholar] [CrossRef] [PubMed]
  86. Mynarek, M.; Bettoni da Cunha Riehm, C.; Brinkmann, F.; Weiβenborn, K.; Tell-Lüersen, M.; Heuft, H.-G.; Maecker-Kolhoff, B.; Sykora, K.-W. Normalized transcranial Doppler velocities, stroke prevention and improved pulmonary function after stem cell transplantation in children with sickle cell anemia. Klin. Padiatr. 2013, 225, 127–132. [Google Scholar] [CrossRef] [PubMed]
  87. Verlhac, S.; Gabor, F.; Paillard, C.; Chateil, J.-F.; Jubert, C.; Petras, M.; Grevent, D.; Brousse, V.; Petit, P.; Thuret, I. Improved stenosis outcome in stroke-free sickle cell anemia children after transplantation compared to chronic transfusion. Br. J. Haematol. 2021, 193, 188–193. [Google Scholar] [CrossRef] [PubMed]
  88. Pecker, L.H.; Maher, J.Y.; Law, J.Y.; Beach, M.C.; Lanzkron, S.; Christianson, M.S. Risks associated with fertility preservation for women with sickle cell anemia. Fertil. Steril. 2018, 110, 720–731. [Google Scholar] [CrossRef] [Green Version]
  89. Matthews, M.; Pollack, R. Acute pain crisis in a patient with sickle cell disease undergoing ovarian simulation for fertility preservation prior to curative stem cell transplantation: Case report and literature review. J. Assist. Reprod. Genet. 2017, 34, 1445–1448. [Google Scholar] [CrossRef]
  90. Mishkin, A.D.; Mapara, M.Y.; Barhaghi, M.; Reshef, R. Fertility Concerns and Access to Care for Stem Cell Transplantation Candidates with Sickle Cell Disease. Biol. Blood Marrow Transpl. 2020, 26, e192–e197. [Google Scholar] [CrossRef]
  91. Zhao, J.; Beebe, K.; Magee, K.; Salzberg, D.; Stahlecker, J.; Miller, H.K.; Adams, R.H.; Lipskind, S.; Walsh, A.; Mirea, L.; et al. Adolescent male fertility following reduced-intensity conditioning regimen for hematopoietic stem cell transplantation in non-malignant disorders. Pediatr. Transpl. 2019, 23, e13496. [Google Scholar] [CrossRef]
  92. Pedersen, S.J.V.; Monagel, D.A.; Mammen, C.; Lewis, V.A.; Guilcher, G.M.T.; Bruce, A.A. Stable renal function in children and adolescents with sickle cell disease after nonmyeloablative hematopoietic stem cell transplantation. Pediatr. Blood Cancer 2020, 67, e28568. [Google Scholar] [CrossRef]
  93. Oostenbrink, L.V.E.; Pool, E.S.; Jol-van der Zijde, C.M.; Jansen-Hoogendijk, A.M.; Vervat, C.; van Halteren, A.G.S.; Bredius, R.G.M.; Smiers, F.J.W.; van Tol, M.J.D. Successful mismatched hematopoietic stem cell transplantation for pediatric hemoglobinopathy by using ATG and post-transplant cyclophosphamide. Bone Marrow Transpl. 2021, 56, 2203–2211. [Google Scholar] [CrossRef]
  94. Katewa, S.; Kharya, G.; Karnik, L.; Kassim, A.A.; de la Fuente, J. Pre-Transplantation Suppression of Haemopoiesis Is Associated with a High Rate of Macrophage Activation Syndrome in Ptcy Haploidentical Transplantation for Haemoglobinopathies. Blood 2017, 130, 1931. [Google Scholar]
  95. Shah, N.C.; Bhoopatiraju, S.; Abraham, A.; Anderson, E.; Andreansky, M.; Bhatia, M.; Chaudhury, S.; Cuvelier, G.D.E.; Godder, K.; Grimley, M.; et al. Granulocyte Colony-Stimulating Factor Is Safe and Well Tolerated following Allogeneic Transplantation in Patients with Sickle Cell Disease. Transpl. Cell Ther. 2022, 28, 174.e1–174.e5. [Google Scholar] [CrossRef]
  96. Palchaudhuri, R.; Saez, B.; Hoggatt, J.; Schajnovitz, A.; Sykes, D.B.; Tate, T.A.; Czechowicz, A.; Kfoury, Y.; Ruchika, F.; Rossi, D.J.; et al. Non-genotoxic conditioning for hematopoietic stem cell transplantation using a hematopoietic-cell-specific internalizing immunotoxin. Nat. Biotechnol. 2016, 34, 738–745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Persaud, S.P.; Ritchey, J.K.; Kim, S.; Lim, S.; Ruminski, P.G.; Cooper, M.L.; Rettig, M.P.; Choi, J.; DiPersio, J.F. Antibody-drug conjugates plus Janus kinase inhibitors enable MHC-mismatched allogeneic hematopoietic stem cell transplantation. J. Clin. Investig. 2021, 131, 145501. [Google Scholar] [CrossRef]
  98. Arai, Y.; Choi, U.; Corsino, C.I.; Koontz, S.M.; Tajima, M.; Sweeney, C.L.; Black, M.A.; Feldman, S.A.; Dinauer, M.C.; Malech, H.L. Myeloid Conditioning with c-kit-Targeted CAR-T Cells Enables Donor Stem Cell Engraftment. Mol. Ther. 2018, 26, 1181–1197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Common transplant conditioning regimens.
Table 1. Common transplant conditioning regimens.
Conditioning Regimen
Myeloablative
Busulfan (BU) > 7.2 mg/kg IV (or >9.0 mg/kg PO)
BU > 300 mg/m2 IV (or 375 mg/m2 PO)
Melphalan (Mel) > 150 mg/m2
Thiotepa (TT) > 10 mg/kg
Treosulfan (treo) > 30,000 mg/2 (or >30 g/m2)
Reduced Intensity and Non-myeloablative
Total Body Irradiation (TBI) ≤ 5 Gy (single) or TBI ≤8 Gy (fractionated)
Cyclophosphamide +/− Anti-Thymocyte Globulin (ATG) +/− Fludarabine (Flu)
BU ≤ 7.2 mg/kg IV or ≤9.0 mg/kg PO
BU ≤ 300 mg/m2 IV or ≤375 mg/m2 PO
Mel ≤ 150 mg/m2 +/− Flu
Treo ≤ 30 g/m2 +/− Flu
TT ≤ 10 mg/kg
PO per os (by mouth).
Table 2. Relative risk of outcomes after transplant by conditioning intensity.
Table 2. Relative risk of outcomes after transplant by conditioning intensity.
MyeloablativeReduced IntensityNon-Myeloablative
Overall survival *🡅 🡅 🡅🡅
Event-Free Survival
(= alive without graft failure)
🡅 🡅🡅
aGVHD🡅🡅🡅🡅 🡅
cGVHD 🡅🡅 🡅🡅🡅
Post-transplant malignancy ^none reported 🡅 🡅🡅
* In analyses limited to matched sibling donor setting, no association found between overall survival and regimen intensity. ^ difference not statistically significant.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Limerick, E.; Abraham, A. Across the Myeloablative Spectrum: Hematopoietic Cell Transplant Conditioning Regimens for Pediatric Patients with Sickle Cell Disease. J. Clin. Med. 2022, 11, 3856. https://doi.org/10.3390/jcm11133856

AMA Style

Limerick E, Abraham A. Across the Myeloablative Spectrum: Hematopoietic Cell Transplant Conditioning Regimens for Pediatric Patients with Sickle Cell Disease. Journal of Clinical Medicine. 2022; 11(13):3856. https://doi.org/10.3390/jcm11133856

Chicago/Turabian Style

Limerick, Emily, and Allistair Abraham. 2022. "Across the Myeloablative Spectrum: Hematopoietic Cell Transplant Conditioning Regimens for Pediatric Patients with Sickle Cell Disease" Journal of Clinical Medicine 11, no. 13: 3856. https://doi.org/10.3390/jcm11133856

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

Article Metrics

Back to TopTop