martes, 16 de abril de 2019

Childhood Hematopoietic Cell Transplantation (PDQ®) 1/2 —Health Professional Version - National Cancer Institute

Childhood Hematopoietic Cell Transplantation (PDQ®)—Health Professional Version - National Cancer Institute

National Cancer Institute



Childhood Hematopoietic Cell Transplantation (PDQ®)–Health Professional Version







General Information About Hematopoietic Cell Transplantation (HCT)



Rationale for HCT

Blood and marrow transplantation, or HCT, is a procedure that involves infusion of hematopoietic stem cells (hematopoietic progenitor cells) to reconstitute the hematopoietic system of a patient. The infusion of hematopoietic stem cells generally follows a preparative regimen consisting of agents designed to do the following:
  • Create marrow space.
  • Suppress the patient's immune system to prevent rejection.
  • Eradicate malignant cells in patients with cancer.
HCT is currently used in the following three clinical scenarios:
  1. Treatment of malignancies.
  2. Replacement or modulation of an absent or poorly functioning hematopoietic or immune system.
  3. Treatment of genetic diseases in which an insufficient expression of the affected gene product can be partially or completely overcome by circulating hematopoietic cells transplanted from a donor with normal gene expression.

Autologous Versus Allogeneic HCT

The two major transplant approaches currently in use are the following:
  • Autologous (using the patient's own hematopoietic stem cells).
  • Allogeneic (using related- or unrelated-donor hematopoietic stem cells).
An autologous transplant treats cancer by exposing patients to high-dose therapy with the intent of overcoming chemotherapy resistance in tumor cells, followed by infusion of the patient’s previously stored hematopoietic stem cells. The transplant can be performed in a single procedure or tandem sequential procedures. For autologous transplants to work for malignancies, the following must apply:
  • A dose-intensified chemotherapy/radiation therapy regimen with hematopoietic stem cell support is used to achieve a significantly higher cell kill than could be achieved without the use of hematopoietic stem cell support. This may include increased tumor kill in areas where standard-dose chemotherapy has less penetration (central nervous system).
  • Meaningful percentages of cure or long-term remission from the disease must occur without significant nonhematopoietic toxicities that would otherwise limit the therapeutic benefit achieved.
Autologous transplants have also been used to reset the immune system in patients with severe autoimmune disorders.
Current pediatric indications for autologous transplant include patients with certain lymphomas, neuroblastoma, and brain tumors. Autologous transplant techniques are also being used to enable engraftment of genetically modified autologous hematopoietic stem cell progenitors to correct/ameliorate inherited disorders (e.g., immunodeficiencies, metabolic disorders, and hemoglobinopathies).
Allogeneic transplant approaches to cancer treatment also may involve high-dose therapy, but because of immunologic differences between the donor and recipient, an additional graft-versus-tumor (GVT) or graft-versus-leukemia (GVL) treatment effect can occur. Although autologous approaches are associated with less short-term mortality, many malignancies are resistant to even high doses of chemotherapy and/or involve the bone marrow, thus requiring allogeneic approaches for optimal outcome.

Determining When HCT Is Indicated: Comparison of HCT and Chemotherapy Outcomes

Because the outcomes using chemotherapy and HCT treatments have been changing over time, regular comparisons between these approaches should be performed to continually redefine optimal therapy for a given patient. For some diseases, randomized trials or intent-to-treat using an HLA-matched sibling donor have established the benefit of HCT by direct comparison.[1,2] However, for very high-risk patients such as those with early relapse of acute lymphoblastic leukemia (ALL), randomized trials have not been feasible because of investigator bias.[3,4]
In general, HCT typically offers benefit only to children at high risk of relapse with standard chemotherapy approaches. Accordingly, treatment schemas that accurately identify these high-risk patients and offer HCT if reasonably HLA-matched donors are available have come to be the preferred approach for many diseases.[5] Less well-established, higher-risk approaches to HCT are generally reserved for only the very highest-risk patients. However, higher-risk approaches such as haploidentical transplantation are becoming safer and more efficacious and are increasingly being used interchangeably with fully matched allogeneic approaches.[6-9] (Refer to the Haploidentical HCT section of this summary for more information.)
When comparisons of similar patients treated with HCT or chemotherapy are made in the setting where randomized or intent-to-treat studies are not feasible, the following issues should be considered:
  1. Remission/disease status: Comparisons between HCT and chemotherapy should include only patients who obtain remission, preferably after similar approaches to salvage therapy, because patients failing to obtain remission do very poorly with any therapy.[10]
    To account for time-to-transplant bias, the chemotherapy comparator arm should include only patients who maintained remission until the median time to HCT. The HCT comparator arm should also include only patients who achieved the initial remission mentioned above and maintained that remission until the time of HCT.[10]
    High-risk and intermediate-risk patient groups should not be combined because a benefit for HCT in the high-risk group can be masked when outcomes are similar to those achieved in the intermediate-risk group.[10]
  2. Therapy approaches used for comparison: Comparisons should be made with the best or most commonly used chemotherapy and HCT approaches utilized during the time frame under study.
  3. HCT approach: HCT approaches that are very high risk or have documented lower rates of survival should not be combined for analysis with standard-risk HCT approaches.
  4. Criteria for relapse: Risk factors for relapse should be carefully defined, and analysis should be based on the most current knowledge of risk.
  5. Selection bias: Attempts should be made to understand and eliminate or correct for selection bias. Examples include the following:
    • Higher-risk patients preferentially undergoing HCT (i.e., patients who take several rounds to achieve remission or who relapse after obtaining remission and go back into a subsequent remission before HCT).
    • Sicker patients deferred from HCT because of comorbidities.
    • Related to the time-to-transplant bias noted above, patients who undergo HCT after relapse or recurrence are a subset of all patients with a disease recurrence and will be selected from those who are able to obtain a remission and remain healthy enough to undergo HCT.
    • Patient or parent refusal.
    • Lack of or inability to obtain insurance approval for HCT.
    • Lack of access to HCT because of distance or inability to travel.
One source of bias difficult to control for or detect is physician bias for or against HCT. The effects of access to HCT and therapeutic bias on outcomes of pediatric malignancies for which HCT may be indicated have been poorly studied to date.


References
  1. Matthay KK, Villablanca JG, Seeger RC, et al.: Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13-cis-retinoic acid. Children's Cancer Group. N Engl J Med 341 (16): 1165-73, 1999. [PUBMED Abstract]
  2. Woods WG, Neudorf S, Gold S, et al.: A comparison of allogeneic bone marrow transplantation, autologous bone marrow transplantation, and aggressive chemotherapy in children with acute myeloid leukemia in remission. Blood 97 (1): 56-62, 2001. [PUBMED Abstract]
  3. Lawson SE, Harrison G, Richards S, et al.: The UK experience in treating relapsed childhood acute lymphoblastic leukaemia: a report on the medical research council UKALLR1 study. Br J Haematol 108 (3): 531-43, 2000. [PUBMED Abstract]
  4. Gaynon PS, Harris RE, Altman AJ, et al.: Bone marrow transplantation versus prolonged intensive chemotherapy for children with acute lymphoblastic leukemia and an initial bone marrow relapse within 12 months of the completion of primary therapy: Children's Oncology Group study CCG-1941. J Clin Oncol 24 (19): 3150-6, 2006. [PUBMED Abstract]
  5. Schrauder A, von Stackelberg A, Schrappe M, et al.: Allogeneic hematopoietic SCT in children with ALL: current concepts of ongoing prospective SCT trials. Bone Marrow Transplant 41 (Suppl 2): S71-4, 2008. [PUBMED Abstract]
  6. Bertaina A, Merli P, Rutella S, et al.: HLA-haploidentical stem cell transplantation after removal of αβ+ T and B cells in children with nonmalignant disorders. Blood 124 (5): 822-6, 2014. [PUBMED Abstract]
  7. Handgretinger R, Chen X, Pfeiffer M, et al.: Feasibility and outcome of reduced-intensity conditioning in haploidentical transplantation. Ann N Y Acad Sci 1106: 279-89, 2007. [PUBMED Abstract]
  8. 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 38 (4): 291-7, 2006. [PUBMED Abstract]
  9. Luznik L, Fuchs EJ: High-dose, post-transplantation cyclophosphamide to promote graft-host tolerance after allogeneic hematopoietic stem cell transplantation. Immunol Res 47 (1-3): 65-77, 2010. [PUBMED Abstract]
  10. Pulsipher MA, Peters C, Pui CH: High-risk pediatric acute lymphoblastic leukemia: to transplant or not to transplant? Biol Blood Marrow Transplant 17 (1 Suppl): S137-48, 2011. [PUBMED Abstract]

Autologous HCT



Collection and Storage of Autologous Hematopoietic Stem Cells

Autologous procedures require collection of growth-factor–mobilized peripheral blood stem cells (PBSCs) from patients by the process of leukapheresis. Bone marrow can be used for autologous transplants, but PBSCs have been shown to lead to quicker blood count recovery and cause less toxicity.
Patients being considered for autologous hematopoietic cell transplantation (HCT) are generally given chemotherapy to determine tumor responsiveness and minimize the risk of tumor contamination in their bone marrow. After a number of rounds of chemotherapy, patients undergo the leukapheresis procedure, either as their blood counts recover from chemotherapy or during a break between chemotherapy treatments. Growth factors such as granulocyte colony-stimulating factor (G-CSF) are used to increase the number of circulating stem and progenitor cells (CD34+ cells). Collection centers monitor the CD34-positive number in the patient and product each day to determine the best time to begin collection and when collection is complete. Patients with poorly mobilized CD34-positive cells can often have their cells successfully collected using alternative mobilization approaches (e.g., plerixafor).[1] The collected PBSCs are cryopreserved for later use. After completion of an intensive preparative regimen using high-dose chemotherapy, which varies according to the tumor type, the PBSCs are administered back into the patient at the time of transplant.

General Indications for Autologous Procedures/Preparative Regimens/Tumor Purging

In pediatrics, the most common autologous transplant indications are the following:
Tumor-specific regimens are described in disease-specific PDQ treatment summaries.
The tumor-specific activity and intensity of agents used for autologous regimens have been shown to be important in improving survival. This is not the case for allogeneic procedures.
One concern with autologous approaches for these and other tumor types has been the contamination of the collected stem cell product by persistent tumor cells. Although many techniques have been developed to remove or purge tumor cells from products, most studies have shown no benefit to tumor purging.[2]


References
  1. Patel B, Pearson H, Zacharoulis S: Mobilisation of haematopoietic stem cells in paediatric patients, prior to autologous transplantation following administration of plerixafor and G-CSF. Pediatr Blood Cancer 62 (8): 1477-80, 2015. [PUBMED Abstract]
  2. Kreissman SG, Seeger RC, Matthay KK, et al.: Purged versus non-purged peripheral blood stem-cell transplantation for high-risk neuroblastoma (COG A3973): a randomised phase 3 trial. Lancet Oncol 14 (10): 999-1008, 2013. [PUBMED Abstract]

Allogeneic HCT



Improved Outcomes After Allogeneic Transplantation

Over the past one to two decades, significant advances have led to improved outcomes after allogeneic hematopoietic cell transplantation (HCT).[1-3] The most significant improvements in survival occurred in unrelated and alternative donor procedures.[4-6] Possible explanations for these improvements in survival include improved patient selection, better supportive care, refined treatment regimens, improved approaches specific to stem cell sources, and better HLA typing. All of these factors may have contributed to better outcomes; however, the section below focuses on modifiable aspects of HCT (i.e., optimization of HLA typing and selection of stem cell sources).

HLA Matching and Hematopoietic Stem Cell Sources

HLA overview

Appropriate matching between donor and recipient HLA in the major histocompatibility complex located on chromosome 6 is essential to successful allogeneic HCT (refer to Figure 1 and Tables 1 and 2).
ENLARGEHuman lymphocyte antigen (HLA) complex; drawing shows the long and short arms of human chromosome 6 with amplification of the HLA region, including the class I A, B, and C alleles, and the class II DP, DQ, and DR alleles.
Figure 1. HLA Complex. Human chromosome 6 with amplification of the HLA region. The locations of specific HLA loci for the class I B, C, and A alleles and the class II DP, DQ, and DR alleles are shown.
HLA class I (A, B, C, etc.) and class II (DRB1, DQB1, DPB1, etc.) alleles are highly polymorphic; therefore, finding appropriately matched unrelated donors is a challenge for some patients, especially those of certain racial groups (e.g., African Americans and Hispanics).[7,8] Full siblings of cancer patients have a 25% chance of being HLA matched.
Early serologic techniques of HLA assessment defined a number of HLA antigens, but more precise DNA methodologies have shown HLA allele-level mismatches in up to 40% of serologic HLA antigen matches. These differences are clinically relevant because the use of donors with allele-level mismatches affects survival and rates of graft-versus-host disease (GVHD) to a degree similar to that in patients with antigen-level mismatches.[9] Because of this, DNA-based allele-level HLA typing is standard when unrelated donors are being chosen.
Table 1. Level of HLA Typing Currently Used for Different Hematopoietic Stem Cell Sourcesa,b,c
ENLARGE
Class I AntigensClass II Antigens
BM = bone marrow; PBSCs = peripheral blood stem cells.
aHLA antigen: A serologically defined, low-resolution method of defining an HLA protein. Differs from allele-level typing half of the time. Designated by the first two numbers (i.e., HLA B 35:01—antigen is HLA B 35).
bHLA allele: A higher-resolution method of defining unique HLA proteins by typing their gene through sequencing or other DNA-based methods that detect unique differences. Designated by at least four numbers (i.e., HLA B 35:01—35 is the antigen and 01 is the allele).
cConsensus recommendations for HLA typing, including extended class II typing of mismatched donors, have been published by the National Cancer Institute/National Heart, Lung, and Blood Institute–sponsored Blood and Marrow Transplant Clinical Trials Network.[10]
dSiblings need confirmation that they have fully matched haplotypes with no crossovers in the A to DRB1 region. If parental typing is performed and haplotypes are established, antigen-level typing of class I is adequate. With no parental haplotypes, allele-level typing of eight alleles is recommended.
eParent, cousin, etc., with a phenotypic match or near-complete HLA match.
Stem Cell SourceHLA AHLA BHLA CHLA DRB1HLA DQB1;HLA DPB1; HLA DR3,4,5
Matched siblingdBM/PBSCsAntigen or alleleAntigen or alleleOptionalAllele
Mismatched sibling/other related-donoreBM/PBSCsAlleleAlleleAlleleAlleleRecommended, if mismatches are present
Unrelated-donor BM/PBSCsAlleleAlleleAlleleAlleleRecommended, if mismatches are present
Unrelated-donor cord bloodAntigen (allele recommended)Antigen (allele recommended)Allele recommendedAllele
Table 2. Definitions of the Numbers Describing HLA Antigens and Alleles Matching
If These HLA Antigens and Alleles Match:Then the Donor Is Considered to be This Type of Match:
A, B, and DRB16/6
A, B, C, and DRB18/8
A, B, C, DRB1, and DQB110/10
A, B, C, DRB1, DQB1, and DPB112/12

HLA matching considerations for sibling and related donors

The most commonly used related donor is a sibling from the same parents who, at a minimum, is HLA matched for HLA A, HLA B, and HLA DRB1 at the antigen level. Given the distance between HLA A and HLA DRB1 on chromosome 6, there is approximately a 1% possibility of a crossover event occurring in a possible sibling match. Because a crossover event could involve the HLA C antigen and because parents may share HLA antigens that actually differ at the allele level, many centers perform allele-level typing of possible sibling donors at all of the key HLA antigens (HLA A, B, C, and DRB1). Any related donor that is not a full sibling should have full HLA typing because similar haplotypes from different parents could differ at the allele level.
Although single-antigen mismatched related donors (5/6 antigen matched) were used interchangeably with matched sibling donors in some studies, a large Center for International Blood and Marrow Transplant Research (CIBMTR) study in pediatric HCT recipients showed that the use of 5/6 antigen-matched related donors resulted in rates of GVHD and overall survival (OS) equivalent to rates in 8/8-allele-level-matched unrelated donors and slightly inferior survival than in fully matched siblings.[11] Any siblings with single mismatches should have extended typing to ensure that if the mismatch is caused by a crossover, it only occurs with one antigen. If clinicians choose siblings with multiple antigen mismatches as donors, haploidentical approaches may be warranted.

HLA matching considerations for unrelated donors

Optimal outcomes are achieved in unrelated allogeneic marrow transplantation when the pairs of antigens at HLA A, B, C, and DRB1 are matched between the donor and the recipient at the allele level (termed an 8/8 match) (refer to Table 2).[12] A single antigen/allele mismatch at any of these antigens (7/8 match) lowers the probability of survival between 5% and 10%, with a similar increase in the amount of significant (grades III–IV) acute GVHD.[12] Of these four antigen pairs, different reports have shown HLA A, C, and DRB1 mismatches to potentially be more highly associated with mortality than the other antigens,[9,12,13] but the differences in outcome are small and inconsistent, making it very difficult to conclude that one can pick a more favorable mismatch by choosing one type of antigen mismatch over another. Many groups are attempting to define specific antigens or pairs of antigens that are associated with either good or poor outcomes. For example, a specific HLA C mismatch (HLA-C*03:03/03:04) has outcomes similar to a match; therefore, selection of this mismatch is desirable in an otherwise matched donor/pair combination.[14]
It is well understood that class II antigen DRB1 mismatches increase GVHD incidence and worsen survival.[13] Subsequent data have also shown that multiple mismatches of DQB1, DPB1, and DR3,4,5 lead to worse outcomes in the setting of less than 8/8 matches.[15] DPB1 mismatches have been extensively studied and classified as permissive or nonpermissive based on T-cell epitope matching. Patients with 10/10 matches and nonpermissive DPB1 mismatches have more transplant-related mortality but have survival rates similar to those with DPB1 matches or permissive matches. Those with 9/10 matches who have nonpermissive DPB1 mismatches had worse survival than did those with permissive mismatches or DPB1 matches.[16-18]
With these findings in mind, although a 7/8- or 8/8-matched unrelated donor can be used routinely, centers may be able to further improve outcomes by the following:
  • Extended typing of DQB1, DPB1, and DR3,4,5, especially in the context of a less-than-8/8-matched donor.[16-18]
  • Extended HLA testing to select appropriate donors in the context of HLA-sensitized patients to avoid the potential risk of graft failure.[19,20] HLA sensitization is detected by testing for the presence of specific anti-HLA antibodies and avoidance of donors who have any HLA antigens associated with the antibodies present in the recipient.
  • Use of younger donors.[10]
  • Matching cytomegalovirus (CMV)-positive or CMV-negative recipients with positive or negative donors.[21]
  • Use of blood type–compatible unrelated donors.[10]
ENLARGEChart showing HLA allele duplication and type of match between donor and recipient: an allele match (0201 and 0401 for both donor and recipient); a mismatch (0201 for both donor and recipient and 0201 for donor, 0401 for recipient) shown by an arrow pointing in a direction that promotes GVHD (GVH-O); a mismatch (0201 for both donor and recipient and 0401 for donor, 0201 for recipient) shown by an arrow pointing in a direction that promotes rejection (R-O); and a bidirectional mismatch (0201 for donor, 0301 for recipient, and 0401 for both  donor and recipient) shown by arrows pointing in two directions, a direction that promotes rejection (R-O) and a direction that promotes GVHD (GVH-O).
Figure 2. HLA allele duplication in a donor or recipient results in a half match and a mismatch that will either occur in a direction that promotes GVHD (GVH-O) or a direction that promotes rejection (R-O).
If a donor or recipient has a duplication of one of their HLA alleles, they will have a halfmatch and a mismatch only in one direction. Figure 2 illustrates that these mismatches will occur in either a direction that promotes GVHD (GVH-O) or a direction that promotes rejection (R-O). When 8/8-matched unrelated donors are compared with 7/8 donors mismatched in the GVH-O direction, 7/8 mismatched in the R-O direction, or 7/8 mismatched in both directions, the mismatch in the R-O direction leads to rates of grades III and IV acute GVHD similar to rates in the 8/8 matched and better than in the other two combinations. The 7/8 mismatched in only the R-O direction is preferred over GVH-O and bidirectional mismatches.[22] It is important to note that this observation in unrelated donors differs from observations in cord blood recipients, outlined below.

HLA matching and cell dose considerations for unrelated cord blood HCT

Another commonly used hematopoietic stem cell source is that of unrelated umbilical cord blood, which is harvested from donor placentas moments after birth. The cord blood is processed, HLA typed, cryopreserved, and banked.
Unrelated cord blood transplantation has been successful with less stringent HLA matching requirements compared with standard related or unrelated donors, probably because of limited antigen exposure experienced in utero and different immunological composition. Cord blood matching has traditionally been performed at an intermediate level for HLA A and B and at an allele level (high resolution) for DRB1. This means that until just recently, attempted matching of only six antigens has been necessary to choose units for transplantation.
Although better outcomes occur when 6/6 or 5/6 HLA-matched units are used,[23] successful HCT has occurred even with 4/6 or less HLA-matched units in many patients. In a large CIBMTR/Eurocord study, better matching at the allele level using eight antigens (matching for HLA A, B, C, and DRB1) resulted in less transplant-related mortality and improved survival. Best outcome was noted with 8/8 allele matching versus 4/8 to 7/8 matches, with poor survival in patients with five or more allele mismatches. Patients receiving 8/8-matched cord blood did not require higher cell doses for better outcomes; however, those with one to three allele mismatches had less transplant-related mortality with total nucleated cell counts higher than 3 × 107/kg, and those with four allele mismatches required a total nucleated cell count higher than 5 × 107/kg to decrease transplant-related mortality.[24] This observation was noted to be especially important in cord blood transplantation for nonmalignant disorders, where any mismatching below 7/8 alleles led to inferior survival.[25] Many centers will type additional alleles and use the best match possible, but the impact of DQB1, DPB1, and DR3,4,5 mismatches has not been studied in detail.
As in unrelated peripheral blood stem cells (PBSCs) or bone marrow donors, extended HLA testing can support the selection of appropriate cord blood units in HLA-sensitized patients to avoid the potential risk of graft failure.[26,27] Evidence also suggests that selecting a mismatched cord blood unit, where the mismatch involves a noninherited maternal antigen, may improve survival.[28,29]
As with unrelated donors, individuals can occasionally have duplicate HLA antigens (e.g., the HLA A antigen is 01 on both chromosomes). When this occurs in a donor product and the antigen is matched to one of the recipient antigens, the recipient immune response will see the donor antigens as matched (matched, in the rejection direction), but the donor immune response will see a mismatch in the recipient (mismatched in the GVHD direction). This variation of partial mismatching has been shown to be important in cord blood transplant outcomes. Mismatches that are only in the GVHD direction (GVH-O) lead to lower transplant-related mortality and overall mortality than in those with recipient direction only (R-O) mismatches.[30] R-O mismatches have outcomes similar to those of bidirectional mismatches.[31] Although these studies suggest that using unidirectional mismatching as a criteria for cord blood selection may be beneficial, a Eurocord–European Society for Blood and Marrow Transplantation analysis disputes the value of this type of mismatching.[32]
Two aspects of umbilical cord blood HCT have made the practice more widely applicable. First, because a successful procedure can occur with multiple HLA mismatches, more than 95% of patients from a wide variety of ethnicities are able to find at least a 4/6-matched cord blood unit.[7,33] Second, as mentioned above, adequate cell dose (minimum 2–3 × 107total nucleated cells/kg and 1.7 × 105 CD34+ cells/kg) has been shown to be associated with improved survival.[34,35] Total nucleated cells are generally used to judge units because techniques to measure CD34-positive doses have not been standardized. Because even large single umbilical cord blood units are only able to supply these minimum doses to recipients weighing up to 40 kg to 50 kg, early umbilical cord blood HCT focused mainly on smaller children. Later studies showed that barriers of this size could be overcome by using two umbilical cord blood units, as long as each of the units is at least a 4/6 HLA match with the recipient; because two cords provide higher cell doses, umbilical cord blood transplantation is now used widely for larger children and adults.[36]
If a single unit provides an adequate cell dose, there may be disadvantages to adding a second unit.[37][Level of evidence: 1iiA] Two randomized trials showed that in children who had adequately sized single units, the addition of a second unit did not alter relapse, transplant-related mortality, or survival rates, but was associated with higher rates of extensive chronic GVHD.[37,38]
Investigators have shown that by using combinations of cytokines and other compounds to expand cord blood for a period of time before infusion, engraftment of cord blood cells can occur more rapidly than after standard approaches.[39-42] Although some studies using multiple units or split units have shown that expanded units will engraft early and then give way to nonexpanded units for long-term reconstitution,[43] other studies are showing persistence of expanded cells, implying preservation of stem cells through the expansion process.[41,42] A number of these approaches are currently under investigation; their effect on efficacy and survival of children using cord blood as a stem cell source has yet to be established and none are approved by the U.S. Food and Drug Administration (FDA).

Comparison of stem cell products

Currently, the following three stem cell products are used from both related and unrelated donors:
  • Bone marrow.
  • PBSCs.
  • Cord blood.
In addition, bone marrow or PBSCs can be T-cell depleted by several methods, and the resultant stem cell product has very different properties. Finally, partially HLA-matched (half or more antigens [haploidentical]) related bone marrow or PBSCs can be used after in vitro or in vivo T-cell depletion, and this product also behaves differently from other stem cell products. A comparison of stem cell products is presented in Table 3.
Table 3. Comparison of Hematopoietic Stem Cell Products
ENLARGE
PBSCsBMCord BloodT-cell–Depleted BM/PBSCsHaploidentical T-cell–Depleted BM/PBSCs
BM = bone marrow; EBV-LPD = Epstein-Barr virus–associated lymphoproliferative disorder; GVHD = graft-versus-host disease; HCT = hematopoietic cell transplantation; PBSCs = peripheral blood stem cells.
aAssuming no development of GVHD. If patients develop GVHD, immune reconstitution is delayed until resolution of the GVHD and discontinuation of immune suppression.
bIf a haploidentical donor is used, longer times to immune reconstitution may occur.
T-cell contentHighModerateLowVery lowVery low
CD34+ contentModerate–highModerateLow (but higher potency)Moderate–highModerate–high
Time to neutrophil recoveryRapid: median, 16 d (11–29 d) [44]Moderate: median, 21 d (12–35 d) [44]Slower: median, 23 d (11–133 d) [38]Rapid: median, 16 d (9–40 d) [45]Rapid: median, 13 d (10–20 d) [46]
Early post-HCT risk of infections, EBV-LPDLow–moderateModerateHighVery HighVery High
Risk of graft rejectionLowLow–moderateModerate–highModerate–highModerate–high
Time to immune reconstitutionaRapid (6–12 mo)Moderate (6–18 mo)Slow (6–24 mo)Slow (6–24 mo)Slow (9–24 mo)b
Risk of acute GVHDModerateModerateModerateLowLow
Risk of chronic GVHDHighModerateLowLowLow
The main differences between the products are associated with the numbers of T cells and CD34-positive progenitor cells present; very high levels of T cells are present in PBSCs, intermediate numbers in bone marrow, and low numbers in cord blood and T-cell–depleted products. Patients receiving T-cell–depleted products or cord blood generally have slower hematopoietic recovery, increased risk of infection, late immune reconstitution, higher risks of nonengraftment, and increased risk of Epstein-Barr virus (EBV)–associated lymphoproliferative disorder. This is countered by lower rates of GVHD and an ability to offer transplantation to patients where full HLA matching is not available. Higher doses of T cells and other cells in PBSCs result in rapid neutrophil recovery and immune reconstitution, but also increase rates of chronic GVHD.
Only a few studies directly compared outcomes of different stem cell sources/products in pediatric patients.
Evidence (comparison of outcomes of stem cell sources/products in children):
  1. A retrospective registry study of pediatric patients who underwent HCT for acute leukemia compared those who received related-donor bone marrow with those who received related-donor PBSCs.[47]
    • Although the bone marrow and PBSC recipient cohorts differed some in their risk profiles, after statistical correction, increased risk of GVHD and transplant-related mortality associated with PBSCs led to poorer survival in the PBSC group.
  2. A retrospective study of Japanese children with acute leukemia compared 90 children who received PBSCs with 571 children who received bone marrow.[48]
    • The study confirmed higher transplant-related mortality caused by GVHD and inferior survival among the children who received PBSCs.
These reports, combined with a lack of prospective studies comparing bone marrow and PBSCs, have led most pediatric transplant protocols to prefer bone marrow over PBSCs from related donors.
For those requiring unrelated donors, a large Blood and Marrow Transplant Clinical Trials Network (BMT CTN) trial that included a number of pediatric patients randomly assigned enrollees to receive either bone marrow or PBSCs. This trial demonstrated that OS was identical using either source, but rates of chronic GVHD were significantly higher in the PBSC arm, with a small increase in rejection in the bone marrow arm.[49] Rejections were rare in pediatric patients, and there was an insufficient number of patients to draw specific conclusions about rejection risk in children who received bone marrow.
Published studies comparing unrelated cord blood and bone marrow have been retrospective, with weaknesses inherent in such analyses.
Evidence (comparison of unrelated cord blood versus bone marrow outcomes):
  1. In one study, pediatric patients with acute lymphoblastic leukemia (ALL) who underwent HCT and received 8/8-HLA-allele–matched unrelated-donor bone marrow were compared with those who received unrelated cord blood.[23]
    • The analysis showed that the best survival occurred in recipients of 6/6 HLA-matched cord blood; survival after 8/8 HLA-matched unrelated bone marrow was slightly less and was statistically identical to survival for patients receiving 5/6 and 4/6 HLA-matched cord blood units.
  2. In a second study from a single center consisting of mostly adult patients with acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), and ALL, outcomes for cord blood recipients were compared with outcomes for recipients of matched and mismatched unrelated-donor bone marrow/PBSCs.[50]
    • Better survival because of less relapse was noted in cord blood recipients, mainly resulting from superior survival in patients with minimal residual disease (MRD) present just before transplant.
    • No difference was seen in relapse and survival between patients with pre-HCT MRD and patients without pre-HCT MRD if they received cord blood.
    • This result is controversial because it contradicts many other studies that showed that the presence of pre-HCT MRD in cord blood recipients led to increased relapse and inferior survival.[51-54]
On the basis of these studies, most transplant centers consider matched sibling bone marrow to be the preferred stem cell source/product. If a sibling donor is not available, fully matched unrelated-donor bone marrow or PBSCs or HLA-matched (4/6 to 6/6) cord blood leads to similar survival rates. Although adult studies of T-cell–depleted unrelated bone marrow or PBSCs have shown outcomes similar to non–T-cell–depleted approaches, large pediatric trials or retrospective studies comparing T-cell–depleted matched or haploidentical bone marrow or PBSCs have not been conducted.

Haploidentical HCT

Early HCT studies demonstrated progressively higher percentages of patients experiencing severe GVHD and lower survival rates as the number of donor/recipient HLA mismatches increased.[55] Other studies showed that even with very high numbers of donors in unrelated-donor registries, patients with rare HLA haplotypes and patients with certain ethnic backgrounds (e.g., Hispanic, African American, Asian-Pacific Islander, etc.) have a low chance of achieving desired levels of HLA matching (7/8 or 8/8 match at the allele level).[8]
To allow access to HCT for patients without fully HLA-matched donor options, investigators have developed techniques allowing the use of siblings, parents, or other relatives who share only a single haplotype of the HLA complex with the patient and are thus halfmatches. Most approaches developed to date rely on intense T-cell depletion of the product before infusion into the patient. The main challenge associated with this approach is intense immune suppression with delayed immune recovery, which can result in lethal infections,[56] increased risk of EBV-associated lymphoproliferative disorder, and high rates of relapse.[57] This has generally led to inferior survival compared with matched HCT and has resulted in the procedure being practiced mainly at larger academic centers with a specific research focus on studying and developing this approach.
Current approaches, however, are rapidly evolving, resulting in improved outcomes, with some pediatric groups reporting survival similar to that of standard approaches.[58,59] These approaches include the following:
  • Newer techniques of T-cell depletion and add-back of specific cell populations (e.g., CD3 or alpha-beta CD3/CD19-negative selection) may decrease transplant-related mortality.[60]; [61,62][Level of evidence: 3iiiDii]
  • Reduced toxicity regimens have led to improved survival.
  • Better supportive care has decreased the chance of morbidity from infection or EBV-associated lymphoproliferative disorder.[63]
  • Some patient-donor combinations that have specific killer immunoglobulin-like receptor mismatches have shown decreased likelihood of relapse (refer to the Role of killer immunoglobulin-like receptor mismatching in HCT section of this summary for more information).
  • Certain techniques, such as using combinations of granulocyte colony-stimulating factor (G-CSF)–primed bone marrow and PBSCs with posttransplant antibody–based T-cell depletion [64] or post-HCT cyclophosphamide (chemotherapeutic T-cell depletion),[65]; [66][Level of evidence: 3iiiA] have made these procedures more accessible to centers because the expensive and complicated processing necessary for traditional T-cell depletion are not used.
Reported rates of survival using many different types of haploidentical approaches varies between 25% and 80%, depending on the technique used and the risk of the patient undergoing the procedure.[57,58,64,65]; [66][Level of evidence: 3iiiA] Whether haploidentical approaches are superior to cord blood or other stem cell sources for a given patient group has not been determined because comparative studies have yet to be performed.[57]
Even more than with other stem cell sources, patients undergoing haploidentical procedures can develop anti-HLA antibodies that, if directed against nonshared haploidentical antigens, can greatly increase the risks of rejection. Clinicians should choose donors with HLA types against whom the recipient does not have an antibody present, if possible. Guidelines on how to best approach this issue have been published.[67]

Other donor characteristics associated with outcome

Although HLA matching has consistently been the most important factor associated with improved survival in nonhaploidentical allogeneic HCT, a number of other characteristics of the donor have been shown in studies to affect key outcomes. Higher cell dose from the donor (refer to the HLA matching and cell dose considerations for unrelated cord blood HCT section of this summary for more information) has also been shown to be important when related, unrelated, or haploidentical bone marrow or PBSC donors are used.[68,69] The effects of donor age, blood type, CMV status, sex, and parity of female donors have also been studied.
Ideally, after HLA matching, transplant centers should select donors based on the following characteristics:
  • Donor age. The youngest donor available is preferred.[70,71]
  • CMV status of the recipient. CMV-negative donor matched to CMV-negative recipient and CMV-positive donor matched to CMV-positive recipient are preferred.[72]
  • Matched donor blood type.[73-75]
  • Donor sex and parity of female donors. Male or nonparous female donors are preferred over parous female donors.[71,76]
Rarely can a donor/recipient pair fit perfectly into this algorithm, and determining which of these characteristics should be chosen over others has been controversial. A CIBMTR study involving 6,349 patients who underwent transplant for hematological malignancies from 1988 to 2006, with a confirmation cohort of 4,690 patients who underwent transplant between 2007 and 2011, tested the effect of donor characteristics while adjusting for disease risk and other key transplant characteristics.[71,77]
  • The earlier data set showed that in addition to HLA mismatching, older donor age and major or minor ABO blood-type mismatching increased overall mortality; parous female graft recipients experienced lower rates of relapse; recipients of younger donor grafts had lower rates of acute GVHD; and recipients of parous female grafts had higher rates of chronic GVHD. Recipient CMV status was more important than donor CMV status (recipients who are CMV-positive are at higher risk of mortality independent of the donor CMV status), although a CMV-negative donor to a CMV-negative recipient combination improves survival.[71]
  • The more recent confirmation cohort was tested by a multivariate analysis for independent predictors of survival in an EBMT study. Older donor age was confirmed to be independently associated with worse OS; every 10 years of donor age increased the risk of mortality by 5.5%. HLA matching continued to have the most important effect on survival; ABO mismatching was not confirmed to have a continuing effect.[77]
Thus, after HLA matching, donor age is likely the most important second factor to optimize, unless the recipient is CMV negative, at which point finding a CMV-negative donor would take priority.
A number of studies have attempted to identify characteristics of the best donors for haploidentical procedures. As with conventional bone marrow transplantation, use of younger donors appears to be beneficial, but data regarding donor sex are inconclusive. Studies involving intense T-cell depletion have noted better outcomes using maternal donors,[78] but studies using posttransplant cyclophosphamide or intense immune suppression seem to favor male donors.[79,80] Further study is needed to clarify this important issue. One large comparison of haploidentical donors showed an effect of ABO incompatibility on engraftment (risk of rejection doubling from 6% to 12%, ABO match vs. ABO major mismatch), and patients receiving bidirectionally mismatched donors had a 2.4-fold increase in grades II to IV acute GVHD.[81]

Immunotherapeutic Effects of Allogeneic HCT

Graft-versus-leukemia (GVL) effect

Early studies in HCT focused on the delivery of intense myeloablative preparative regimens followed by rescue of the hematopoietic system with either an autologous or allogeneic bone marrow. Investigators quickly showed that allogeneic approaches led to a decreased risk of relapse caused by an immunotherapeutic reaction of the new bone marrow graft against tumor antigens. This phenomenon came to be termed the GVL or graft-versus-tumor (GVT) effect, and has been shown to be associated with mismatches to both major and minor HLA antigens.
The GVL effect is challenging to use therapeutically because of a strong association between GVL and clinical GVHD. For standard approaches to HCT, the highest survival rates have been associated with mild or moderate GVHD (grades I to II in AML and grades I to III in ALL), compared with patients who have no GVHD and experience more relapse or patients with severe GVHD who experience more transplant-related mortality.[82,83]; [84][Level of evidence: 3iDi]
Understanding when GVL occurs and how to use GVL optimally is challenging. One method of study is comparing rates of relapse and survival between patients undergoing myeloablative HCT with either autologous or allogeneic donors for a given disease.
  • Leukemia and MDS: A clear advantage has been noted when allogeneic approaches are used for ALL, AML, chronic myelogenous leukemia (CML), and MDS. For ALL and AML specifically, autologous HCT approaches for most high-risk patient groups have shown results similar to those obtained with chemotherapy, while allogeneic approaches produced superior results.[85,86]
  • Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL): Patients with HL or NHL generally fare better with autologous approaches, although there may be a role for allogeneic approaches in relapsed lymphoblastic lymphoma, lymphoma that is poorly responsive to chemotherapy, or lymphoma that has relapsed after autologous HCT.[87]
Further insights into the therapeutic benefit of GVL/GVT for given diseases have come from the use of reduced-intensity preparative regimens (refer to the Principles of Allogeneic HCT Preparative Regimens section of this summary for more information). This approach to transplantation relies on GVL because, in most cases, the intensity of the preparative regimen is not sufficient for cure. Although studies have shown benefit for patients pursuing this approach when they are ineligible for standard transplantation,[88] this approach has not been used for most children with cancer who require HCT because pediatric cancer patients can generally undergo myeloablative approaches safely.
Using donor lymphocyte infusions (DLI) or early withdrawal of immune suppression to enhance GVL
GVL can be delivered therapeutically through the infusion of cells after transplant that either specifically or nonspecifically target the tumor. The most common approach is the use of DLI. This approach relies on the persistence of donor T-cell engraftment after transplant to prevent rejection of donor lymphocytes infused to induce the GVL.
Therapeutic DLI results in potent responses in patients with CML who relapse after transplant (60%–80% enter into long-term remission),[89] but responses in patients with other diseases (AML and ALL) have been less potent, with only 20% to 30% long-term survival.[90] DLI works poorly in patients with acute leukemia who relapse early and who have high levels of active disease. Late relapse (>6 months after transplant) and the treatment of patients into complete remission with chemotherapy before DLI have been associated with improved outcomes.[91] Infusions of DLI modified to enhance GVL or other donor cells (natural killer [NK] cells, etc.) have also been studied, but have yet to be generally adopted.
Another method of delivering GVL therapeutically is the rapid withdrawal of immune suppression after HCT. Some studies have scheduled more rapid immune suppression tapers based on donor type (related donors are tapered more quickly than are unrelated donors because of less GVHD risk), and others have used sensitive measures of either low levels of persistent recipient cells (recipient chimerism [from the Greek chimera, a mythical animal with parts from various animals]) or MRD to assess the risk of relapse and trigger rapid taper of immune suppression.
A combination of early withdrawal of immune suppression after HCT with addition of DLI to prevent relapse in patients at high risk of relapse due to persistent/progressive recipient chimerism has been tested in patients who underwent transplant for both ALL and AML.[92][Level of evidence: 2A]; [93][Level of evidence: 3iiDii]
  • ALL: For patients with ALL, one study found increasing recipient chimerism in 46 of 101 patients. Thirty-one of those patients had withdrawal of immune suppression, and a portion went on to receive DLI if GVHD did not occur. This group had a 37% survival rate, compared with 0% in the 15 patients who did not undergo this approach (P < .001).[94]
  • AML: For patients with AML after HCT, about 20% experienced mixed chimerism after HCT and were identified as high risk. Of these, 54% survived if they underwent withdrawal of immune suppression with or without DLI; there were no survivors among those who did not receive this therapy.[95]
Other immunological and cell therapy approaches under evaluation
The role of killer immunoglobulin-like receptor (KIR) mismatching in HCT
Donor-derived NK cells in the post-HCT setting have been shown to promote the following:[96-98]
  • Engraftment.
  • Decreased GVHD.
  • Fewer relapses of hematological malignancies.
  • Improved survival.
NK-cell function is modulated by interactions with a number of receptor families, including activating and inhibiting KIR. The KIR effect in the allogeneic HCT setting hinges on the expression of specific inhibitory KIR on donor-derived NK cells and either the presence or the absence of their matching HLA class I molecules (KIR ligands) on recipient leukemic and normal cells. Normally, the presence of specific KIR ligands interacting with paired inhibitory KIR molecules prevents NK cell attack on healthy cells. In the allogeneic transplant setting, recipient leukemia cells genetically differ from donor NK cells, and they may not have the appropriate inhibitory KIR ligand. Mismatch of ligand and receptor allows NK-cell–based killing of recipient leukemia cells to proceed for certain donor-recipient genetic combinations.
The original observation of decreased relapse with certain KIR-ligand combinations was made in the setting of T-cell–depleted haploidentical transplantation and was strongest after HCT for AML.[97,99] Along with decreasing relapse, these studies have suggested a decrease in GVHD with appropriate KIR-ligand combinations. Many subsequent studies did not detect survival effects for KIR-incompatible HCT using standard transplantation methods,[100,101] which has led to the conclusion that T-cell depletion may be necessary to remove other forms of inhibitory cellular interactions.
Decreased relapse and better survival have been noted with donor/recipient KIR-ligand incompatibility after cord blood HCT, a relatively T-cell–depleted procedure.[102,103] In contrast to this notion, one study demonstrated that some KIR mismatching combinations (activating receptor KIR2DS1 with the HLA C1 ligand) can lead to decreased relapse after AML HCT without T-cell depletion.[104] The role of KIR incompatibility in sibling donor HCT and in diseases other than AML is controversial, but in pediatrics, at least two groups have found better outcomes with specific types of KIR mismatching in ALL.[58,105,106]
A current challenge associated with studies of KIR is that several different approaches have been used to determine what is KIR compatible and incompatible.[99,107] The standardization of classification and prospective studies should help clarify the utility and importance of this approach. Because a limited number of centers perform haploidentical HCT and the results of the data in cord blood HCT are preliminary, most transplant programs do not use KIR mismatching as part of their strategy for choosing a donor. Full HLA matching is considered most important for outcome, with considerations of KIR incompatibility remaining secondary.
NK-cell transplantation
With a low risk of GVHD and demonstrated efficacy in decreasing relapse in post-haploidentical HCT settings, NK-cell infusions as a method of treating high-risk patients and consolidating patients in remission have been studied:
Evidence (NK-cell transplantation outcomes):
  1. The University of Minnesota group initially failed to demonstrate efficacy with autologous NK cells, but found that intense immunoablative therapy followed by purified haploidentical NK cells and interleukin-2 (IL-2) maintenance led to remission in 5 of 19 high-risk AML patients.[108]
  2. Researchers at St. Jude Children’s Research Hospital treated ten intermediate-risk AML patients who had completed chemotherapy and were in remission with lower-dose immunosuppression followed by haploidentical NK-cell infusions and IL-2 for consolidation.[109] Expansion of NK cells was noted in all nine of the KIR-incompatible donor/recipient pairs. All ten children remained in remission at 2 years. A follow-up phase II study is under way, as are many investigations into NK-cell therapy for a number of cancer types.
    Other investigators have used expanded/activated NK cells before and after HCT.[110] One approach that included the culturing of haploidentical NK cells with membrane-bound IL-21 showed marked expansion and high activity. These cells were then infused just before haploidentical HCT, followed by additional infusions on day +7 and +28 after HCT.[110]
  3. Although early survival rates in this high-risk AML cohort are high, multicenter confirmatory studies will be necessary to establish the efficacy of these types of NK-cell approaches.
Chimeric antigen receptor (CAR) T-cell therapy
For T cells to attack cellular targets (viruses or cancer cells), they must bind to class I major histocompatibility complex (MHC) molecules on the surface of the target cells and avoid suppressor signals sent by regulatory T cells and other surface molecule interactions. Gene transfer technologies can modify T cells to express MHC-independent antibody-binding domains (CAR molecules) aimed at specific target proteins on the surface of tumors. To minimize the chance of suppressor mechanisms affecting CAR T-cell function and to create a cytokine milieu conducive to CAR T-cell expansion,[111] lymphodepleting chemotherapy is generally given before CAR T-cell infusions. CAR T-cell–mediated responses are further enhanced by the addition of intracellular costimulatory domains (e.g., CD28, 4-1BB), which cause significant CAR T-cell expansion and may increase the lifespan of these cells in the recipient.[111]
Investigators using this technology have targeted a variety of tumors/surface molecules, but the best-studied example in pediatric patients is CAR T cells aimed at CD19, a surface receptor on B cells. Several groups have reported significant rates of remission (70%–90%) in children and adults with refractory B-cell ALL,[112-115] and several groups have reported persistence of CAR T cells and remission beyond 6 months in most patients studied.[115,116] Early loss of the CAR T cells is associated with relapse, and the best use of this therapy (bridge to transplant vs. definitive therapy) is under study.
Responses have been associated with a significant increase in inflammatory cytokines (termed cytokine release syndrome), which presents as a sepsis-like picture that can be successfully treated with anti–interleukin-6 receptor (IL-6R) therapies (tocilizumab), often in combination with steroids.[117,118] Cytokine release syndrome presents as fever, headache, myalgias, hypotension, capillary leak, hypoxia, and renal dysfunction. Neurotoxicity, including aphasia, altered mental status, and seizures, has also been observed with CAR T-cell therapy and the symptoms usually resolve spontaneously. Central nervous system symptoms have not responded to IL-6R–targeting agents or other approaches. Other CAR T-cell therapy side effects include coagulopathy, hemophagocytic lymphohistiocytosis–like laboratory changes, and cardiac dysfunction. Between 20% and 40% of patients require treatment in the intensive care unit, mostly pressor support, with 10% to 20% of patients requiring intubation and/or dialysis.[112,115,116]
An international trial in children led to FDA approval of tisagenlecleucel for multiply relapsed or refractory CD19-positive B-cell ALL for patients aged 1 to 25 years.[119] Tisagenlecleucel has also been approved for adults with B-cell lymphoma, as has a second agent, axicabtagene ciloleucel.[120,121]

Principles of Allogeneic HCT Preparative Regimens

In the days just before infusion of the stem cell product (bone marrow, peripheral blood stem cells, or cord blood), HCT recipients receive chemotherapy/immunotherapy, sometimes combined with radiation therapy. This is called a preparative regimen, and the original intent of this treatment was to:
  • Create bone marrow space in the recipient for the donor cells to engraft.
  • Suppress the immune system or eliminate the recipient T cells to minimize risks of rejection.
  • Intensely treat cancer (if present) with high doses of active agents, with the intent to overcome therapy resistance.
With the recognition that donor T cells can facilitate engraftment and kill tumors through GVL effects (obviating the need to create bone marrow space and intensely treat cancer), reduced-intensity or minimal-intensity HCT approaches focusing on immune suppression rather than myeloablation have been developed. The resultant lower toxicity associated with these regimens has led to lower rates of transplant-related mortality and an expanded eligibility for allogeneic HCT to older individuals and younger patients with pre-HCT comorbidities that put them at risk of severe toxicity after standard HCT approaches.[122]
The preparative regimens available now vary tremendously in the amount of immunosuppression and myelosuppression that they cause, with the lowest-intensity regimens relying heavily on a strong GVT effect (refer to Figure 3).
ENLARGEChart showing selected preparative regimens frequently used in pediatric HCT categorized by current definitions as non-myeloablative, reduced-intensity, or myeloablative.
Figure 3. Selected preparative regimens frequently used in pediatric HCT categorized by current definitions as nonmyeloablative, reduced-intensity, or myeloablative. Although FLU plus treosulfan and FLU plus busulfan (full-dose) are considered myeloablative approaches, some refer to them as reduced-toxicity regimens.
Although these regimens lead to varying degrees of myelosuppression and immune suppression, they have been grouped clinically into the following three major categories (refer to Figure 4):[123]
  • Myeloablative: Intense approaches that cause irreversible pancytopenia that requires stem cell rescue for restoration of hematopoiesis.
  • Nonmyeloablative: Regimens that cause minimal cytopenias and do not require stem cell support.
  • Reduced-intensity conditioning: Regimens that are of intermediate intensity and do not meet the definitions of nonmyeloablative or myeloablative regimens.
ENLARGEFigure 3; chart shows classification of conditioning regimens based on duration of pancytopenia and requirement for stem cell support; chart shows myeloablative regimens, nonmyeloablative regimens, and reduced intensity regimens.
Figure 4. Classification of conditioning regimens in 3 categories, based on duration of pancytopenia and requirement for stem cell support. Myeloablative regimens (MA) produce irreversible pancytopenia and require stem cell support. Nonmyeloablative regimens (NMA) produce minimal cytopenia and would not require stem cell support. Reduced-intensity regimens (RIC) are regimens which cannot be classified as MA nor NMA. Reprinted from Biology of Blood and Marrow Transplantation, Volume 15 (Issue 12), Andrea Bacigalupo, Karen Ballen, Doug Rizzo, Sergio Giralt, Hillard Lazarus, Vincent Ho, Jane Apperley, Shimon Slavin, Marcelo Pasquini, Brenda M. Sandmaier, John Barrett, Didier Blaise, Robert Lowski, Mary Horowitz, Defining the Intensity of Conditioning Regimens: Working Definitions, Pages 1628-1633, Copyright 2009, with permission from Elsevier.
For a number of years, retrospective studies showed similar outcomes using reduced-intensity and myeloablative approaches.[68,124] However, a Blood and Marrow Transplant Clinical Trials Network (BMT CTN) trial of adults with AML and MDS that randomly assigned patients to receive either myeloablative or reduced-intensity HCT approaches demonstrated the importance of regimen intensity.[125]
  • At 18 months, relapse was markedly higher in the reduced-intensity cohort (48% vs. 13.5%, P < .001).
  • Although treatment-related mortality was higher in the myeloablative arm (16% vs. 4%, P = .002), relapse-free survival was superior in the myeloablative arm (69% vs. 47%, P < .01) and overall survival was higher (76% vs. 68%), with a nonsignificant P value of .07.
With this in mind, the use of reduced-intensity conditioning and nonmyeloablative regimens is well established in older adults who cannot tolerate more intense myeloablative approaches,[126-128] but these approaches have been studied in a limited number of younger patients with malignancies.[129-133] A large Pediatric Blood and Marrow Transplant Consortium study identified patients at high risk of transplant-related mortality with myeloablative regimens (e.g., history of previous myeloablative transplant, severe organ system dysfunction, or active invasive fungal infection) and successfully treated them with a reduced-intensity regimen.[88] Transplant-related mortality was low in this high-risk group, and long-term survival occurred in most patients with minimal or no detectable disease at the time of transplantation. Because the risks of relapse are higher with these approaches, their use in pediatric cancer is currently limited to patients ineligible for myeloablative regimens and is most likely to be successful when patients have achieved MRD-negative remissions.[88]

Establishing donor chimerism

Intense myeloablative approaches almost invariably result in rapid establishment of hematopoiesis derived completely from donor cells upon count recovery within weeks of the transplant. The introduction of reduced-intensity conditioning and nonmyeloablative approaches into HCT practice has resulted in a slower pace of transition to donor hematopoiesis (gradually increasing from partial to full donor hematopoiesis over months) that is sometimes only partial. DNA-based techniques have been established to differentiate donor and recipient hematopoiesis, applying the word chimerism to describe whether all or part of hematopoiesis after HCT is from the donor or recipient.
There are several implications for the pace and extent of donor chimerism eventually achieved by an HCT recipient. For patients receiving reduced-intensity conditioning or nonmyeloablative regimens, rapid progression to full donor chimerism is associated with less relapse but more GVHD.[134] The delayed pace of obtaining full donor chimerism after reduced-intensity regimens has led to late-onset acute GVHD, occurring as long as 6 months to 7 months after HCT (generally within 100 days after myeloablative approaches).[135] A portion of patients achieve stable mixed chimerism of both donor and recipient. Mixed chimerism is associated with more relapse after HCT for malignancies and less GVHD; however, this condition is often advantageous for nonmalignant HCT, where usually only a percentage of normal hematopoiesis is needed to correct the underlying disorder and GVHD is not beneficial.[136] Finally, serially measured decreasing donor chimerism, especially T-cell–specific chimerism, has been associated with increased risk of rejection.[137]
Because of the implications of persistent recipient chimerism, most transplant programs test for chimerism shortly after engraftment and continue testing regularly until stable full donor hematopoiesis has been achieved. Investigators have defined two approaches to treat the increased risks of relapse and rejection associated with increasing recipient chimerism: rapid withdrawal of immune suppression and DLI. (Refer to the Using donor lymphocyte infusions (DLI) or early withdrawal of immune suppression to enhance GVLsection of this summary for more information.) These approaches are frequently used to address this issue, and have been shown in some cases to decrease relapse risk and stop rejection.[94,138,139] The timing of tapers of immune suppression and doses and approaches to the administration of DLI to increase or stabilize donor chimerism vary tremendously among transplant regimens and institutions.


References
  1. Hahn T, McCarthy PL Jr, Hassebroek A, et al.: Significant improvement in survival after allogeneic hematopoietic cell transplantation during a period of significantly increased use, older recipient age, and use of unrelated donors. J Clin Oncol 31 (19): 2437-49, 2013. [PUBMED Abstract]
  2. Horan JT, Logan BR, Agovi-Johnson MA, et al.: Reducing the risk for transplantation-related mortality after allogeneic hematopoietic cell transplantation: how much progress has been made? J Clin Oncol 29 (7): 805-13, 2011. [PUBMED Abstract]
  3. Wood WA, Lee SJ, Brazauskas R, et al.: Survival improvements in adolescents and young adults after myeloablative allogeneic transplantation for acute lymphoblastic leukemia. Biol Blood Marrow Transplant 20 (6): 829-36, 2014. [PUBMED Abstract]
  4. MacMillan ML, Davies SM, Nelson GO, et al.: Twenty years of unrelated donor bone marrow transplantation for pediatric acute leukemia facilitated by the National Marrow Donor Program. Biol Blood Marrow Transplant 14 (9 Suppl): 16-22, 2008. [PUBMED Abstract]
  5. Harvey J, Green A, Cornish J, et al.: Improved survival in matched unrelated donor transplant for childhood ALL since the introduction of high-resolution matching at HLA class I and II. Bone Marrow Transplant 47 (10): 1294-300, 2012. [PUBMED Abstract]
  6. Majhail NS, Chitphakdithai P, Logan B, et al.: Significant improvement in survival after unrelated donor hematopoietic cell transplantation in the recent era. Biol Blood Marrow Transplant 21 (1): 142-50, 2015. [PUBMED Abstract]
  7. Barker JN, Byam CE, Kernan NA, et al.: Availability of cord blood extends allogeneic hematopoietic stem cell transplant access to racial and ethnic minorities. Biol Blood Marrow Transplant 16 (11): 1541-8, 2010. [PUBMED Abstract]
  8. Gragert L, Eapen M, Williams E, et al.: HLA match likelihoods for hematopoietic stem-cell grafts in the U.S. registry. N Engl J Med 371 (4): 339-48, 2014. [PUBMED Abstract]
  9. Woolfrey A, Klein JP, Haagenson M, et al.: HLA-C antigen mismatch is associated with worse outcome in unrelated donor peripheral blood stem cell transplantation. Biol Blood Marrow Transplant 17 (6): 885-92, 2011. [PUBMED Abstract]
  10. Howard CA, Fernandez-Vina MA, Appelbaum FR, et al.: Recommendations for donor human leukocyte antigen assessment and matching for allogeneic stem cell transplantation: consensus opinion of the Blood and Marrow Transplant Clinical Trials Network (BMT CTN). Biol Blood Marrow Transplant 21 (1): 4-7, 2015. [PUBMED Abstract]
  11. Shaw PJ, Kan F, Woo Ahn K, et al.: Outcomes of pediatric bone marrow transplantation for leukemia and myelodysplasia using matched sibling, mismatched related, or matched unrelated donors. Blood 116 (19): 4007-15, 2010. [PUBMED Abstract]
  12. Flomenberg N, Baxter-Lowe LA, Confer D, et al.: Impact of HLA class I and class II high-resolution matching on outcomes of unrelated donor bone marrow transplantation: HLA-C mismatching is associated with a strong adverse effect on transplantation outcome. Blood 104 (7): 1923-30, 2004. [PUBMED Abstract]
  13. Petersdorf EW, Kollman C, Hurley CK, et al.: Effect of HLA class II gene disparity on clinical outcome in unrelated donor hematopoietic cell transplantation for chronic myeloid leukemia: the US National Marrow Donor Program Experience. Blood 98 (10): 2922-9, 2001. [PUBMED Abstract]
  14. Fernandez-Viña MA, Wang T, Lee SJ, et al.: Identification of a permissible HLA mismatch in hematopoietic stem cell transplantation. Blood 123 (8): 1270-8, 2014. [PUBMED Abstract]
  15. Fernández-Viña MA, Klein JP, Haagenson M, et al.: Multiple mismatches at the low expression HLA loci DP, DQ, and DRB3/4/5 associate with adverse outcomes in hematopoietic stem cell transplantation. Blood 121 (22): 4603-10, 2013. [PUBMED Abstract]
  16. Fleischhauer K, Shaw BE, Gooley T, et al.: Effect of T-cell-epitope matching at HLA-DPB1 in recipients of unrelated-donor haemopoietic-cell transplantation: a retrospective study. Lancet Oncol 13 (4): 366-74, 2012. [PUBMED Abstract]
  17. Crocchiolo R, Zino E, Vago L, et al.: Nonpermissive HLA-DPB1 disparity is a significant independent risk factor for mortality after unrelated hematopoietic stem cell transplantation. Blood 114 (7): 1437-44, 2009. [PUBMED Abstract]
  18. Pidala J, Lee SJ, Ahn KW, et al.: Nonpermissive HLA-DPB1 mismatch increases mortality after myeloablative unrelated allogeneic hematopoietic cell transplantation. Blood 124 (16): 2596-606, 2014. [PUBMED Abstract]
  19. Spellman S, Bray R, Rosen-Bronson S, et al.: The detection of donor-directed, HLA-specific alloantibodies in recipients of unrelated hematopoietic cell transplantation is predictive of graft failure. Blood 115 (13): 2704-8, 2010. [PUBMED Abstract]
  20. Ciurea SO, Thall PF, Wang X, et al.: Donor-specific anti-HLA Abs and graft failure in matched unrelated donor hematopoietic stem cell transplantation. Blood 118 (22): 5957-64, 2011. [PUBMED Abstract]
  21. Shaw BE, Mayor NP, Szydlo RM, et al.: Recipient/donor HLA and CMV matching in recipients of T-cell-depleted unrelated donor haematopoietic cell transplants. Bone Marrow Transplant 52 (5): 717-725, 2017. [PUBMED Abstract]
  22. Hurley CK, Woolfrey A, Wang T, et al.: The impact of HLA unidirectional mismatches on the outcome of myeloablative hematopoietic stem cell transplantation with unrelated donors. Blood 121 (23): 4800-6, 2013. [PUBMED Abstract]
  23. Eapen M, Rubinstein P, Zhang MJ, et al.: Outcomes of transplantation of unrelated donor umbilical cord blood and bone marrow in children with acute leukaemia: a comparison study. Lancet 369 (9577): 1947-54, 2007. [PUBMED Abstract]
  24. Eapen M, Klein JP, Ruggeri A, et al.: Impact of allele-level HLA matching on outcomes after myeloablative single unit umbilical cord blood transplantation for hematologic malignancy. Blood 123 (1): 133-40, 2014. [PUBMED Abstract]
  25. Eapen M, Wang T, Veys PA, et al.: Allele-level HLA matching for umbilical cord blood transplantation for non-malignant diseases in children: a retrospective analysis. Lancet Haematol 4 (7): e325-e333, 2017. [PUBMED Abstract]
  26. Takanashi M, Atsuta Y, Fujiwara K, et al.: The impact of anti-HLA antibodies on unrelated cord blood transplantations. Blood 116 (15): 2839-46, 2010. [PUBMED Abstract]
  27. Cutler C, Kim HT, Sun L, et al.: Donor-specific anti-HLA antibodies predict outcome in double umbilical cord blood transplantation. Blood 118 (25): 6691-7, 2011. [PUBMED Abstract]
  28. Rocha V, Spellman S, Zhang MJ, et al.: Effect of HLA-matching recipients to donor noninherited maternal antigens on outcomes after mismatched umbilical cord blood transplantation for hematologic malignancy. Biol Blood Marrow Transplant 18 (12): 1890-6, 2012. [PUBMED Abstract]
  29. van Rood JJ, Stevens CE, Smits J, et al.: Reexposure of cord blood to noninherited maternal HLA antigens improves transplant outcome in hematological malignancies. Proc Natl Acad Sci U S A 106 (47): 19952-7, 2009. [PUBMED Abstract]
  30. Kanda J, Atsuta Y, Wake A, et al.: Impact of the direction of HLA mismatch on transplantation outcomes in single unrelated cord blood transplantation. Biol Blood Marrow Transplant 19 (2): 247-54, 2013. [PUBMED Abstract]
  31. Stevens CE, Carrier C, Carpenter C, et al.: HLA mismatch direction in cord blood transplantation: impact on outcome and implications for cord blood unit selection. Blood 118 (14): 3969-78, 2011. [PUBMED Abstract]
  32. Cunha R, Loiseau P, Ruggeri A, et al.: Impact of HLA mismatch direction on outcomes after umbilical cord blood transplantation for hematological malignant disorders: a retrospective Eurocord-EBMT analysis. Bone Marrow Transplant 49 (1): 24-9, 2014. [PUBMED Abstract]
  33. Barker JN, Rocha V, Scaradavou A: Optimizing unrelated donor cord blood transplantation. Biol Blood Marrow Transplant 15 (1 Suppl): 154-61, 2009. [PUBMED Abstract]
  34. Wagner JE, Barker JN, DeFor TE, et al.: Transplantation of unrelated donor umbilical cord blood in 102 patients with malignant and nonmalignant diseases: influence of CD34 cell dose and HLA disparity on treatment-related mortality and survival. Blood 100 (5): 1611-8, 2002. [PUBMED Abstract]
  35. Rubinstein P, Carrier C, Scaradavou A, et al.: Outcomes among 562 recipients of placental-blood transplants from unrelated donors. N Engl J Med 339 (22): 1565-77, 1998. [PUBMED Abstract]
  36. Barker JN, Weisdorf DJ, DeFor TE, et al.: Transplantation of 2 partially HLA-matched umbilical cord blood units to enhance engraftment in adults with hematologic malignancy. Blood 105 (3): 1343-7, 2005. [PUBMED Abstract]
  37. Michel G, Galambrun C, Sirvent A, et al.: Single- vs double-unit cord blood transplantation for children and young adults with acute leukemia or myelodysplastic syndrome. Blood 127 (26): 3450-7, 2016. [PUBMED Abstract]
  38. Wagner JE Jr, Eapen M, Carter S, et al.: One-unit versus two-unit cord-blood transplantation for hematologic cancers. N Engl J Med 371 (18): 1685-94, 2014. [PUBMED Abstract]
  39. Stiff PJ, Montesinos P, Peled T, et al.: Cohort-Controlled Comparison of Umbilical Cord Blood Transplantation Using Carlecortemcel-L, a Single Progenitor-Enriched Cord Blood, to Double Cord Blood Unit Transplantation. Biol Blood Marrow Transplant : , 2018. [PUBMED Abstract]
  40. Anand S, Thomas S, Hyslop T, et al.: Transplantation of Ex Vivo Expanded Umbilical Cord Blood (NiCord) Decreases Early Infection and Hospitalization. Biol Blood Marrow Transplant 23 (7): 1151-1157, 2017. [PUBMED Abstract]
  41. Horwitz ME, Chao NJ, Rizzieri DA, et al.: Umbilical cord blood expansion with nicotinamide provides long-term multilineage engraftment. J Clin Invest 124 (7): 3121-8, 2014. [PUBMED Abstract]
  42. Wagner JE Jr, Brunstein CG, Boitano AE, et al.: Phase I/II Trial of StemRegenin-1 Expanded Umbilical Cord Blood Hematopoietic Stem Cells Supports Testing as a Stand-Alone Graft. Cell Stem Cell 18 (1): 144-55, 2016. [PUBMED Abstract]
  43. Delaney C, Heimfeld S, Brashem-Stein C, et al.: Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution. Nat Med 16 (2): 232-6, 2010. [PUBMED Abstract]
  44. Bensinger WI, Martin PJ, Storer B, et al.: Transplantation of bone marrow as compared with peripheral-blood cells from HLA-identical relatives in patients with hematologic cancers. N Engl J Med 344 (3): 175-81, 2001. [PUBMED Abstract]
  45. Rocha V, Cornish J, Sievers EL, et al.: Comparison of outcomes of unrelated bone marrow and umbilical cord blood transplants in children with acute leukemia. Blood 97 (10): 2962-71, 2001. [PUBMED Abstract]
  46. Bertaina A, Merli P, Rutella S, et al.: HLA-haploidentical stem cell transplantation after removal of αβ+ T and B cells in children with nonmalignant disorders. Blood 124 (5): 822-6, 2014. [PUBMED Abstract]
  47. Eapen M, Horowitz MM, Klein JP, et al.: Higher mortality after allogeneic peripheral-blood transplantation compared with bone marrow in children and adolescents: the Histocompatibility and Alternate Stem Cell Source Working Committee of the International Bone Marrow Transplant Registry. J Clin Oncol 22 (24): 4872-80, 2004. [PUBMED Abstract]
  48. Shinzato A, Tabuchi K, Atsuta Y, et al.: PBSCT is associated with poorer survival and increased chronic GvHD than BMT in Japanese paediatric patients with acute leukaemia and an HLA-matched sibling donor. Pediatr Blood Cancer 60 (9): 1513-9, 2013. [PUBMED Abstract]
  49. Anasetti C, Logan BR, Lee SJ, et al.: Peripheral-blood stem cells versus bone marrow from unrelated donors. N Engl J Med 367 (16): 1487-96, 2012. [PUBMED Abstract]
  50. Milano F, Gooley T, Wood B, et al.: Cord-Blood Transplantation in Patients with Minimal Residual Disease. N Engl J Med 375 (10): 944-53, 2016. [PUBMED Abstract]
  51. Ruggeri A, Michel G, Dalle JH, et al.: Impact of pretransplant minimal residual disease after cord blood transplantation for childhood acute lymphoblastic leukemia in remission: an Eurocord, PDWP-EBMT analysis. Leukemia 26 (12): 2455-61, 2012. [PUBMED Abstract]
  52. Bachanova V, Burke MJ, Yohe S, et al.: Unrelated cord blood transplantation in adult and pediatric acute lymphoblastic leukemia: effect of minimal residual disease on relapse and survival. Biol Blood Marrow Transplant 18 (6): 963-8, 2012. [PUBMED Abstract]
  53. Sutton R, Shaw PJ, Venn NC, et al.: Persistent MRD before and after allogeneic BMT predicts relapse in children with acute lymphoblastic leukaemia. Br J Haematol 168 (3): 395-404, 2015. [PUBMED Abstract]
  54. Sanchez-Garcia J, Serrano J, Serrano-Lopez J, et al.: Quantification of minimal residual disease levels by flow cytometry at time of transplant predicts outcome after myeloablative allogeneic transplantation in ALL. Bone Marrow Transplant 48 (3): 396-402, 2013. [PUBMED Abstract]
  55. Beatty PG, Clift RA, Mickelson EM, et al.: Marrow transplantation from related donors other than HLA-identical siblings. N Engl J Med 313 (13): 765-71, 1985. [PUBMED Abstract]
  56. Aversa F, Tabilio A, Velardi A, et al.: Treatment of high-risk acute leukemia with T-cell-depleted stem cells from related donors with one fully mismatched HLA haplotype. N Engl J Med 339 (17): 1186-93, 1998. [PUBMED Abstract]
  57. Barrett J, Gluckman E, Handgretinger R, et al.: Point-counterpoint: haploidentical family donors versus cord blood transplantation. Biol Blood Marrow Transplant 17 (1 Suppl): S89-93, 2011. [PUBMED Abstract]
  58. Leung W, Campana D, Yang J, et al.: High success rate of hematopoietic cell transplantation regardless of donor source in children with very high-risk leukemia. Blood 118 (2): 223-30, 2011. [PUBMED Abstract]
  59. González-Vicent M, Molina B, Andión M, et al.: Allogeneic hematopoietic transplantation using haploidentical donor vs. unrelated cord blood donor in pediatric patients: a single-center retrospective study. Eur J Haematol 87 (1): 46-53, 2011. [PUBMED Abstract]
  60. Handgretinger R, Chen X, Pfeiffer M, et al.: Feasibility and outcome of reduced-intensity conditioning in haploidentical transplantation. Ann N Y Acad Sci 1106: 279-89, 2007. [PUBMED Abstract]
  61. Locatelli F, Merli P, Pagliara D, et al.: Outcome of children with acute leukemia given HLA-haploidentical HSCT after αβ T-cell and B-cell depletion. Blood 130 (5): 677-685, 2017. [PUBMED Abstract]
  62. Bertaina A, Zecca M, Buldini B, et al.: Unrelated donor vs HLA-haploidentical alpha/beta T-cell and B-cell depleted HSCT in children with acute leukemia. Blood : , 2018. [PUBMED Abstract]
  63. Leen AM, Christin A, Myers GD, et al.: Cytotoxic T lymphocyte therapy with donor T cells prevents and treats adenovirus and Epstein-Barr virus infections after haploidentical and matched unrelated stem cell transplantation. Blood 114 (19): 4283-92, 2009. [PUBMED Abstract]
  64. 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 38 (4): 291-7, 2006. [PUBMED Abstract]
  65. Luznik L, Fuchs EJ: High-dose, post-transplantation cyclophosphamide to promote graft-host tolerance after allogeneic hematopoietic stem cell transplantation. Immunol Res 47 (1-3): 65-77, 2010. [PUBMED Abstract]
  66. Berger M, Lanino E, Cesaro S, et al.: Feasibility and Outcome of Haploidentical Hematopoietic Stem Cell Transplantation with Post-Transplant High-Dose Cyclophosphamide for Children and Adolescents with Hematologic Malignancies: An AIEOP-GITMO Retrospective Multicenter Study. Biol Blood Marrow Transplant 22 (5): 902-9, 2016. [PUBMED Abstract]
  67. Ciurea SO, Cao K, Fernadez-Vina M, et al.: The European Society for Blood and Marrow Transplantation (EBMT) Consensus Guidelines for the Detection and Treatment of Donor-specific Anti-HLA Antibodies (DSA) in Haploidentical Hematopoietic Cell Transplantation. Bone Marrow Transplant 53 (5): 521-534, 2018. [PUBMED Abstract]
  68. Pulsipher MA, Chitphakdithai P, Logan BR, et al.: Donor, recipient, and transplant characteristics as risk factors after unrelated donor PBSC transplantation: beneficial effects of higher CD34+ cell dose. Blood 114 (13): 2606-16, 2009. [PUBMED Abstract]
  69. Aversa F, Terenzi A, Tabilio A, et al.: Full haplotype-mismatched hematopoietic stem-cell transplantation: a phase II study in patients with acute leukemia at high risk of relapse. J Clin Oncol 23 (15): 3447-54, 2005. [PUBMED Abstract]
  70. Kollman C, Howe CW, Anasetti C, et al.: Donor characteristics as risk factors in recipients after transplantation of bone marrow from unrelated donors: the effect of donor age. Blood 98 (7): 2043-51, 2001. [PUBMED Abstract]
  71. Kollman C, Spellman SR, Zhang MJ, et al.: The effect of donor characteristics on survival after unrelated donor transplantation for hematologic malignancy. Blood 127 (2): 260-7, 2016. [PUBMED Abstract]
  72. Boeckh M, Nichols WG: The impact of cytomegalovirus serostatus of donor and recipient before hematopoietic stem cell transplantation in the era of antiviral prophylaxis and preemptive therapy. Blood 103 (6): 2003-8, 2004. [PUBMED Abstract]
  73. Seebach JD, Stussi G, Passweg JR, et al.: ABO blood group barrier in allogeneic bone marrow transplantation revisited. Biol Blood Marrow Transplant 11 (12): 1006-13, 2005. [PUBMED Abstract]
  74. Logan AC, Wang Z, Alimoghaddam K, et al.: ABO mismatch is associated with increased nonrelapse mortality after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 21 (4): 746-54, 2015. [PUBMED Abstract]
  75. Stussi G, Muntwyler J, Passweg JR, et al.: Consequences of ABO incompatibility in allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 30 (2): 87-93, 2002. [PUBMED Abstract]
  76. Loren AW, Bunin GR, Boudreau C, et al.: Impact of donor and recipient sex and parity on outcomes of HLA-identical sibling allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 12 (7): 758-69, 2006. [PUBMED Abstract]
  77. Canaani J, Savani BN, Labopin M, et al.: ABO incompatibility in mismatched unrelated donor allogeneic hematopoietic cell transplantation for acute myeloid leukemia: A report from the acute leukemia working party of the EBMT. Am J Hematol 92 (8): 789-796, 2017. [PUBMED Abstract]
  78. Stern M, Ruggeri L, Mancusi A, et al.: Survival after T cell-depleted haploidentical stem cell transplantation is improved using the mother as donor. Blood 112 (7): 2990-5, 2008. [PUBMED Abstract]
  79. Ciurea SO, Champlin RE: Donor selection in T cell-replete haploidentical hematopoietic stem cell transplantation: knowns, unknowns, and controversies. Biol Blood Marrow Transplant 19 (2): 180-4, 2013. [PUBMED Abstract]
  80. Wang Y, Chang YJ, Xu LP, et al.: Who is the best donor for a related HLA haplotype-mismatched transplant? Blood 124 (6): 843-50, 2014. [PUBMED Abstract]
  81. Canaani J, Savani BN, Labopin M, et al.: Impact of ABO incompatibility on patients' outcome after haploidentical hematopoietic stem cell transplantation for acute myeloid leukemia - a report from the Acute Leukemia Working Party of the EBMT. Haematologica 102 (6): 1066-1074, 2017. [PUBMED Abstract]
  82. Pulsipher MA, Langholz B, Wall DA, et al.: The addition of sirolimus to tacrolimus/methotrexate GVHD prophylaxis in children with ALL: a phase 3 Children's Oncology Group/Pediatric Blood and Marrow Transplant Consortium trial. Blood 123 (13): 2017-25, 2014. [PUBMED Abstract]
  83. Neudorf S, Sanders J, Kobrinsky N, et al.: Allogeneic bone marrow transplantation for children with acute myelocytic leukemia in first remission demonstrates a role for graft versus leukemia in the maintenance of disease-free survival. Blood 103 (10): 3655-61, 2004. [PUBMED Abstract]
  84. Boyiadzis M, Arora M, Klein JP, et al.: Impact of Chronic Graft-versus-Host Disease on Late Relapse and Survival on 7,489 Patients after Myeloablative Allogeneic Hematopoietic Cell Transplantation for Leukemia. Clin Cancer Res 21 (9): 2020-8, 2015. [PUBMED Abstract]
  85. Woods WG, Neudorf S, Gold S, et al.: A comparison of allogeneic bone marrow transplantation, autologous bone marrow transplantation, and aggressive chemotherapy in children with acute myeloid leukemia in remission. Blood 97 (1): 56-62, 2001. [PUBMED Abstract]
  86. Ribera JM, Ortega JJ, Oriol A, et al.: Comparison of intensive chemotherapy, allogeneic, or autologous stem-cell transplantation as postremission treatment for children with very high risk acute lymphoblastic leukemia: PETHEMA ALL-93 Trial. J Clin Oncol 25 (1): 16-24, 2007. [PUBMED Abstract]
  87. Gross TG, Hale GA, He W, et al.: Hematopoietic stem cell transplantation for refractory or recurrent non-Hodgkin lymphoma in children and adolescents. Biol Blood Marrow Transplant 16 (2): 223-30, 2010. [PUBMED Abstract]
  88. Pulsipher MA, Boucher KM, Wall D, et al.: Reduced-intensity allogeneic transplantation in pediatric patients ineligible for myeloablative therapy: results of the Pediatric Blood and Marrow Transplant Consortium Study ONC0313. Blood 114 (7): 1429-36, 2009. [PUBMED Abstract]
  89. Porter DL, Collins RH Jr, Shpilberg O, et al.: Long-term follow-up of patients who achieved complete remission after donor leukocyte infusions. Biol Blood Marrow Transplant 5 (4): 253-61, 1999. [PUBMED Abstract]
  90. Levine JE, Barrett AJ, Zhang MJ, et al.: Donor leukocyte infusions to treat hematologic malignancy relapse following allo-SCT in a pediatric population. Bone Marrow Transplant 42 (3): 201-5, 2008. [PUBMED Abstract]
  91. Warlick ED, DeFor T, Blazar BR, et al.: Successful remission rates and survival after lymphodepleting chemotherapy and donor lymphocyte infusion for relapsed hematologic malignancies postallogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 18 (3): 480-6, 2012. [PUBMED Abstract]
  92. Horn B, Petrovic A, Wahlstrom J, et al.: Chimerism-based pre-emptive immunotherapy with fast withdrawal of immunosuppression and donor lymphocyte infusions after allogeneic stem cell transplantation for pediatric hematologic malignancies. Biol Blood Marrow Transplant 21 (4): 729-37, 2015. [PUBMED Abstract]
  93. Horn B, Wahlstrom JT, Melton A, et al.: Early mixed chimerism-based preemptive immunotherapy in children undergoing allogeneic hematopoietic stem cell transplantation for acute leukemia. Pediatr Blood Cancer 64 (8): , 2017. [PUBMED Abstract]
  94. Bader P, Kreyenberg H, Hoelle W, et al.: Increasing mixed chimerism is an important prognostic factor for unfavorable outcome in children with acute lymphoblastic leukemia after allogeneic stem-cell transplantation: possible role for pre-emptive immunotherapy? J Clin Oncol 22 (9): 1696-705, 2004. [PUBMED Abstract]
  95. Rettinger E, Willasch AM, Kreyenberg H, et al.: Preemptive immunotherapy in childhood acute myeloid leukemia for patients showing evidence of mixed chimerism after allogeneic stem cell transplantation. Blood 118 (20): 5681-8, 2011. [PUBMED Abstract]
  96. Ruggeri L, Capanni M, Urbani E, et al.: Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 295 (5562): 2097-100, 2002. [PUBMED Abstract]
  97. Giebel S, Locatelli F, Lamparelli T, et al.: Survival advantage with KIR ligand incompatibility in hematopoietic stem cell transplantation from unrelated donors. Blood 102 (3): 814-9, 2003. [PUBMED Abstract]
  98. Bari R, Rujkijyanont P, Sullivan E, et al.: Effect of donor KIR2DL1 allelic polymorphism on the outcome of pediatric allogeneic hematopoietic stem-cell transplantation. J Clin Oncol 31 (30): 3782-90, 2013. [PUBMED Abstract]
  99. Ruggeri L, Mancusi A, Capanni M, et al.: Donor natural killer cell allorecognition of missing self in haploidentical hematopoietic transplantation for acute myeloid leukemia: challenging its predictive value. Blood 110 (1): 433-40, 2007. [PUBMED Abstract]
  100. Davies SM, Ruggieri L, DeFor T, et al.: Evaluation of KIR ligand incompatibility in mismatched unrelated donor hematopoietic transplants. Killer immunoglobulin-like receptor. Blood 100 (10): 3825-7, 2002. [PUBMED Abstract]
  101. Farag SS, Bacigalupo A, Eapen M, et al.: The effect of KIR ligand incompatibility on the outcome of unrelated donor transplantation: a report from the center for international blood and marrow transplant research, the European blood and marrow transplant registry, and the Dutch registry. Biol Blood Marrow Transplant 12 (8): 876-84, 2006. [PUBMED Abstract]
  102. Cooley S, Trachtenberg E, Bergemann TL, et al.: Donors with group B KIR haplotypes improve relapse-free survival after unrelated hematopoietic cell transplantation for acute myelogenous leukemia. Blood 113 (3): 726-32, 2009. [PUBMED Abstract]
  103. Willemze R, Rodrigues CA, Labopin M, et al.: KIR-ligand incompatibility in the graft-versus-host direction improves outcomes after umbilical cord blood transplantation for acute leukemia. Leukemia 23 (3): 492-500, 2009. [PUBMED Abstract]
  104. Venstrom JM, Pittari G, Gooley TA, et al.: HLA-C-dependent prevention of leukemia relapse by donor activating KIR2DS1. N Engl J Med 367 (9): 805-16, 2012. [PUBMED Abstract]
  105. Leung W: Use of NK cell activity in cure by transplant. Br J Haematol 155 (1): 14-29, 2011. [PUBMED Abstract]
  106. Oevermann L, Michaelis SU, Mezger M, et al.: KIR B haplotype donors confer a reduced risk for relapse after haploidentical transplantation in children with ALL. Blood 124 (17): 2744-7, 2014. [PUBMED Abstract]
  107. Leung W, Iyengar R, Triplett B, et al.: Comparison of killer Ig-like receptor genotyping and phenotyping for selection of allogeneic blood stem cell donors. J Immunol 174 (10): 6540-5, 2005. [PUBMED Abstract]
  108. Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al.: Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 105 (8): 3051-7, 2005. [PUBMED Abstract]
  109. Rubnitz JE, Inaba H, Ribeiro RC, et al.: NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J Clin Oncol 28 (6): 955-9, 2010. [PUBMED Abstract]
  110. Ciurea SO, Schafer JR, Bassett R, et al.: Phase 1 clinical trial using mbIL21 ex vivo-expanded donor-derived NK cells after haploidentical transplantation. Blood 130 (16): 1857-1868, 2017. [PUBMED Abstract]
  111. Kalos M, Levine BL, Porter DL, et al.: T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med 3 (95): 95ra73, 2011. [PUBMED Abstract]
  112. Grupp SA, Kalos M, Barrett D, et al.: Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 368 (16): 1509-18, 2013. [PUBMED Abstract]
  113. Lee DW, Kochenderfer JN, Stetler-Stevenson M, et al.: T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 385 (9967): 517-28, 2015. [PUBMED Abstract]
  114. Davila ML, Riviere I, Wang X, et al.: Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 6 (224): 224ra25, 2014. [PUBMED Abstract]
  115. Gardner RA, Finney O, Annesley C, et al.: Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood 129 (25): 3322-3331, 2017. [PUBMED Abstract]
  116. Maude SL, Frey N, Shaw PA, et al.: Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 371 (16): 1507-17, 2014. [PUBMED Abstract]
  117. Lee DW, Gardner R, Porter DL, et al.: Current concepts in the diagnosis and management of cytokine release syndrome. Blood 124 (2): 188-95, 2014. [PUBMED Abstract]
  118. Maude SL, Barrett D, Teachey DT, et al.: Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J 20 (2): 119-22, 2014 Mar-Apr. [PUBMED Abstract]
  119. Maude SL, Laetsch TW, Buechner J, et al.: Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N Engl J Med 378 (5): 439-448, 2018. [PUBMED Abstract]
  120. Chow VA, Shadman M, Gopal AK: Translating anti-CD19 CAR T-Cell therapy into clinical practice for relapsed/refractory diffuse large B-Cell lymphoma. Blood : , 2018. [PUBMED Abstract]
  121. Neelapu SS, Locke FL, Bartlett NL, et al.: Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N Engl J Med 377 (26): 2531-2544, 2017. [PUBMED Abstract]
  122. Deeg HJ, Sandmaier BM: Who is fit for allogeneic transplantation? Blood 116 (23): 4762-70, 2010. [PUBMED Abstract]
  123. Bacigalupo A, Ballen K, Rizzo D, et al.: Defining the intensity of conditioning regimens: working definitions. Biol Blood Marrow Transplant 15 (12): 1628-33, 2009. [PUBMED Abstract]
  124. Luger SM, Ringdén O, Zhang MJ, et al.: Similar outcomes using myeloablative vs reduced-intensity allogeneic transplant preparative regimens for AML or MDS. Bone Marrow Transplant 47 (2): 203-11, 2012. [PUBMED Abstract]
  125. Scott BL, Pasquini MC, Logan BR, et al.: Myeloablative Versus Reduced-Intensity Hematopoietic Cell Transplantation for Acute Myeloid Leukemia and Myelodysplastic Syndromes. J Clin Oncol 35 (11): 1154-1161, 2017. [PUBMED Abstract]
  126. Giralt S, Estey E, Albitar M, et al.: Engraftment of allogeneic hematopoietic progenitor cells with purine analog-containing chemotherapy: harnessing graft-versus-leukemia without myeloablative therapy. Blood 89 (12): 4531-6, 1997. [PUBMED Abstract]
  127. Slavin S, Nagler A, Naparstek E, et al.: Nonmyeloablative stem cell transplantation and cell therapy as an alternative to conventional bone marrow transplantation with lethal cytoreduction for the treatment of malignant and nonmalignant hematologic diseases. Blood 91 (3): 756-63, 1998. [PUBMED Abstract]
  128. Storb R, Yu C, Sandmaier BM, et al.: Mixed hematopoietic chimerism after marrow allografts. Transplantation in the ambulatory care setting. Ann N Y Acad Sci 872: 372-5; discussion 375-6, 1999. [PUBMED Abstract]
  129. Bradley MB, Satwani P, Baldinger L, et al.: Reduced intensity allogeneic umbilical cord blood transplantation in children and adolescent recipients with malignant and non-malignant diseases. Bone Marrow Transplant 40 (7): 621-31, 2007. [PUBMED Abstract]
  130. Del Toro G, Satwani P, Harrison L, et al.: A pilot study of reduced intensity conditioning and allogeneic stem cell transplantation from unrelated cord blood and matched family donors in children and adolescent recipients. Bone Marrow Transplant 33 (6): 613-22, 2004. [PUBMED Abstract]
  131. Gómez-Almaguer D, Ruiz-Argüelles GJ, Tarín-Arzaga Ldel C, et al.: Reduced-intensity stem cell transplantation in children and adolescents: the Mexican experience. Biol Blood Marrow Transplant 9 (3): 157-61, 2003. [PUBMED Abstract]
  132. Pulsipher MA, Woolfrey A: Nonmyeloablative transplantation in children. Current status and future prospects. Hematol Oncol Clin North Am 15 (5): 809-34, vii-viii, 2001. [PUBMED Abstract]
  133. Roman E, Cooney E, Harrison L, et al.: Preliminary results of the safety of immunotherapy with gemtuzumab ozogamicin following reduced intensity allogeneic stem cell transplant in children with CD33+ acute myeloid leukemia. Clin Cancer Res 11 (19 Pt 2): 7164s-7170s, 2005. [PUBMED Abstract]
  134. Baron F, Baker JE, Storb R, et al.: Kinetics of engraftment in patients with hematologic malignancies given allogeneic hematopoietic cell transplantation after nonmyeloablative conditioning. Blood 104 (8): 2254-62, 2004. [PUBMED Abstract]
  135. Vigorito AC, Campregher PV, Storer BE, et al.: Evaluation of NIH consensus criteria for classification of late acute and chronic GVHD. Blood 114 (3): 702-8, 2009. [PUBMED Abstract]
  136. Marsh RA, Vaughn G, Kim MO, et al.: Reduced-intensity conditioning significantly improves survival of patients with hemophagocytic lymphohistiocytosis undergoing allogeneic hematopoietic cell transplantation. Blood 116 (26): 5824-31, 2010. [PUBMED Abstract]
  137. McSweeney PA, Niederwieser D, Shizuru JA, et al.: Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versus-tumor effects. Blood 97 (11): 3390-400, 2001. [PUBMED Abstract]
  138. Horn B, Soni S, Khan S, et al.: Feasibility study of preemptive withdrawal of immunosuppression based on chimerism testing in children undergoing myeloablative allogeneic transplantation for hematologic malignancies. Bone Marrow Transplant 43 (6): 469-76, 2009. [PUBMED Abstract]
  139. Haines HL, Bleesing JJ, Davies SM, et al.: Outcomes of donor lymphocyte infusion for treatment of mixed donor chimerism after a reduced-intensity preparative regimen for pediatric patients with nonmalignant diseases. Biol Blood Marrow Transplant 21 (2): 288-92, 2015. [PUBMED Abstract]

No hay comentarios:

Publicar un comentario