Anemia after allogeneic hematopoietic stem cell transplantation (HSCT) can be immune or non–immune mediated. Auto- or alloimmunity resulting from blood group incompatibility remains an important cause in post-HSCT immune-mediated anemia. ABO incompatibility is commonly encountered in HSCT and may lead to serious clinical complications, including acute hemolysis, pure red cell aplasia, and passenger lymphocyte syndrome. It remains controversial whether ABO incompatibility may affect HSCT outcomes, such as relapse, nonrelapse mortality, graft-versus-host disease, and survival. Non-ABO incompatibility is less frequently encountered but can have similar complications to ABO incompatibility, causing adverse clinical outcomes. It is crucial to identify the driving etiology of post-HSCT anemia in order to prevent and treat this condition. This requires a comprehensive understanding of the mechanism of anemia in blood group–incompatible HSCT and the temporal association between HSCT and anemia. In this review, we summarize the literature on post-HSCT immune-mediated anemia with a focus on ABO and non-ABO blood group incompatibility, describe the underlying mechanism of anemia, and outline preventive and treatment approaches.

There are at least 43 recognized blood groups in humans involving 345 red blood cell (RBC) antigens.1  Approximately 30% to 50% of hematopoietic stem cell transplantations (HSCTs) are performed across the ABO blood group barrier, and clinically significant hemolysis is encountered in 10% to 15% of cases.2,3  Other complications of ABO-mismatched HSCT include hemolytic infusion reactions, delayed engraftment, and pure red cell aplasia (PRCA).4,5  Compared with those of ABO-mismatched HSCT, complications of non-ABO blood group–mismatched HSCT are less well-characterized. Recipients and donors are not routinely checked for non-ABO blood group phenotypes unless clinically indicated, such as when there is a known history of alloantibodies or heavy transfusion requirement in the recipient before transplantation.6  The incidence of non-ABO blood group mismatch–related immunohematologic complications after HSCT is reported to be <10%, which likely reflects underreporting.7-10 

Auto- or alloimmunity against RBCs is an important etiology of immune-mediated anemia in blood group–mismatched HSCT. The source of auto- or alloimmunity may be donor or recipient related, including passive antibody transfusion, passenger lymphocyte syndrome (PLS), alloantibodies formed against engrafting hematopoietic cells, or new auto- or alloantibodies resulting from transfusions (Figures 1 and 2). Antibodies can mediate phagocytosis and cellular toxicity and activate the complement pathway, resulting in intra- and extravascular hemolysis.11  Immune-mediated cytopenia can also occur as a manifestation of chronic graft-versus-host disease (GVHD).12,13  The underlying mechanism of cytopenia in GVHD is unclear, but it is likely to result from altered T-cell immune reconstitution.14 

Figure 1.

Pathophysiology and targeted therapy for post-HSCT immune-mediated anemia. Post-HSCT anemia is multifactorial as a result of new auto- or alloantibodies in combination with T cell– and cytokine-mediated inflammatory processes. There are no formal guidelines in management. Several therapeutic approaches that target different pathophysiologic aspects of post-HSCT immune-mediated anemia are outlined. DLI, donor lymphocyte infusion; IVIG, IV immunoglobulin; Syk, spleen tyrosine kinase.

Figure 1.

Pathophysiology and targeted therapy for post-HSCT immune-mediated anemia. Post-HSCT anemia is multifactorial as a result of new auto- or alloantibodies in combination with T cell– and cytokine-mediated inflammatory processes. There are no formal guidelines in management. Several therapeutic approaches that target different pathophysiologic aspects of post-HSCT immune-mediated anemia are outlined. DLI, donor lymphocyte infusion; IVIG, IV immunoglobulin; Syk, spleen tyrosine kinase.

Close modal

The complexity of post-HSCT immune-mediated anemia requires close collaboration between transfusion medicine, hematology, and transplantation services. Detailed knowledge about recipient/donor blood group phenotypes and the temporal association between transplantation and hemolysis can help identify the underlying mechanism of hemolysis and direct appropriate management. This review summarizes the literature on post-HSCT immune-mediated anemia to provide insight into its pathophysiology, clinical manifestations, and preventive and proposed therapeutic strategies.

ABO mismatch is divided into major, minor, and bidirectional categories (Table 1).

Table 1.

Directions of ABO mismatch

ABO mismatchRecipient phenotypeDonor phenotype
Major OAB A, B, ABAB, BAB, A 
Minor ABAB OOO, A, B 
Bidirectional AB BA 
ABO mismatchRecipient phenotypeDonor phenotype
Major OAB A, B, ABAB, BAB, A 
Minor ABAB OOO, A, B 
Bidirectional AB BA 

Major ABO mismatch

Major ABO mismatch is characterized by the presence of naturally existing host isohemagglutinins against the corresponding carbohydrate ABO antigens of the donor RBCs. The most common scenario is when a group O recipient receives a non-O graft; less commonly, a group A or B recipient may receive a B or A graft, respectively, or an AB graft. The naturally occurring isohemagglutinins in recipients can attack donor RBC ABO antigens early after stem cell infusion or can affect erythroid precursors later in the post-HSCT course.

ABO isohemagglutinins are immunoglobulin M (IgM) antibodies that can robustly activate the classical complement pathway.15  Through formation of the membrane attack complex, major ABO incompatibility may cause acute hemolytic transfusion reactions at the time of stem cell infusion, especially when the graft contains a significant volume of RBCs, such as in unmanipulated bone marrow (BM) grafts.16  Furthermore, recipient isohemagglutinins against donor-derived erythroid precursor cells can lead to reticulocytopenia with subsequent PRCA, which is seen in 8% to 26% of major ABO-mismatched HSCTs.17  PRCA usually occurs ∼30 to 90 days post-HSCT and is characterized by an absence of erythroid precursors in otherwise normal BM.18  Because a majority of recipients clear the donor-specific isohemagglutinins within 120 days, the development of PRCA has been attributed to residual recipient plasma cells that continue to produce isohemagglutinins (Figure 3).4  Patients with PRCA can be transfusion dependent for months, become iron overloaded, and develop alloimmunization to other blood group antigens.19  ABO antigens are also expressed on granulocytes and platelets; therefore, major ABO mismatch can also lead to prolonged neutropenia and thrombocytopenia, resulting in severe infections and catastrophic bleeding.20 

Whether major ABO mismatch can affect HSCT outcomes, such as survival, relapse, and GVHD, is controversial (Table 2). A single-center study of 1502 patients undergoing HSCT with different graft sources, including peripheral blood (PB), BM or cord blood, showed that ABO mismatch was not associated with neutrophil or platelet engraftment delay, incidence of acute or chronic GVHD, OS, or NRM, regardless of graft source.21  A meta-analysis of 7 cohort studies showed that OS was not affected by ABO matching status.22  In contrast, in a Center for International Blood and Marrow Transplant Research study that included 5179 patients, major ABO mismatch was associated with decreased OS (hazard ratio [HR], 1.19; 95% confidence interval [CI], 1.19-1.31; P < .001) and increased NRM (HR, 1.23; 95% CI, 1.08-1.4; P = .002).23  In a European Blood and Marrow Transplant Acute Leukemia Working Group Registry study that included 837 patients who underwent haploidentical HSCT, major ABO mismatch was associated with an inferior day-100 engraftment rate, and in BM HSCT, it was associated with inferior OS.24 

Table 2.

Selected studies evaluating ABO compatibility and transplantation outcomes (2000-2020)

ReferenceYearABO match, n (%)*RIC, n (%)Related donor, n (%)BM graft, n (%)*GVHD prophylaxisEngraftmentGVHD rateRelapse rateNRMOS
16  2000 083 (53)65 (41)10 (6) None All related —62 (74.7)46 (70.8)7 (70.0) CNI + MTX NS No data No data No data No data 
118  2001 2860 (41)1670 (24)1802 (26)587 (8) No data All matched unrelated All BM Various regimens NS NS No data No data NS 
119  2005 2108 (68)451 (21)430 (14)114 (4) 258 (12)46 (10)54 (13)17 (15) All related All BM CNI + MTX Delayed engraftment in major ABO incompatibility NS NS NS NS 
120  2007 121 (56)40 (19)40 (19)15 (7) 0.5% of all patients 86 (71)No data17 (43)No data No data CNI ± MTX; MTX alone NS NS NS NS NS 
121  2008 Major 205 (18.5)Minor 187 (17) All patients 932 (84) of all patients 213 (19) of all patients CNI only in 430 (39%) No data Higher in minor ABO incompatibility No data No data Worse in minor ABO incompatibility 
122  2008 2820 (47)1834 (31)1202 (20)143 (2) 348 (12)152 (8)136 (11)30 (21) No data No data CNI + MTX Delayed engraftment in major ABO incompatibility Higher in major and minor ABO incompatibility NS Higher in major and minor ABO incompatibility Worse in major and minor ABO incompatibility 
43  2009 58 (38)30 (19)44 (29)22 (14) 17 (29)8 (27)15 (34)16 (73) All matched unrelated 27 (47)8 (27)22 (50)9 (41) CNI + MTX/MMF; 10% with CNI only No data Higher acute GVHD rate in minor ABO incompatibility NS NS NS 
23  (Stanford) 2015 1053 (61)297 (17)309 (18)78 (4) 526 (30) of all patients 1303 (75) of all patients 727 (42) of all patients Various No data NS NS Higher in minor ABO incompatibility Worse in minor ABO incompatibility, especially in BM grafts 
CIBMTR(B-cell lymphoma)  240 (59)73 (18)73 (18)22 (5) 238 (55) of all patients 330 (76) of all patients None Various No data NS NS Higher in minor ABO incompatibility Worse in minor ABO incompatibility 
CIBMTR (AML or MDS)  2608 (50)1084 (21)977 (19)311 (6) 1448 (28) of all patients 2079 (40) of all patients 2333 (45) of all patients Various No data NS NS Higher in major ABO incompatibility Worse in major ABO incompatibility 
42  2016 252 (49)105 (21)117 (23)38 (7) 192 (76)78 (74)86 (74)30 (79) 108 (43)25 (24)32 (27)7 (18) None No data Delayed PLT engraftment in major ABO incompatibility NS NS No data NS 
123 (mismatched unrelated donor, AML) 2017 349 (40)215 (25)241 (28)71 (8) 193 (55)122 (57)154 (64)38 (54) None None CNI-based in 87% of all patients NS Lower grade 2-4 acute GVHD rate in major ABO incompatibility NS NS NS 
24 (haploidentical, AML) 2017 522 (63)127 (15)150 (18)38 (5) 215 (41)45 (35)68 (45)12 (32) All haploidentical 279 (53)76 (60)83 (55)19 (55) No data Delayed engraftment in major ABO incompatibility Higher grade 2-4 acute GVHD in bidirectional NS NS Worse in major ABO incompatibility + BM graft 
21  2017 704 (47)324 (22)372 (25)102 (7) 301 (43)128 (40)151 (41)54 (53) 372 (53)101 (31)88 (24)23 (23) 296 (42)87 (27)73 (17)19 (19) No data Delayed neutrophil engraftment only in bidirectional with umbilical cord blood graft NS NS NS NS 
41 (severe aplastic anemia) 2020 114 (57)47 (24)38 (19)0 All patients All haploidentical Both BM and PB in all patients CNI + MMF NS Grade 3-4 acute GVHD more common in minor ABO incompatibility No data No data NS 
124  2020 590 (59.0)164 (16.4)191 (19.1)55 (5.5) 110 (17)32 (20)35 (18)18 (33) 531 (90)124 (76)145 (76)34 (62) 189 (32)33 (20)41 (22)8 (15) CNI + MTX Neutrophil engraftment delayed in mismatched groups NS NS NS NS 
ReferenceYearABO match, n (%)*RIC, n (%)Related donor, n (%)BM graft, n (%)*GVHD prophylaxisEngraftmentGVHD rateRelapse rateNRMOS
16  2000 083 (53)65 (41)10 (6) None All related —62 (74.7)46 (70.8)7 (70.0) CNI + MTX NS No data No data No data No data 
118  2001 2860 (41)1670 (24)1802 (26)587 (8) No data All matched unrelated All BM Various regimens NS NS No data No data NS 
119  2005 2108 (68)451 (21)430 (14)114 (4) 258 (12)46 (10)54 (13)17 (15) All related All BM CNI + MTX Delayed engraftment in major ABO incompatibility NS NS NS NS 
120  2007 121 (56)40 (19)40 (19)15 (7) 0.5% of all patients 86 (71)No data17 (43)No data No data CNI ± MTX; MTX alone NS NS NS NS NS 
121  2008 Major 205 (18.5)Minor 187 (17) All patients 932 (84) of all patients 213 (19) of all patients CNI only in 430 (39%) No data Higher in minor ABO incompatibility No data No data Worse in minor ABO incompatibility 
122  2008 2820 (47)1834 (31)1202 (20)143 (2) 348 (12)152 (8)136 (11)30 (21) No data No data CNI + MTX Delayed engraftment in major ABO incompatibility Higher in major and minor ABO incompatibility NS Higher in major and minor ABO incompatibility Worse in major and minor ABO incompatibility 
43  2009 58 (38)30 (19)44 (29)22 (14) 17 (29)8 (27)15 (34)16 (73) All matched unrelated 27 (47)8 (27)22 (50)9 (41) CNI + MTX/MMF; 10% with CNI only No data Higher acute GVHD rate in minor ABO incompatibility NS NS NS 
23  (Stanford) 2015 1053 (61)297 (17)309 (18)78 (4) 526 (30) of all patients 1303 (75) of all patients 727 (42) of all patients Various No data NS NS Higher in minor ABO incompatibility Worse in minor ABO incompatibility, especially in BM grafts 
CIBMTR(B-cell lymphoma)  240 (59)73 (18)73 (18)22 (5) 238 (55) of all patients 330 (76) of all patients None Various No data NS NS Higher in minor ABO incompatibility Worse in minor ABO incompatibility 
CIBMTR (AML or MDS)  2608 (50)1084 (21)977 (19)311 (6) 1448 (28) of all patients 2079 (40) of all patients 2333 (45) of all patients Various No data NS NS Higher in major ABO incompatibility Worse in major ABO incompatibility 
42  2016 252 (49)105 (21)117 (23)38 (7) 192 (76)78 (74)86 (74)30 (79) 108 (43)25 (24)32 (27)7 (18) None No data Delayed PLT engraftment in major ABO incompatibility NS NS No data NS 
123 (mismatched unrelated donor, AML) 2017 349 (40)215 (25)241 (28)71 (8) 193 (55)122 (57)154 (64)38 (54) None None CNI-based in 87% of all patients NS Lower grade 2-4 acute GVHD rate in major ABO incompatibility NS NS NS 
24 (haploidentical, AML) 2017 522 (63)127 (15)150 (18)38 (5) 215 (41)45 (35)68 (45)12 (32) All haploidentical 279 (53)76 (60)83 (55)19 (55) No data Delayed engraftment in major ABO incompatibility Higher grade 2-4 acute GVHD in bidirectional NS NS Worse in major ABO incompatibility + BM graft 
21  2017 704 (47)324 (22)372 (25)102 (7) 301 (43)128 (40)151 (41)54 (53) 372 (53)101 (31)88 (24)23 (23) 296 (42)87 (27)73 (17)19 (19) No data Delayed neutrophil engraftment only in bidirectional with umbilical cord blood graft NS NS NS NS 
41 (severe aplastic anemia) 2020 114 (57)47 (24)38 (19)0 All patients All haploidentical Both BM and PB in all patients CNI + MMF NS Grade 3-4 acute GVHD more common in minor ABO incompatibility No data No data NS 
124  2020 590 (59.0)164 (16.4)191 (19.1)55 (5.5) 110 (17)32 (20)35 (18)18 (33) 531 (90)124 (76)145 (76)34 (62) 189 (32)33 (20)41 (22)8 (15) CNI + MTX Neutrophil engraftment delayed in mismatched groups NS NS NS NS 

AML, acute myeloid leukemia; CIBMTR, Center for International Blood and Marrow Transplant Research; CNI, calcineurin inhibitor; MDS, myelodysplastic syndrome; MMF, mycophenolate mofetil; MTX, methotrexate; NRM, nonrelapse mortality; NS, not significant; OS, overall survival; PLT, platelets; RIC, reduced-intensity conditioning.

*

Data given in the following order: matched, major, minor, bidirectional.

Patients with ABO mismatch may have lower rates of complications if they receive myeloablative (MA) conditioning.25  One study showed that fludarabine/cyclophosphamide conditioning, compared with a total-body irradiation–based MA regimen, was associated with delayed full donor erythroid chimerism by a median of 74 days and increased risk of PRCA. The clearance of antidonor isohemagglutinins was faster in the latter.25  Other studies have also shown impaired HSCT outcomes in ABO-mismatched HSCTs involving RIC.26,27  ABO mismatch was associated with a 1.7-fold increased risk of extensive chronic GVHD in a cohort of 594 patients who underwent alemtuzumab-based RIC HSCT.27 

Preventive strategies for immunohematologic complications.

Although ABO matching is not required for donor selection per current guidelines, it is worth taking into consideration when multiple suitable donors are available.28  Besides using MA regimens when feasible, strategies to prevent complications in major ABO mismatch include graft RBC reduction and removal of host isohemagglutinins by therapeutic plasma exchange before HSCT.29-31  In recipients with antidonor isohemagglutinin titers ≥1:32, RBC depletion should be performed to ensure infused RBCs <20 mL. When titers are ≤1:16, RBC depletion is generally not required.3 

Because ABO antigens can be secreted into the plasma, pretransplantation immunoabsorption (ie, infusion of donor-type fresh frozen plasma [FFP] positive in A or B antigens to neutralize respective isohemagglutinins) is another approach to prevent complications. In a single-center study in Singapore, 79 of 99 major or bidirectional ABO-mismatched recipients were treated with donor FFP from day −5 to −2 before transplantation, including 70 patients with isohemagglutinin titers >1:32. Those with a titer level ≤1:32 at the time of stem cell infusion did not develop acute hemolysis. Three patients developed PRCA irrespective of pre- or post-FFP isohemagglutinin levels.32  In another single-center study from Australia, 75 of 110 major or bidirectional ABO-mismatched recipients had isohemagglutinin titers >1:32 and received FFP with or without plasma exchange before HSCT. The incidence of PRCA was 5%. The risk of PRCA or delayed RBC engraftment was significantly higher in patients with pretransplantation isohemagglutinin titers ≥1:128.17 

Donor-type RBC transfusion is another immunoabsorption method.33,34  In a single-center study in thalassemia, 20 of 55 major or bidirectional ABO-mismatched recipients with isohemagglutinin titers ≥1:32 received small incremental doses of uncrossmatched donor RBCs during conditioning; 12 had mild hemolysis, and none had severe hemolysis or anaphylaxis. In patients who had no hemolytic reaction at the last transfusion or who had titers <1:32, BM grafts were infused without RBC depletion. No hemolytic reactions were observed post-HSCT, and all patients experienced engraftment without delay.33  Despite these successes, immunoabsorption requires high expertise in transfusion medicine and close patient monitoring, and it remains to be validated in larger studies.

Minor ABO mismatch

Minor ABO mismatch is characterized by the interaction of donor-derived isohemagglutinins with corresponding recipient RBC antigens. These isohemagglutinins may be infused along with the stem cell products, causing acute hemolysis of recipient RBCs. Alternatively, passenger B-lymphocytes from the donor graft may proliferate and secrete isohemagglutinins and other RBC antibodies, causing clinically significantly delayed hemolysis called PLS (Figure 4).35  PLS typically occurs 1 to 2 weeks post-HSCT, more frequently after PB than BM HSCT, because the former contains a higher concentration of lymphocytes.35,36  RIC is associated with a higher risk of PLS because recipient antigens may be left intact and subsequently stimulate donor B cells.37,38  A cyclosporine-only GVHD prophylactic regimen has been associated with higher risks of immunohematologic complications because cyclosporine boosts B-cell antibody production.2,39 

The impact of minor ABO mismatch on HSCT outcomes is also controversial (Table 1). Some studies suggest that minor ABO mismatch is a risk factor for acute GVHD, decreased OS, and increased NRM.23,40-42  One study found it was associated with a threefold increased risk of grade 2 to 4 acute GVHD and fourfold increased risk of grade 3 to 4 acute GVHD.43  In a 1737-patient cohort at Stanford University between 1986 and 2011, minor ABO mismatch was an independent risk factor for OS, with an HR of 1.56 (95% CI, 1.05-2.05; P = .001).23  In a Center for International Blood and Marrow Transplant Research analysis of patients with lymphoma receiving PB grafts, minor ABO mismatch was associated with reduced OS and higher NRM (HR, 1.55 and 1.72; P = .021 and .03, respectively); however, these associations were not observed in patients with acute myeloid leukemia or myelodysplastic syndrome.23 

Preventive strategies for immunohematologic complications.

In minor ABO-mismatched HSCT, immediate hemolysis can be prevented by graft plasma volume reduction; this does not prevent PLS because it does not affect the B-cell component in the graft.44  There is a scarcity of literature on how to prevent PLS. Patients need close monitoring for hemolysis, especially at ∼1 to 2 weeks post-HSCT when PLS may occur. A direct antiglobulin test may assist in early detection of subclinical hemolysis. It is often checked once or twice weekly according to institutional protocols. Maintaining higher hemoglobin levels than the normal transfusion threshold is preferred, especially during the high-risk period, to avoid nadir in case severe or life-threatening PLS occurs. Collaborative efforts between transfusion and transplantation services to ensure the awareness of minor ABO mismatch and transfusion-compatible RBC components peritransplantation is critical.

Bidirectional ABO mismatch

Bidirectional mismatch occurs when the donor/recipient pair has both a major and minor ABO mismatch, such as type A donor/type B recipient or vice versa. The recipient is at risk of complications associated with both directions of incompatibility (Table 1), and preventive methods should address both accordingly.

Unlike ABO isohemagglutinins, which exist in the absence of previous exposure, alloimmunization to protein antigens occurs only after exposure. The incidence of alloimmunization after transfusion is 2% to 6% and most commonly involves the Rhesus (Rh), Kell (K), and Kidd (Jk) blood groups.45-47  RhD antigen is the most potent non-ABO antigen, followed by K antigen. Patients with a history of alloimmunization are at higher risk of developing additional alloantibodies.48  Although rare, non-ABO blood group alloantibodies may result in poor HSCT outcomes.49,50  Complement cascade is implicated in immune-mediated anemia in some of the non-ABO–mismatched HSCT. For example, the IgG or IgM antibodies against Jk antigens are able to fix complements and induce intra- or extravascular hemolysis.51  Mismatch of the Jk antigen system has been reported to cause severe hemolytic anemia or PLS.8,52 

RBC alloimmunization resulting from non-ABO mismatch could originate from the donor, recipient, or both, depending on the temporal relationship between antibody development and chimerism status. Donor-derived alloantibodies include the preexisting antibodies in the graft and new antibodies produced by passenger or engrafted lymphocytes. Recipient-derived alloantibodies include preexisting antibodies and antibodies produced by residual lymphocytes and plasma cells.

Post-HSCT, a majority of the de novo non-ABO alloantibodies develop within the first month.8  PLS can also occur in non-ABO blood group–mismatched HSCT, usually involving RhD, E, s, Jkb, or Jka.52-54 Table 3 outlines previously described clinically significant non-ABO blood group systems associated with alloimmunization after HSCT.

Table 3.

Non-ABO blood group systems with potential to cause clinically significant alloimmunization after HCT

RBC blood group systemAntibodies
Rh Anti-D,8,58,125  anti-C, anti-c,58,126,127  anti-E8,58,125,127,128  
Jk Anti-Jka,8,128  anti-Jkb 
Anti-K,50,58  anti-Jsa58  
MNS Anti-M,50  anti-N,7  anti-S, anti-s8  
Lewis Anti-Lua,129  anti-Leb7,58  
Diego Anti-Dia, anti-Dib128  
Other Anti-McC,58  anti-V,58  anti-Knop58  
RBC blood group systemAntibodies
Rh Anti-D,8,58,125  anti-C, anti-c,58,126,127  anti-E8,58,125,127,128  
Jk Anti-Jka,8,128  anti-Jkb 
Anti-K,50,58  anti-Jsa58  
MNS Anti-M,50  anti-N,7  anti-S, anti-s8  
Lewis Anti-Lua,129  anti-Leb7,58  
Diego Anti-Dia, anti-Dib128  
Other Anti-McC,58  anti-V,58  anti-Knop58  

Patients receiving chronic transfusion therapy before HSCT are generally at higher risk of developing auto- and alloimmunization. Patients with sickle cell disease have high rates of RBC alloimmunization (23.8% to 45.7%) despite leukoreduction and prophylactic antigen matching.55,56  Frequent transfusions, lack of phenotypically matched products, inadequate alloantibody testing, and presence of antigen variations, especially in donors of African descent, increase the risk of alloimmunization.57  In 1 cohort, RBC antibodies were detected in 9 (15%) of 61 patients post-HSCT, including 3 patients with preformed and 6 with new antibodies, and 4 patients developed reticulocytopenia or hemolysis.58  In a cohort of patients undergoing matched-sibling HSCT without MA conditioning, 31% (11 of 31) had a history of RBC alloantibodies, which was correlated with decreased donor T-cell chimerism at 1 year compared with patients without alloimmunization (median, 24% vs 55%; P = .035).59  As the number of HSCTs in sickle cell disease is increasing, it remains a challenge to prevent and treat RBC alloimmunization peri-HSCT.

Thalassemia is another hemoglobinopathy that requires chronic transfusion therapy. Alloantibodies occur in 5% to 30% of patients with thalassemia and are more common after splenectomy, long duration of treatment, and frequent transfusions.60-63  The dominant antibodies are against the Rh and K groups, each comprising 20% to 30% of the antibodies.63  When prophylactically transfusing Rh (D, C, and E) antigen– and K antigen–matched RBC units, alloantibodies were still detected in 32.5% of patients, with an alloimmunization rate of 0.26 antibodies per 100 units, and 72.5% of antibodies were directed against Rh.62  There are limited data on how non-ABO alloantibodies affect HSCT outcomes in patients with thalassemia.

Chronic granulomatous disease can coexist with the McLeod phenotype (ie, reduced expression of K blood group antigens and absence of XK protein on RBCs). This is due to the genetic proximity between the cytochrome B β subunit gene and the XK gene. Hönig et al64  reported a case of successful HSCT in a patient with chronic granulomatous disease/McLeod phenotype with anti-K and anti-Kx alloantibodies. The patient received rituximab, antithymoglobin, MA conditioning, and a K HLA-matched unrelated graft. The patient had an uneventful HSCT course with prompt engraftment. The anti-K alloantibodies remained detectable until 20 months post-HCT.64  Another patient received MA conditioning with busulfan, cyclophosphamide, and antithymoglobin, followed by a K BM graft. He developed refractory severe hemolytic anemia after day +100 as a result of anti-Kx and anti-K and was found to have low donor T-cell chimerism, despite B-cell and erythroid conversion. His hemolytic anemia resolved after 3 DLI.65 

Outside of the chronically transfused population, the rate of non-ABO alloimmunization is reportedly low.7,8,10  De la Rubia et al10  reported a rate of 3.7% (8 of 217) of patients developing new non-ABO alloantibodies post-HCT, 2 of whom developed severe immune hemolytic anemia early after HCT; recipient age and ABO incompatibility were associated with the development of non-ABO alloantibodies. Ting et al7  reported a rate of 8.7% (13 of 150) of patients experiencing new RBC alloantibody production from 12 days to 11 months post-HCT. In another retrospective study, the incidence of alloimmune hemolysis post-HCT involving non-ABO antigens was 1% (5 of 427 patients).8  When alloantibodies are detected promptly, these complications may be prevented.8,66  In 1 case, a recipient with anti-Jka antibodies received fludarabine/melphalan conditioning with rituximab, followed by Jka+ donor PB stem cells without adverse events.66  Complications related to non-ABO antigen incompatibility are expected to increase given the progressive success in HSCT and its wide use in malignant and nonmalignant diseases.67 

Laboratory investigations in non-ABO blood group–mismatched HSCT

Immunohematologic complications caused by non-ABO blood group mismatch can be underdiagnosed because of inadequate phenotyping of the donor-recipient RBCs. In the absence of known preexisting alloantibodies in the recipient, testing beyond the ABO and Rh blood groups is generally not considered before transplantation. Some alloantibodies may be not detected, such as the Jka/Jkb antibodies, because of fluctuation of the titers.8  Jk antibodies are known to have an amnestic response such that a patient may test negative initially but have a subsequent reemergence of antibodies later in the post-HSCT course.

Extended serologic RBC phenotype matching is being performed for patients receiving frequent transfusions in order to provide appropriate antigen-negative blood products. Despite this, alloantibodies can still form and lead to additional challenges in transfusion.68  Extended genotype RBC matching can detect and predict minor variations in RBC antigens that are hard to identify serologically; it is increasingly used in clinical practice.69,70  Real-time polymerase chain reaction is another method for quick RBC phenotyping in HSCT, especially when donor-recipient myeloid chimerism status is dynamically changing.71 

AIHA occurs in 4% to 6% of those undergoing HSCT.72,73  In 1 study of T cell–depleted haploidentical HSCT in patients with severe combined immunodeficiency, the incidence of AIHA was 19.5%.74  Patients post–second HSCT have higher incidence and earlier presentation of AIHA than those post–first HSCT.75  AIHA may occur alone or in conjunction with other immune-mediated cytopenias.73  RBC phenotyping of the donor and recipient, determined before HSCT, can help differentiate autoimmune-mediated anemia from alloimmunity.76  In different reports, median time of onset ranges from 4 to 10 months post-HSCT.75  Transplantation for nonmalignant disease is the most consistent risk factor for post-HSCT AIHA in the literature; other factors, such as unrelated donor, acute or chronic GVHD, cytomegalovirus activation, and alemtuzumab-containing conditioning regimen, have been reported, but reports are inconsistent.72,76-78 

The pathophysiology of AIHA post-HSCT has not been fully elucidated, but it may be related to preformed autoantibodies and dysregulated immune tolerance. In the chronically transfused population, autoantibodies against RBCs have been reported in 7% to 25% of patients; a majority of the autoantibodies are transient, but some may lead to clinically significant hemolysis.79  Patients who develop AIHA are severely lymphopenic, with low numbers of regulatory T cells; delayed T-cell immune reconstitution, resulting from either in vivo or ex vivo T-cell depletion, may allow autoreactive B cells to activate and expand without regulation.74,80  In contrast, flowcytometry and cytokine analysis of patients with AIHA post-HSCT identified a decreased CD3+CD8+ T-cell ratio and a T-helper cell 2–related cytokine profile compared with control patients.76  Patients who develop AIHA are at a higher risk for developing alloimmunity.

Mild to moderate cases of hemolytic anemia, including PLS, are self-limited and are usually treated with simple transfusions of antigen-negative RBCs; however, finding the appropriate RBC unit can be challenging when multiple alloantibodies or antibodies to high-frequency antigens are present.81  Approximately 50% of PRCA cases resolve spontaneously within 100 to 200 days post-HSCT, but in cases of prolonged anemia dependent on chronic frequent transfusions, treatment is necessary to prevent complications such as iron overload.18,82-84 

Most of the therapeutic options for post-HSCT immune-mediated anemia are derived from AIHA, which does not have a licensed treatment itself (Figure 2).80  Based on mechanism of action, the options can be categorized into immunosuppression, which includes the use of systemic steroids, IV immunoglobulin, rituximab, cyclophosphamide, azathioprine, and splenectomy in the severe cases; immunoabsorption; RBC or plasma exchange; and stimulation of erythropoiesis using erythropoietin or thrombopoietin mimetics (Figure 1).85-89  For PRCA, the most commonly used therapies are rituximab, erythropoietin, and DLI (Figure 3).90 

Figure 2.

General approach for posttransplantation immune-mediated hemolysis. The graph outlines underlying mechanisms of posttransplantation hemolysis, timing posttransplantation, preventive methods, and treatment strategies. Note there is no consensus or guideline on how to manage posttransplantation immune-mediated anemia. The management approaches listed are based on expert opinions and available literature. AIHA, autoimmune hemolytic anemia; IVIG, IV immunoglobulin; TPO, thrombopoietin.

Figure 2.

General approach for posttransplantation immune-mediated hemolysis. The graph outlines underlying mechanisms of posttransplantation hemolysis, timing posttransplantation, preventive methods, and treatment strategies. Note there is no consensus or guideline on how to manage posttransplantation immune-mediated anemia. The management approaches listed are based on expert opinions and available literature. AIHA, autoimmune hemolytic anemia; IVIG, IV immunoglobulin; TPO, thrombopoietin.

Close modal
Figure 3.

PRCA. PRCA is more frequently encountered in major ABO mismatch during the first 1 to 3 months posttransplantation. Management approach is similar to PLS, with frequent monitoring and RBC transfusion support. Other pharmacologic interventions can be considered in refractory cases. CBC, complete blood count; Hb, hemoglobin; IVIG, IV immunoglobulin; TMA, thrombotic microangiopathy.

Figure 3.

PRCA. PRCA is more frequently encountered in major ABO mismatch during the first 1 to 3 months posttransplantation. Management approach is similar to PLS, with frequent monitoring and RBC transfusion support. Other pharmacologic interventions can be considered in refractory cases. CBC, complete blood count; Hb, hemoglobin; IVIG, IV immunoglobulin; TMA, thrombotic microangiopathy.

Close modal

There are reports of efficacy using anti-CD20 monoclonal antibodies such as rituximab to treat PRCA.91  Because B cells are responsible for isohemagglutinins or alloantibody production in PLS, rituximab can also be used in severe cases of PLS (Figure 4).92  Other immunosuppressive treatments reported to work in refractory cases include abatacept, a fusion protein that blocks interaction between T cells and antigen-presenting cells.93 

Figure 4.

PLS. Proactive approach with identifying patients at risk before transplantation, alerting the blood bank service, conducting frequent count monitoring, and providing transfusion support in the first 2 weeks posttransplantation is key for better clinical outcomes. Hb, hemoglobin; IVIG, IV immunoglobulin; TMA, thrombotic microangiopathy.

Figure 4.

PLS. Proactive approach with identifying patients at risk before transplantation, alerting the blood bank service, conducting frequent count monitoring, and providing transfusion support in the first 2 weeks posttransplantation is key for better clinical outcomes. Hb, hemoglobin; IVIG, IV immunoglobulin; TMA, thrombotic microangiopathy.

Close modal

Erythropoietin-stimulating agents can increase RBC production and can be used as a therapeutic option in severe hemolytic anemia or PRCA post-HSCT.88,94  Eltrombopag is a thrombopoietin receptor agonist approved for the treatment of aplastic anemia. It has shown efficacy in refractory acquired PRCA outside the HSCT setting.95  Busca et al89  reported 2 cases of PRCA that were refractory to erythropoietin, plasma exchange, rituximab, and bortezomib; these patients achieved sustained PRCA remission with eltrombopag.

DLI and tapering of immunosuppressants represent unique therapeutic options in the HSCT setting. These methods may improve donor chimerism, eliminate recipient plasma cells, and reduce alloreactivity from either the donor or recipient direction. The efficacy of both methods in PRCA is ∼50% in case reports.83  In 1 case report, a patient with PRCA received DLI with a CD34+ stem cell boost and achieved normal blood counts after 2 months.96  However, in a recent multicenter retrospective study, rituximab, DLI, and erythropoietin had no impact on the resolution of PRCA.90  Mesenchymal stem cell infusion has also been used in refractory PRCA cases, with promising results.97,98 

Plasma cells are a viable target for therapy because they may contribute to antibody production, especially in refractory hemolytic anemia and PRCA. Bortezomib was reported to be effective in 40% of cases in the literature.83  Compared with other plasma cell–targeted therapy, daratumumab has a more appealing safety profile and mechanism of action.99,100  A few case reports have noted its efficacy in refractory post-HSCT AIHA and PRCA.101-103  Daratumumab may have efficacy in other forms of post-HCT immune-mediated cytopenia, such as thrombocytopenia or Evan’s syndrome.104  We previously reported durable response after daratumumab in a patient with posttransplantation thrombocytopenia.105  Daratumumab impairs RBC crossmatch testing, which necessitates transfusing K units posttreatment, unless the RBC phenotype is identified pretreatment.106  However, daratumumab is not always effective in post-HSCT immune-mediated cytopenia, likely because of the suppressive effect of daratumumab on CD38+ regulatory T cells.107,108 

A novel modulatory therapy that is worth testing is the Syk inhibitor fostamatinib. Its active metabolite, R406, was shown to reduce antibody-mediated platelet destruction. It was US Food and Drug Administration approved in April 2018 for patients with chronic immune thrombocytopenia for whom 1 line of therapy failed. Additionally, it is currently being studied in a phase 3 clinical trial for patients with warm AIHA (registered at www.clinicaltrials.gov as #NCT03764618) and in an early-phase chronic GVHD trial (#NCT02611063). The Syk pathway is critical for B-cell activation and proliferation; activated B cells may trigger T-cell activation and cytokine production.109,110  Preclinical reports in xenograft models using PB mononuclear cells from patients with active chronic GVHD showed that the Syk inhibitor augments B-cell apoptosis while not affecting normal T-cell function.111 

Therapies targeting the complement pathway are being more frequently used in immune-mediated cytopenia. Vo et al112  used eculizumab, a complement C5 inhibitor, to treat heavily alloimmunized patients with platelet transfusion refractoriness. Four of the 10 patients enrolled overcame platelet transfusion refractoriness with 1 dose of eculizumab. Eculizumab has shown efficacy in warm antibody AIHA and cold agglutinin disease.113,114  In post-HSCT AIHA, eculizumab was effective in 1 of the 3 cases reported in the literature.72  Several other complement inhibitors, such as pegcetacoplan and sutimlimab, are actively being investigated in complement-mediated hemolytic anemia.115,116  These agents may be viable options for refractory hemolytic anemia post-HSCT.

Therapies carry their own risks. Infection is the most concerning complication in patients receiving immunosuppression. In 1 study analyzing the outcome of 46 patients with PRCA, 22 received treatment other than supportive care, and 7 died as a result of infection.18  IV immunoglobulin, rituximab, and plasma exchange may cause anaphylactic reactions. DLI may lead to higher incidence of GVHD.117  Therefore, careful evaluation of the risks and benefits before initiation of therapy is crucial, requiring multidisciplinary collaboration between transfusion medicine, hematology, and HSCT providers.

Donor-recipient RBC antigen mismatch is commonly encountered and results in clinically significant immunohematologic events, complicating the posttransplantation course. ABO phenotype is part of the donor-recipient pretransplantation workup to plan transfusion needs, clinical laboratory monitoring, and possible graft manipulations. Donor-recipient extended phenotyping for non-ABO antigens, such as Rh, K, and Jk, should be considered in high-risk groups, especially those receiving chronic transfusion therapy. HSCT across the barrier of RBC antigen mismatch may lead to acute hemolysis, PLS, and PRCA. Early detection of hemolysis, supportive care with antigen-negative or matched RBC transfusion, and appropriate interventions such as immunosuppression are necessary to improve clinical outcomes. Active collaboration between transfusion and transplantation services might help with early identification of clinically significant antibodies and prompt necessary treatment interventions.

The authors extend their gratitude to the patients reported in this review and thank Charles Bolan for his contribution and review of this manuscript.

Contribution: Y.M. and Y.P. provided conception and design and wrote, reviewed, and/or revised the manuscript; S.S.K. reviewed and revised the manuscript; and S.A. and R.C. wrote and revised the manuscript and provided expert guidance.

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

Correspondence: Yazan Migdady, 3181 SW Sam Jackson Park Rd, Portland, OR 97239; e-mail: migdady@ohsu.edu.

1.
International Society of Blood Transfusion
. Red cell immunogenetics and blood group terminology. Available at: https://www.isbtweb.org/working-parties/red-cell-immunogenetics-and-blood-group-terminology. Accessed 1 October 2021
2.
Hows
J
,
Beddow
K
,
Gordon-Smith
E
, et al
.
Donor-derived red blood cell antibodies and immune hemolysis after allogeneic bone marrow transplantation
.
Blood.
1986
;
67
(
1
):
177
-
181
.
3.
Rowley
SD
,
Donato
ML
,
Bhattacharyya
P
.
Red blood cell-incompatible allogeneic hematopoietic progenitor cell transplantation
.
Bone Marrow Transplant.
2011
;
46
(
9
):
1167
-
1185
.
4.
Griffith
LM
,
McCoy
JP
Jr
,
Bolan
CD
, et al
.
Persistence of recipient plasma cells and anti-donor isohaemagglutinins in patients with delayed donor erythropoiesis after major ABO incompatible non-myeloablative haematopoietic cell transplantation
.
Br J Haematol.
2005
;
128
(
5
):
668
-
675
.
5.
Stussi
G
,
Muntwyler
J
,
Passweg
JR
, et al
.
Consequences of ABO incompatibility in allogeneic hematopoietic stem cell transplantation
.
Bone Marrow Transplant.
2002
;
30
(
2
):
87
-
93
.
6.
Allen
ES
,
Nelson
RC
,
Flegel
WA
.
How we evaluate red blood cell compatibility and transfusion support for patients with sickle cell disease undergoing hematopoietic progenitor cell transplantation
.
Transfusion.
2018
;
58
(
11
):
2483
-
2489
.
7.
Ting
A
,
Pun
A
,
Dodds
AJ
,
Atkinson
K
,
Biggs
JC
.
Red cell alloantibodies produced after bone marrow transplantation
.
Transfusion.
1987
;
27
(
2
):
145
-
147
.
8.
Young
PP
,
Goodnough
LT
,
Westervelt
P
,
Diersio
JF
.
Immune hemolysis involving non-ABO/RhD alloantibodies following hematopoietic stem cell transplantation
.
Bone Marrow Transplant.
2001
;
27
(
12
):
1305
-
1310
.
9.
Abou-Elella
AA
,
Camarillo
TA
,
Allen
MB
, et al
.
Low incidence of red cell and HLA antibody formation by bone marrow transplant patients
.
Transfusion.
1995
;
35
(
11
):
931
-
935
.
10.
de La Rubia
J
,
Arriaga
F
,
Andreu
R
, et al
.
Development of non-ABO RBC alloantibodies in patients undergoing allogeneic HPC transplantation. Is ABO incompatibility a predisposing factor?
Transfusion.
2001
;
41
(
1
):
106
-
110
.
11.
Flegel
WA
.
Pathogenesis and mechanisms of antibody-mediated hemolysis
.
Transfusion.
2015
;
55
(
suppl 2
):
S47
-
S58
.
12.
Filipovich
AH
,
Weisdorf
D
,
Pavletic
S
, et al
.
National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and staging working group report
.
Biol Blood Marrow Transplant.
2005
;
11
(
12
):
945
-
956
.
13.
Couriel
D
,
Carpenter
PA
,
Cutler
C
, et al
.
Ancillary therapy and supportive care of chronic graft-versus-host disease: National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: V. Ancillary Therapy and Supportive Care Working Group report
.
Biol Blood Marrow Transplant.
2006
;
12
(
4
):
375
-
396
.
14.
Szanto
CL
,
Langenhorst
J
,
de Koning
C
, et al
.
Predictors for autoimmune cytopenias after allogeneic hematopoietic cell transplantation in children
.
Biol Blood Marrow Transplant.
2020
;
26
(
1
):
114
-
122
.
15.
Stowell
SR
,
Winkler
AM
,
Maier
CL
, et al
.
Initiation and regulation of complement during hemolytic transfusion reactions
.
Clin Dev Immunol.
2012
;
2012
:
307093
.
16.
Rowley
SD
,
Liang
PS
,
Ulz
L
.
Transplantation of ABO-incompatible bone marrow and peripheral blood stem cell components
.
Bone Marrow Transplant.
2000
;
26
(
7
):
749
-
757
.
17.
Curley
C
,
Pillai
E
,
Mudie
K
, et al
.
Outcomes after major or bidirectional ABO-mismatched allogeneic hematopoietic progenitor cell transplantation after pretransplant isoagglutinin reduction with donor-type secretor plasma with or without plasma exchange
.
Transfusion.
2012
;
52
(
2
):
291
-
297
.
18.
Hirokawa
M
,
Fukuda
T
,
Ohashi
K
, et al;
PRCA Collaborative Study Group
.
Efficacy and long-term outcome of treatment for pure red cell aplasia after allogeneic stem cell transplantation from major ABO-incompatible donors
.
Biol Blood Marrow Transplant.
2013
;
19
(
7
):
1026
-
1032
.
19.
Gmür
JP
,
Burger
J
,
Schaffner
A
, et al
.
Pure red cell aplasia of long duration complicating major ABO-incompatible bone marrow transplantation
.
Blood.
1990
;
75
(
1
):
290
-
295
.
20.
Worel
N
,
Greinix
HT
,
Schneider
B
, et al
.
Regeneration of erythropoiesis after related- and unrelated-donor BMT or peripheral blood HPC transplantation: a major ABO mismatch means problems
.
Transfusion.
2000
;
40
(
5
):
543
-
550
.
21.
Damodar
S
,
Shanley
R
,
MacMillan
M
,
Ustun
C
,
Weisdorf
D
.
Donor-to-recipient ABO mismatch does not impact outcomes of allogeneic hematopoietic cell transplantation regardless of graft source
.
Biol Blood Marrow Transplant.
2017
;
23
(
5
):
795
-
804
.
22.
Kanda
J
,
Ichinohe
T
,
Matsuo
K
, et al
.
Impact of ABO mismatching on the outcomes of allogeneic related and unrelated blood and marrow stem cell transplantations for hematologic malignancies: IPD-based meta-analysis of cohort studies
.
Transfusion.
2009
;
49
(
4
):
624
-
635
.
23.
Logan
AC
,
Wang
Z
,
Alimoghaddam
K
, et al;
Center for International Blood and Marrow Transplantation
.
ABO mismatch is associated with increased nonrelapse mortality after allogeneic hematopoietic cell transplantation
.
Biol Blood Marrow Transplant.
2015
;
21
(
4
):
746
-
754
.
24.
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.
2017
;
102
(
6
):
1066
-
1074
.
25.
Bolan
CD
,
Leitman
SF
,
Griffith
LM
, et al
.
Delayed donor red cell chimerism and pure red cell aplasia following major ABO-incompatible nonmyeloablative hematopoietic stem cell transplantation
.
Blood.
2001
;
98
(
6
):
1687
-
1694
.
26.
Worel
N
,
Kalhs
P
,
Keil
F
, et al
.
ABO mismatch increases transplant-related morbidity and mortality in patients given nonmyeloablative allogeneic HPC transplantation
.
Transfusion.
2003
;
43
(
8
):
1153
-
1161
.
27.
Brierley
CK
,
Littlewood
TJ
,
Peniket
AJ
, et al
.
Impact of ABO blood group mismatch in alemtuzumab-based reduced-intensity conditioned haematopoietic SCT
.
Bone Marrow Transplant.
2015
;
50
(
7
):
931
-
938
.
28.
Dehn
J
,
Spellman
S
,
Hurley
CK
, et al
.
Selection of unrelated donors and cord blood units for hematopoietic cell transplantation: guidelines from the NMDP/CIBMTR
.
Blood.
2019
;
134
(
12
):
924
-
934
.
29.
Buckner
CD
,
Clift
RA
,
Sanders
JE
, et al
.
ABO-incompatible marrow transplants
.
Transplantation.
1978
;
26
(
4
):
233
-
238
.
30.
Bensinger
WI
,
Buckner
CD
,
Thomas
ED
,
Clift
RA
.
ABO-incompatible marrow transplants
.
Transplantation.
1982
;
33
(
4
):
427
-
429
.
31.
Benjamin
RJ
,
McGurk
S
,
Ralston
MS
,
Churchill
WH
,
Antin
JH
.
ABO incompatibility as an adverse risk factor for survival after allogeneic bone marrow transplantation
.
Transfusion.
1999
;
39
(
2
):
179
-
187
.
32.
Quek
J
,
Lee
JJ
,
Lim
FL
, et al
.
Donor-type fresh frozen plasma is effective in preventing hemolytic reaction in major ABO incompatible allogeneic stem cell transplant
.
Transfusion.
2019
;
59
(
1
):
335
-
339
.
33.
Mehta
P
,
Ramprakash
S
,
Raghuram
CP
, et al
.
Pre-transplant donor-type red cell transfusion is a safe and effective strategy to reduce isohemagglutinin titers and prevent donor marrow infusion reactions in major ABO-mismatched transplants
.
Ann Hematol.
2021
;
100
(
8
):
2071
-
2078
.
34.
Scholl
S
,
Klink
A
,
Mügge
LO
,
Schilling
K
,
Höffken
K
,
Sayer
HG
.
Safety and impact of donor-type red blood cell transfusion before allogeneic peripheral blood progenitor cell transplantation with major ABO mismatch
.
Transfusion.
2005
;
45
(
10
):
1676
-
1683
.
35.
Bolan
CD
,
Childs
RW
,
Procter
JL
,
Barrett
AJ
,
Leitman
SF
.
Massive immune haemolysis after allogeneic peripheral blood stem cell transplantation with minor ABO incompatibility
.
Br J Haematol.
2001
;
112
(
3
):
787
-
795
.
36.
Toren
A
,
Dacosta
Y
,
Manny
N
,
Varadi
G
,
Or
R
,
Nagler
A
.
Passenger B-lymphocyte-induced severe hemolytic disease after allogeneic peripheral blood stem cell transplantation
.
Blood.
1996
;
87
(
2
):
843
-
844
.
37.
Noborio
K
,
Muroi
K
,
Izumi
T
, et al
.
Massive immune hemolysis after non-myeloablative allogeneic peripheral blood stem cell transplantation with minor ABO-incompatibility
.
Leuk Lymphoma.
2003
;
44
(
2
):
357
-
359
.
38.
Worel
N
,
Greinix
HT
,
Keil
F
, et al
.
Severe immune hemolysis after minor ABO-mismatched allogeneic peripheral blood progenitor cell transplantation occurs more frequently after nonmyeloablative than myeloablative conditioning
.
Transfusion.
2002
;
42
(
10
):
1293
-
1301
.
39.
Rosenthal
GJ
,
Weigand
GW
,
Germolec
DR
,
Blank
JA
,
Luster
MI
.
Suppression of B cell function by methotrexate and trimetrexate. Evidence for inhibition of purine biosynthesis as a major mechanism of action
.
J Immunol.
1988
;
141
(
2
):
410
-
416
.
40.
Vaezi
M
,
Oulad Dameshghi
D
,
Souri
M
,
Setarehdan
SA
,
Alimoghaddam
K
,
Ghavamzadeh
A
.
ABO incompatibility and hematopoietic stem cell transplantation outcomes
.
Int J Hematol Oncol Stem Cell Res.
2017
;
11
(
2
):
139
-
147
.
41.
Ma
YR
,
Wang
WJ
,
Cheng
YF
, et al
.
Impact of ABO incompatibility on outcomes after haploidentical hematopoietic stem cell transplantation for severe aplastic anemia
.
Bone Marrow Transplant.
2020
;
55
(
6
):
1068
-
1075
.
42.
Grube
M
,
Wolff
D
,
Ahrens
N
,
Herzberg
PY
,
Herr
W
,
Holler
E
.
ABO blood group antigen mismatch has an impact on outcome after allogeneic peripheral blood stem cell transplantation
.
Clin Transplant.
2016
;
30
(
11
):
1457
-
1465
.
43.
Ludajic
K
,
Balavarca
Y
,
Bickeböller
H
, et al
.
Minor ABO-mismatches are risk factors for acute graft-versus-host disease in hematopoietic stem cell transplant patients
.
Biol Blood Marrow Transplant.
2009
;
15
(
11
):
1400
-
1406
.
44.
Booth
GS
,
Gehrie
EA
,
Bolan
CD
,
Savani
BN
.
Clinical guide to ABO-incompatible allogeneic stem cell transplantation
.
Biol Blood Marrow Transplant.
2013
;
19
(
8
):
1152
-
1158
.
45.
Heddle
NM
,
Soutar
RL
,
O’Hoski
PL
, et al
.
A prospective study to determine the frequency and clinical significance of alloimmunization post-transfusion
.
Br J Haematol.
1995
;
91
(
4
):
1000
-
1005
.
46.
Hoeltge
GA
,
Domen
RE
,
Rybicki
LA
,
Schaffer
PA
.
Multiple red cell transfusions and alloimmunization. Experience with 6996 antibodies detected in a total of 159,262 patients from 1985 to 1993
.
Arch Pathol Lab Med.
1995
;
119
(
1
):
42
-
45
.
47.
Seyfried
H
,
Walewska
I
.
Analysis of immune response to red blood cell antigens in multitransfused patients with different diseases
.
Mater Med Pol.
1990
;
22
(
1
):
21
-
25
.
48.
Higgins
JM
,
Sloan
SR
.
Stochastic modeling of human RBC alloimmunization: evidence for a distinct population of immunologic responders
.
Blood.
2008
;
112
(
6
):
2546
-
2553
.
49.
Klumpp
TR
.
Immunohematologic complications of bone marrow transplantation
.
Bone Marrow Transplant.
1991
;
8
(
3
):
159
-
170
.
50.
López
A
,
de la Rubia
J
,
Arriaga
F
, et al
.
Severe hemolytic anemia due to multiple red cell alloantibodies after an ABO-incompatible allogeneic bone marrow transplant
.
Transfusion.
1998
;
38
(
3
):
247
-
251
.
51.
Kay
B
,
Poisson
JL
,
Tuma
CW
,
Shulman
IA
.
Anti-Jka that are detected by solid-phase red blood cell adherence but missed by gel testing can cause hemolytic transfusion reactions
.
Transfusion.
2016
;
56
(
12
):
2973
-
2979
.
52.
Leo
A
,
Mytilineos
J
,
Voso
MT
, et al
.
Passenger lymphocyte syndrome with severe hemolytic anemia due to an anti-Jk(a) after allogeneic PBPC transplantation
.
Transfusion.
2000
;
40
(
6
):
632
-
636
.
53.
Petz
LD
.
Immune hemolysis associated with transplantation
.
Semin Hematol.
2005
;
42
(
3
):
145
-
155
.
54.
Franchini
M
,
Gandini
G
,
Aprili
G
.
Non-ABO red blood cell alloantibodies following allogeneic hematopoietic stem cell transplantation
.
Bone Marrow Transplant.
2004
;
33
(
12
):
1169
-
1172
.
55.
Campbell-Lee
SA
,
Gvozdjan
K
,
Choi
KM
, et al
.
Red blood cell alloimmunization in sickle cell disease: assessment of transfusion protocols during two time periods
.
Transfusion.
2018
;
58
(
7
):
1588
-
1596
.
56.
Hindawi
S
,
Badawi
M
,
Elfayoumi
R
, et al
.
The value of transfusion of phenotyped blood units for thalassemia and sickle cell anemia patients at an academic center
.
Transfusion.
2020
;
60
(
suppl 1
):
S15
-
S21
.
57.
Chou
ST
,
Jackson
T
,
Vege
S
,
Smith-Whitley
K
,
Friedman
DF
,
Westhoff
CM
.
High prevalence of red blood cell alloimmunization in sickle cell disease despite transfusion from Rh-matched minority donors
.
Blood.
2013
;
122
(
6
):
1062
-
1071
.
58.
Allen
ES
,
Srivastava
K
,
Hsieh
MM
, et al
.
Immunohaematological complications in patients with sickle cell disease after haemopoietic progenitor cell transplantation: a prospective, single-centre, observational study
.
Lancet Haematol.
2017
;
4
(
11
):
e553
-
e561
.
59.
Nickel
RS
,
Flegel
WA
,
Adams
SD
, et al
.
The impact of pre-existing HLA and red blood cell antibodies on transfusion support and engraftment in sickle cell disease after nonmyeloablative hematopoietic stem cell transplantation from HLA-matched sibling donors: a prospective, single-center, observational study
.
EClinicalMedicine.
2020
;
24
:
100432
.
60.
Thompson
AA
,
Cunningham
MJ
,
Singer
ST
, et al;
Thalassemia Clinical Research Network Investigators
.
Red cell alloimmunization in a diverse population of transfused patients with thalassaemia
.
Br J Haematol.
2011
;
153
(
1
):
121
-
128
.
61.
Datta
SS
,
Mukherjee
S
,
Talukder
B
,
Bhattacharya
P
,
Mukherjee
K
.
Frequency of red cell alloimmunization and autoimmunization in thalassemia patients: a report from eastern India
.
Adv Hematol.
2015
;
2015
:
610931
.
62.
Waldis
SJ
,
Uter
S
,
Kavitsky
D
, et al
.
Rh alloimmunization in chronically transfused patients with thalassemia receiving RhD, C, E, and K matched transfusions
.
Blood Adv.
2021
;
5
(
3
):
737
-
744
.
63.
El-Beshlawy
A
,
Salama
AA
,
El-Masry
MR
,
El Husseiny
NM
,
Abdelhameed
AM
.
A study of red blood cell alloimmunization and autoimmunization among 200 multitransfused Egyptian β thalassemia patients
.
Sci Rep.
2020
;
10
(
1
):
21079
.
64.
Hönig
M
,
Flegel
WA
,
Schwarz
K
, et al
.
Successful hematopoietic stem-cell transplantation in a patient with chronic granulomatous disease and McLeod phenotype sensitized to Kx and K antigens
.
Bone Marrow Transplant.
2010
;
45
(
1
):
209
-
211
.
65.
Kordes
U
,
Binder
TM
,
Eiermann
TH
, et al
.
Successful donor-lymphocyte infusion for extreme immune-hemolysis following unrelated BMT in a patient with X-linked chronic granulomatous disease and McLeod phenotype
.
Bone Marrow Transplant.
2008
;
42
(
3
):
219
-
220
.
66.
Kim
MY
,
Chaudhary
P
,
Shulman
IA
,
Pullarkat
V
.
Major non-ABO incompatibility caused by anti-Jk(a) in a patient before allogeneic hematopoietic stem cell transplantation
.
Immunohematology.
2013
;
29
(
1
):
11
-
14
.
67.
Passweg
JR
,
Baldomero
H
,
Bader
P
, et al;
European Society for Blood and Marrow Transplantation (EBMT)
.
Hematopoietic SCT in Europe 2013: recent trends in the use of alternative donors showing more haploidentical donors but fewer cord blood transplants
.
Bone Marrow Transplant.
2015
;
50
(
4
):
476
-
482
.
68.
Lasalle-Williams
M
,
Nuss
R
,
Le
T
, et al
.
Extended red blood cell antigen matching for transfusions in sickle cell disease: a review of a 14-year experience from a single center (CME)
.
Transfusion.
2011
;
51
(
8
):
1732
-
1739
.
69.
Casas
J
,
Friedman
DF
,
Jackson
T
,
Vege
S
,
Westhoff
CM
,
Chou
ST
.
Changing practice: red blood cell typing by molecular methods for patients with sickle cell disease
.
Transfusion.
2015
;
55
(
6 Pt 2
):
1388
-
1393
.
70.
Fasano
RM
,
Chou
ST
.
Red blood cell antigen genotyping for sickle cell disease, thalassemia, and other transfusion complications
.
Transfus Med Rev.
2016
;
30
(
4
):
197
-
201
.
71.
Liu
F
,
Li
G
,
Mao
X
,
Hu
L
.
ABO chimerism determined by real-time polymerase chain reaction analysis after ABO-incompatible haematopoietic stem cell transplantation
.
Blood Transfus.
2013
;
11
(
1
):
43
-
52
.
72.
Barcellini
W
,
Fattizzo
B
,
Zaninoni
A
.
Management of refractory autoimmune hemolytic anemia after allogeneic hematopoietic stem cell transplantation: current perspectives
.
J Blood Med.
2019
;
10
:
265
-
278
.
73.
Miller
PDE
,
Snowden
JA
,
De Latour
RP
, et al
.
Autoimmune cytopenias (AIC) following allogeneic haematopoietic stem cell transplant for acquired aplastic anaemia: a joint study of the Autoimmune Diseases and Severe Aplastic Anaemia Working Parties (ADWP/SAAWP) of the European Society for Blood and Marrow Transplantation (EBMT)
.
Bone Marrow Transplant.
2020
;
55
(
2
):
441
-
451
.
74.
Horn
B
,
Viele
M
,
Mentzer
W
,
Mogck
N
,
DeSantes
K
,
Cowan
M
.
Autoimmune hemolytic anemia in patients with SCID after T cell-depleted BM and PBSC transplantation
.
Bone Marrow Transplant.
1999
;
24
(
9
):
1009
-
1013
.
75.
Ahmed
I
,
Teruya
J
,
Murray-Krezan
C
,
Krance
R
.
The incidence of autoimmune hemolytic anemia in pediatric hematopoietic stem cell recipients post-first and post-second hematopoietic stem cell transplant
.
Pediatr Transplant.
2015
;
19
(
4
):
391
-
398
.
76.
Kruizinga
MD
,
van Tol
MJD
,
Bekker
V
, et al
.
Risk factors, treatment, and immune dysregulation in autoimmune cytopenia after allogeneic hematopoietic stem cell transplantation in pediatric patients
.
Biol Blood Marrow Transplant.
2018
;
24
(
4
):
772
-
778
.
77.
O’Brien
TA
,
Eastlund
T
,
Peters
C
, et al
.
Autoimmune haemolytic anaemia complicating haematopoietic cell transplantation in paediatric patients: high incidence and significant mortality in unrelated donor transplants for non-malignant diseases
.
Br J Haematol.
2004
;
127
(
1
):
67
-
75
.
78.
Faraci
M
,
Zecca
M
,
Pillon
M
, et al;
Italian Association of Paediatric Haematology and Oncology
.
Autoimmune hematological diseases after allogeneic hematopoietic stem cell transplantation in children: an Italian multicenter experience
.
Biol Blood Marrow Transplant.
2014
;
20
(
2
):
272
-
278
.
79.
Garratty
G
.
Autoantibodies induced by blood transfusion
.
Transfusion.
2004
;
44
(
1
):
5
-
9
.
80.
González-Vicent
M
,
Sanz
J
,
Fuster
JL
, et al
.
Autoimmune hemolytic anemia (AIHA) following allogeneic hematopoietic stem cell transplantation (HSCT): a retrospective analysis and a proposal of treatment on behalf of the Grupo Español De Trasplante de Medula Osea en Niños (GETMON) and the Grupo Español de Trasplante Hematopoyetico (GETH) [published online ahead of print on 3 March 2018]
.
Transfus Med Rev.
doi: 10.1016/j.tmrv.2018.02.005.
81.
Adams
BR
,
Miller
AN
,
Costa
LJ
.
Self-limited hemolysis due to anti-D passenger lymphocyte syndrome in allogeneic hematopoietic stem cell transplantation
.
Bone Marrow Transplant.
2010
;
45
(
4
):
772
-
773
.
82.
Aung
FM
,
Lichtiger
B
,
Bassett
R
, et al
.
Incidence and natural history of pure red cell aplasia in major ABO-mismatched haematopoietic cell transplantation
.
Br J Haematol.
2013
;
160
(
6
):
798
-
805
.
83.
Marco-Ayala
J
,
Gómez-Seguí
I
,
Sanz
G
,
Solves
P
.
Pure red cell aplasia after major or bidirectional ABO incompatible hematopoietic stem cell transplantation: to treat or not to treat, that is the question
.
Bone Marrow Transplant.
2021
;
56
(
4
):
769
-
778
.
84.
Stussi
G
,
Halter
J
,
Bucheli
E
, et al
.
Prevention of pure red cell aplasia after major or bidirectional ABO blood group incompatible hematopoietic stem cell transplantation by pretransplant reduction of host anti-donor isoagglutinins
.
Haematologica.
2009
;
94
(
2
):
239
-
248
.
85.
Holbro
A
,
Passweg
JR
.
Management of hemolytic anemia following allogeneic stem cell transplantation
.
Hematology Am Soc Hematol Educ Program.
2015
;
2015
:
378
-
384
.
86.
Hill
QA
,
Stamps
R
,
Massey
E
,
Grainger
JD
,
Provan
D
,
Hill
A
;
British Society for Haematology Guidelines
.
Guidelines on the management of drug-induced immune and secondary autoimmune, haemolytic anaemia
.
Br J Haematol.
2017
;
177
(
2
):
208
-
220
.
87.
Silva
VA
,
Seder
RH
,
Weintraub
LR
.
Synchronization of plasma exchange and cyclophosphamide in severe and refractory autoimmune hemolytic anemia
.
J Clin Apher.
1994
;
9
(
2
):
120
-
123
.
88.
Santamaría
A
,
Sureda
A
,
Martino
R
,
Domingo-Albós
A
,
Muñiz-Díaz
E
,
Brunet
S
.
Successful treatment of pure red cell aplasia after major ABO-incompatible T cell-depleted bone marrow transplantation with erythropoietin
.
Bone Marrow Transplant.
1997
;
20
(
12
):
1105
-
1107
.
89.
Busca
A
,
Dellacasa
C
,
Giaccone
L
, et al
.
Eltrombopag for the treatment of refractory pure RBC aplasia after major ABO incompatible hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant.
2018
;
24
(
8
):
1765
-
1770
.
90.
Longval
T
,
Galimard
JE
,
Leprêtre
AC
, et al
.
Treatment for pure red cell aplasia after major ABO-incompatible allogeneic stem cell transplantation: a multicentre study
.
Br J Haematol.
2021
;
193
(
4
):
814
-
826
.
91.
Helbig
G
,
Stella-Holowiecka
B
,
Krawczyk-Kulis
M
, et al
.
Successful treatment of pure red cell aplasia with repeated, low doses of rituximab in two patients after ABO-incompatible allogeneic haematopoietic stem cell transplantation for acute myeloid leukaemia
.
Haematologica.
2005
;
90
(
suppl
):
ECR33
.
92.
Audet
M
,
Panaro
F
,
Piardi
T
, et al
.
Passenger lymphocyte syndrome and liver transplantation
.
Clin Dev Immunol.
2008
;
2008
:
715769
.
93.
Hess
J
,
Su
L
,
Nizzi
F
, et al
.
Successful treatment of severe refractory autoimmune hemolytic anemia after hematopoietic stem cell transplant with abatacept
.
Transfusion.
2018
;
58
(
9
):
2122
-
2127
.
94.
Taniguchi
S
,
Yamasaki
K
,
Shibuya
T
,
Asayama
R
,
Harada
M
,
Niho
Y
.
Recombinant human erythropoietin for long-term persistent anemia after major ABO-incompatible bone marrow transplantation
.
Bone Marrow Transplant.
1993
;
12
(
4
):
423
.
95.
Liu
X
,
Cheng
L
,
He
Y
, et al
.
Eltrombopag restores erythropoiesis in refractory adult acquired pure red cell aplasia
.
Int J Hematol.
2021
;
114
(
1
):
124
-
128
.
96.
Selleri
C
,
Raiola
A
,
De Rosa
G
, et al
.
CD34+-enriched donor lymphocyte infusions in a case of pure red cell aplasia and late graft failure after major ABO-incompatible bone marrow transplantation
.
Bone Marrow Transplant.
1998
;
22
(
6
):
605
-
607
.
97.
Fang
B
,
Song
Y
,
Li
N
,
Li
J
,
Han
Q
,
Zhao
RC
.
Mesenchymal stem cells for the treatment of refractory pure red cell aplasia after major ABO-incompatible hematopoietic stem cell transplantation
.
Ann Hematol.
2009
;
88
(
3
):
261
-
266
.
98.
Sergeevicheva
V
,
Kruchkova
I
,
Chernykh
E
, et al
.
Rapid recovery from chronic PRCA by MSC infusion in patient after major ABO-mismatched alloSCT
.
Case Rep Med.
2012
;
2012
:
862721
.
99.
Sackett
K
,
Cohn
CS
,
Fahey-Ahrndt
K
,
Smith
AR
,
Johnson
AD
.
Successful treatment of pure red cell aplasia because of ABO major mismatched stem cell transplant
.
J Clin Apher.
2018
;
33
(
1
):
108
-
112
.
100.
Poon
LM
,
Koh
LP
.
Successful treatment of isohemagglutinin-mediated pure red cell aplasia after ABO-mismatched allogeneic hematopoietic cell transplant using bortezomib
.
Bone Marrow Transplant.
2012
;
47
(
6
):
870
-
871
.
101.
Tolbert
VP
,
Goldsby
R
,
Huang
J
, et al
.
Daratumumab is effective in the treatment of refractory post-transplant autoimmune hemolytic anemia: a pediatric case report [abstract]
.
Blood.
2016
;
128
(
22
):
XXX
. Abstract 128.
102.
Chapuy
CI
,
Kaufman
RM
,
Alyea
EP
,
Connors
JM
.
Daratumumab for delayed red-cell engraftment after allogeneic transplantation
.
N Engl J Med.
2018
;
379
(
19
):
1846
-
1850
.
103.
Schuetz
C
,
Hoenig
M
,
Moshous
D
, et al
.
Daratumumab in life-threatening autoimmune hemolytic anemia following hematopoietic stem cell transplantation
.
Blood Adv.
2018
;
2
(
19
):
2550
-
2553
.
104.
Blennerhassett
R
,
Sudini
L
,
Gottlieb
D
,
Bhattacharyya
A
.
Post-allogeneic transplant Evans syndrome successfully treated with daratumumab
.
Br J Haematol.
2019
;
187
(
2
):
e48
-
e51
.
105.
Migdady
Y
,
Ediriwickrema
A
,
Jackson
RP
, et al
.
Successful treatment of thrombocytopenia with daratumumab after allogeneic transplant: a case report and literature review
.
Blood Adv.
2020
;
4
(
5
):
815
-
818
.
106.
Chapuy
CI
,
Nicholson
RT
,
Aguad
MD
, et al
.
Resolving the daratumumab interference with blood compatibility testing
.
Transfusion.
2015
;
55
(
6 Pt 2
):
1545
-
1554
.
107.
van de Donk
NWCJ
.
Reprint of “Immunomodulatory effects of CD38-targeting antibodies”
.
Immunol Lett.
2019
;
205
:
71
-
77
.
108.
Krejcik
J
,
Casneuf
T
,
Nijhof
IS
, et al
.
Daratumumab depletes CD38+ immune regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma
.
Blood.
2016
;
128
(
3
):
384
-
394
.
109.
Mócsai
A
,
Ruland
J
,
Tybulewicz
VL
.
The SYK tyrosine kinase: a crucial player in diverse biological functions
.
Nat Rev Immunol.
2010
;
10
(
6
):
387
-
402
.
110.
Kaur
M
,
Singh
M
,
Silakari
O
.
Inhibitors of switch kinase ‘spleen tyrosine kinase’ in inflammation and immune-mediated disorders: a review
.
Eur J Med Chem.
2013
;
67
:
434
-
446
.
111.
Poe
JC
,
Jia
W
,
Di Paolo
JA
, et al
.
SYK inhibitor entospletinib prevents ocular and skin GVHD in mice
.
JCI Insight.
2018
;
3
(
19
):
e122430
.
112.
Vo
P
,
Purev
E
,
West
KA
, et al
.
A pilot trial of complement inhibition using eculizumab to overcome platelet transfusion refractoriness in human leukocyte antigen allo-immunized patients
.
Br J Haematol.
2020
;
189
(
3
):
551
-
558
.
113.
Gauchy
AC
,
Hentzien
M
,
Wynckel
A
, et al
.
Efficacy of eculizumab in refractory life-threatening warm autoimmune hemolytic anemia associated with chronic myelomonocytic leukemia
.
Clin Case Rep.
2020
;
8
(
12
):
2641
-
2644
.
114.
Röth
A
,
Bommer
M
,
Hüttmann
A
, et al
.
Eculizumab in cold agglutinin disease (DECADE): an open-label, prospective, bicentric, nonrandomized phase 2 trial
.
Blood Adv.
2018
;
2
(
19
):
2543
-
2549
.
115.
Röth
A
,
Barcellini
W
,
D’Sa
S
, et al
.
Sutimlimab in cold agglutinin disease
.
N Engl J Med.
2021
;
384
(
14
):
1323
-
1334
.
116.
Hillmen
P
,
Szer
J
,
Weitz
I
, et al
.
Pegcetacoplan versus eculizumab in paroxysmal nocturnal hemoglobinuria
.
N Engl J Med.
2021
;
384
(
11
):
1028
-
1037
.
117.
Frey
NV
,
Porter
DL
.
Graft-versus-host disease after donor leukocyte infusions: presentation and management
.
Best Pract Res Clin Haematol.
2008
;
21
(
2
):
205
-
222
.
118.
Kollman
C
,
Howe
CWS
,
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.
2001
;
98
(
7
):
2043
-
2051
.
119.
Seebach
JD
,
Stussi
G
,
Passweg
JR
, et al;
GVHD Working Committee of Center for International Blood and Marrow Transplant Research
.
ABO blood group barrier in allogeneic bone marrow transplantation revisited
.
Biol Blood Marrow Transplant.
2005
;
11
(
12
):
1006
-
1013
.
120.
Helming
AM
,
Brand
A
,
Wolterbeek
R
,
van Tol
MJD
,
Egeler
RM
,
Ball
LM
.
ABO incompatible stem cell transplantation in children does not influence outcome
.
Pediatr Blood Cancer.
2007
;
49
(
3
):
313
-
317
.
121.
Michallet
M
,
Le
QH
,
Mohty
M
, et al
.
Predictive factors for outcomes after reduced intensity conditioning hematopoietic stem cell transplantation for hematological malignancies: a 10-year retrospective analysis from the Société Française de Greffe de Moelle et de Thérapie Cellulaire
.
Exp Hematol.
2008
;
36
(
5
):
535
-
544
.
122.
Kimura
F
,
Sato
K
,
Kobayashi
S
, et al;
Japan Marrow Donor Program
.
Impact of AB0-blood group incompatibility on the outcome of recipients of bone marrow transplants from unrelated donors in the Japan Marrow Donor Program
.
Haematologica.
2008
;
93
(
11
):
1686
-
1693
.
123.
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.
2017
;
92
(
8
):
789
-
796
.
124.
Ataca Atilla
P
,
Akkus
E
,
Atilla
E
, et al
.
Effects of ABO incompatibility in allogeneic hematopoietic stem cell transplantation
.
Transfus Clin Biol.
2020
;
27
(
3
):
115
-
121
.
125.
Heim
MU
,
Schleuning
M
,
Eckstein
R
, et al
.
Rh antibodies against the pretransplant red cells following Rh-incompatible bone marrow transplantation
.
Transfusion.
1988
;
28
(
3
):
272
-
275
.
126.
Tasaki
T
,
Sasaki
S
,
Gotoh
K
,
Itoh
C
,
Itoh
S
,
Kuriya
S
.
Multiple red blood cell antibodies produced by donor B lymphocytes after ABO-matched allogeneic bone marrow transplantation
.
Transfus Sci.
1999
;
20
(
2
):
121
-
127
.
127.
Izumi
N
,
Fuse
I
,
Furukawa
T
, et al
.
Long-term production of pre-existing alloantibodies to E and c after allogenic BMT in a patient with aplastic anemia resulting in delayed hemolytic anemia
.
Transfusion.
2003
;
43
(
2
):
241
-
245
.
128.
Zupańska
B
,
Zaucha
JM
,
Michalewska
B
,
Malinowska
A
,
Brojer
E
,
Hellmann
A
.
Multiple red cell alloantibodies, including anti-Dib, after allogeneic ABO-matched peripheral blood progenitor cell transplantation
.
Transfusion.
2005
;
45
(
1
):
16
-
20
.
129.
Myser
T
,
Steedman
M
,
Hunt
K
,
Strohm
P
,
Williams
M
,
Kennedy
M
.
A bone marrow transplant with an acquired anti-Le(a): a case study
.
Hum Immunol.
1986
;
17
(
2
):
102
-
106
.