1
|
Nickel RS, Margulies S, Panchapakesan K, Chorvinsky E, Nino G, Gierdalski M, Bost J, Luban NLC, Webb J. Adding hydroxyurea to chronic transfusion therapy for sickle cell anemia reduces transfusion burden. Transfusion 2024. [PMID: 39580793 DOI: 10.1111/trf.18073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/07/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Chronic red blood cell (RBC) transfusion is an established therapy to prevent stroke in patients with sickle cell anemia (SCA). It is unclear if adding daily hydroxyurea treatment to chronic transfusion is beneficial. STUDY DESIGN AND METHODS We conducted a phase 2 clinical trial (NCT03644953) investigating the addition of dose-escalated hydroxyurea to chronic transfusion for patients with SCA receiving simple chronic transfusion for stroke prevention. Simple chronic transfusion therapy was administered as per the same protocol before and after hydroxyurea treatment in which the volume transfused was dependent on the pretransfusion hemoglobin (Hb). RESULTS A total of 14 participants enrolled with nine completing one year of combination hydroxyurea and transfusion (HAT) therapy after reaching hydroxyurea target dose. No participant who discontinued the study prematurely had a serious adverse event attributed to HAT. Among the nine participants who completed the study, eight participants achieved a reduction in RBC transfusion volume with a median reduction of -19.4 mL/kg/year (interquartile range -31.8, -2.8 mL/kg/year), p = .02, when comparing pre- and post-HAT time periods. With the addition of hydroxyurea participants had a significant increase in pretransfusion Hb S% but this was balanced by an increased Hb F% and decreased lactate dehydrogenase. One participant developed a pretransfusion Hb >11 g/dL and Hb S > 45% that required holding hydroxyurea and changing to partial manual exchange transfusions. No patient had evidence of cerebrovascular disease progression. DISCUSSION Hydroxyurea added to chronic transfusion therapy for patients with SCA is feasible and decreases RBC transfusion volume requirements.
Collapse
Affiliation(s)
- Robert Sheppard Nickel
- Children's National Hospital, Washington, DC, USA
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Children's National Research Institute, Washington, DC, USA
| | | | | | | | - Gustavo Nino
- Children's National Hospital, Washington, DC, USA
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Children's National Research Institute, Washington, DC, USA
| | - Marcin Gierdalski
- Children's National Hospital, Washington, DC, USA
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Children's National Research Institute, Washington, DC, USA
| | - James Bost
- Children's National Hospital, Washington, DC, USA
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Children's National Research Institute, Washington, DC, USA
| | - Naomi L C Luban
- Children's National Hospital, Washington, DC, USA
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Children's National Research Institute, Washington, DC, USA
| | - Jennifer Webb
- Children's National Hospital, Washington, DC, USA
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
2
|
Jajosky RP, Zerra PE, Chonat S, Stowell SR, Arthur CM. Harnessing the potential of red blood cells in immunotherapy. Hum Immunol 2024; 85:111084. [PMID: 39255557 DOI: 10.1016/j.humimm.2024.111084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 09/12/2024]
Abstract
Red blood cell (RBC) transfusion represents one of the earliest and most widespread forms of cellular therapy. While the primary purpose of RBC transfusions is to enhance the oxygen-carrying capacity of the recipient, RBCs also possess unique properties that make them attractive vehicles for inducing antigen-specific immune tolerance. Preclinical studies have demonstrated that RBC transfusion alone, in the absence of inflammatory stimuli, often fails to elicit detectable alloantibody formation against model RBC antigens. Several studies also suggest that RBC transfusion without inflammation may not only fail to generate a detectable alloantibody response but can also induce a state of antigen-specific non-responsiveness, a phenomenon potentially influenced by the density of the corresponding RBC alloantigen. The unique properties of RBCs, including their inability to divide and their stable surface antigen expression, make them attractive platforms for displaying exogenous antigens with the goal of leveraging their ability to induce antigen-specific non-responsiveness. This could facilitate antigen presentation to the host's immune system without triggering innate immune activation, potentially enabling the induction of antigen-specific tolerance for therapeutic applications in autoimmune disorders, preventing immune responses against protein therapeutics, or reducing alloreactivity in the setting of transfusion and transplantation.
Collapse
Affiliation(s)
- Ryan P Jajosky
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Patricia E Zerra
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States; Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Satheesh Chonat
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Sean R Stowell
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| | - Connie M Arthur
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
3
|
Arthur CM, Hollenhorst M, Wu SC, Jajosky R, Nakahara H, Jan HM, Zheng L, Covington M, Rakoff-Nahoum S, Yeung M, Lane W, Josephson C, Cummings RD, Stowell SR. ABO blood groups and galectins: Implications in transfusion medicine and innate immunity. Semin Immunol 2024; 74-75:101892. [PMID: 39405833 DOI: 10.1016/j.smim.2024.101892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/05/2024] [Accepted: 10/05/2024] [Indexed: 11/18/2024]
Abstract
ABO blood group antigens, which are complex carbohydrate moieties, and the first human polymorphisms identified, are critical in transfusion medicine and transplantation. Despite their discovery over a century ago, significant questions remain about the development of anti-ABO antibodies and the structural features of ABO antigens that cause hemolytic transfusion reactions. Anti-ABO antibodies develop naturally during the first few months of life, in contrast to other red blood cell (RBC) alloantibodies which form after allogeneic RBC exposure. Anti-ABO antibodies are the most common immune barrier to transfusion and transplantation, but the factors driving their formation are incompletely understood. Some studies suggest that microbes that express glycans similar in structure to the blood group antigens could play a role in anti-blood group antibody formation. While the role of these microbes in clinically relevant anti-blood group antibody formation remains to be defined, the presence of these microbes raises questions about how blood group-positive individuals protect themselves against blood group molecular mimicry. Recent studies suggest that galectins can bind and kill microbes that mimic blood group antigens, suggesting a unique host defense mechanism against microbial molecular mimicry. However, new models are needed to fully define the impact of microbes, galectins, or other factors on the development of clinically relevant naturally occurring anti-blood group antibodies.
Collapse
Affiliation(s)
- Connie M Arthur
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Harvard Medical School Center for Glycosciences, USA
| | - Marie Hollenhorst
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shang-Chuen Wu
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryan Jajosky
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hirotomo Nakahara
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hau-Ming Jan
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Leon Zheng
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mischa Covington
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Melissa Yeung
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - William Lane
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Richard D Cummings
- Harvard Medical School Center for Glycosciences, USA; Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sean R Stowell
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Harvard Medical School Center for Glycosciences, USA.
| |
Collapse
|
4
|
Shold J, Dasgupta A, Ye Z. Prevention of potential delayed hemolytic transfusion reaction in two sickle cell patients using intravenous immunoglobulins and steroids before and after red blood cell exchange with antigen positive units and review literature. Transfus Apher Sci 2024; 63:103920. [PMID: 38570214 DOI: 10.1016/j.transci.2024.103920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/04/2024] [Accepted: 03/31/2024] [Indexed: 04/05/2024]
Abstract
Emergent Red Blood Cell (RBC) exchange is indicated in sickle cell disease (SCD) patients with severe acute chest syndrome. However, fully matched RBC units may not be available for patients with multiple RBC antibodies. Intravenous immunoglobulin (IVIG) and steroids were reported for preventing potential delayed hemolytic transfusion reaction (HTR) in simple transfusion of antigen-positive RBCs. We investigated the efficacy and safety of IVIG and steroids in two SCD patients presented with acute chest syndrome receiving RBC exchange with multiple incompatible units. The first patient had multiple historical alloantibodies, including anti-Jsb, although none of them were reactive. IVIG (1 g/kg) was given before and after RBC exchange with methylprednisolone (500 mg IV) one hour before exchange. Her sickle hemoglobin (HbS) was reduced from 89.4% to 17.4% after the exchange with five Jsb-positive units. The patient improved clinically without acute or delayed hemolysis. The second patient had reactive anti-Jsb on two different admissions 18 months apart. Only one of the sixteen units used in the exchanges was Jsb negative. He received the same IVIG regimen during both admissions but 100 mg IV hydrocortisone instead of methylprednisolone. His HbS was reduced from 63.4% to 22.4% after the first exchange. Significant clinical improvements were achieved after both exchanges. No delayed HTR was observed. Our experience of these two patients suggested that IVIG and steroids may be used in preventing potential delayed HTR in some SCD patients with rare antibodies receiving large amounts of antigen-positive RBC products.
Collapse
Affiliation(s)
- Janna Shold
- University of Kansas Medical Center, Kansas City, KS, United States
| | - Amitava Dasgupta
- University of Kansas Medical Center, Kansas City, KS, United States
| | - Zhan Ye
- University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
5
|
Souza Gonçalves M, Albuquerque CCMX, Yahouedehou SCMA, Francisco MVL, Fraiji NA, de Siqueira IC, Gonçalves MS. Prevalence of arboviruses in sickle cell disease patients from two different regions of Brazil, the North and Northeast. Braz J Infect Dis 2024; 28:103741. [PMID: 38670165 PMCID: PMC11070587 DOI: 10.1016/j.bjid.2024.103741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/29/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
Sickle Cell Disease (SCD) is a hereditary disease characterized by extravascular and intravascular hemolysis and clinical variability, from mild pain to potentially life-threatening. Arboviruses include mainly Zika (ZIKV), Chikungunya (CHKV), and Dengue (DENV) virus, and are considered a public and social health problem. The present cross-sectional observational study aimed to investigate the prevalence of arbovirus infection in SCD patients from two Brazilian cities, Salvador and Manaus located in Bahia and Amazonas states respectively. A total of 409 individuals with SCD were included in the study, and 307 (75.06 %) patients tested positive for DENV-IgG, 161 (39.36 %) for ZIKV-IgG, and 60 (14.67 %) for CHIKV-IgG. Only one individual was positive for DENV-NS1 and another for DENV-IgM, both from Salvador. No individuals had positive serology for ZIKV-IgM or CHIKV-IgM. Arbovirus positivity by IgG testing revealed that the SCD group presented high frequencies in both cities. Interestingly, these differences were only statistically significant for ZIKV-IgG (p = 0.023) and CHIKV-IgG (p = 0.005) among SCD patients from Manaus. The reshaping of arbovirus from its natural habitat by humans due to disorderly urban expansion and the ease of international Mobility has been responsible for facilitating the spread of vector-borne infectious diseases in humans. We found the need for further studies on arboviruses in this population to elucidate the real association and impact, especially in acute infection. We hope that this study will contribute to improvements in the personalized clinical follow-up of SCD patients, identifying the influence of arbovirus infection in severe disease manifestations.
Collapse
Affiliation(s)
- Marilda Souza Gonçalves
- Fundação Oswaldo Cruz (FIOCRUZ), Instituto Gonçalo Moniz, Salvador, BA, Brazil; Faculdade de Ciências Farmacêuticas, Laboratório de Análise Especializada em Biologia Molecular (LAEBM), Universidade Federal do Amazonas, Manaus, AM, Brazil; Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil.
| | | | | | | | - Nelson Abrahim Fraiji
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil
| | | | | |
Collapse
|
6
|
Chang DY, Wankier Z, Arthur CM, Stowell SR. The ongoing challenge of RBC alloimmunization in the management of patients with sickle cell disease. Presse Med 2023; 52:104211. [PMID: 37981194 DOI: 10.1016/j.lpm.2023.104211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2023] Open
Abstract
RBC transfusion remains a cornerstone in the treatment of sickle cell disease (SCD). However, as with many interventions, transfusion of RBCs is not without risk. Allogeneic RBC exposure can result in the development of alloantibodies, which can make it difficult to find compatible RBCs for future transfusion and increases the likelihood of life-threatening complications. The development of RBC alloantibodies occurs when a patient's immune system produces alloantibodies against foreign alloantigens present on RBCs. Despite its longstanding recognition, RBC alloimmunization has increasingly become a challenge when caring for patients with SCD. The growing prominence of alloimmunization can be attributed to several factors, including expanded indications for transfusions, increased lifespan of patients with SCD, and inadequate approaches to prevent alloimmunization. Recognizing these challenges, recent observational studies and preclinical models have begun to elucidate the immune pathways that underpin RBC alloimmunization. These emerging data hold promise in paving the way for innovative prevention strategies, with the goal of increasing the safety and efficacy of RBC transfusion in patients with SCD who are most vulnerable to alloimmunization.
Collapse
Affiliation(s)
- Daniel Y Chang
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Zakary Wankier
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Connie M Arthur
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Sean R Stowell
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
7
|
Guarente J, Tormey C. Transfusion Support of Patients with Myelodysplastic Syndromes. Clin Lab Med 2023; 43:669-683. [PMID: 37865510 DOI: 10.1016/j.cll.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2023]
Abstract
Patients with MDS often suffer from anemia, and less often thrombocytopenia, and thus are a frequently transfused population. Red blood cell (RBC) transfusion may be used to improve functional capacity and quality of life in this population, while platelet transfusion is typically used to decrease bleeding risk. Despite the frequency of transfusion in patients with MDS, there are few well-defined guidelines for RBC and platelet transfusion support in this patient population. Transfusion is not without risk-patients with MDS who are frequently transfused may develop alloantibodies to RBC antigens, which can lead to hemolytic transfusion reactions and delays in obtaining compatible RBCs. Regular communication between clinicians and blood bank physicians is crucial to ensure that patients with MDS receive the most appropriate blood products.
Collapse
Affiliation(s)
- Juliana Guarente
- Department of Pathology and Genomic Medicine, Pathology Residency Program, Thomas Jefferson University Hospital, 111 South 11th Street Gibbon Building, Room 8220, Philadelphia, PA 19107, USA
| | - Christopher Tormey
- Department of Laboratory Medicine, Transfusion Medicine Fellowship, Yale University School of Medicine, Yale-New Haven Hospital, 55 Park Street, Floor 3, Room 329D, New Haven, CT 06511, USA
| |
Collapse
|
8
|
Jajosky RP, Wu SC, Jajosky PG, Stowell SR. Plasmodium knowlesi ( Pk) Malaria: A Review & Proposal of Therapeutically Rational Exchange (T-REX) of Pk-Resistant Red Blood Cells. Trop Med Infect Dis 2023; 8:478. [PMID: 37888606 PMCID: PMC10610852 DOI: 10.3390/tropicalmed8100478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
Plasmodium knowlesi (Pk) causes zoonotic malaria and is known as the "fifth human malaria parasite". Pk malaria is an emerging threat because infections are increasing and can be fatal. While most infections are in Southeast Asia (SEA), especially Malaysia, travelers frequently visit this region and can present with Pk malaria around the world. So, clinicians need to know (1) patients who present with fever after recent travel to SEA might be infected with Pk and (2) Pk is often misdiagnosed as P. malariae (which typically causes less severe malaria). Here we review the history, pathophysiology, clinical features, diagnosis, and treatment of Pk malaria. Severe disease is most common in adults. Signs and symptoms can include fever, abdominal pain, jaundice, acute kidney injury, acute respiratory distress syndrome, hyponatremia, hyperparasitemia, and thrombocytopenia. Dengue is one of the diseases to be considered in the differential. Regarding pathophysiologic mechanisms, when Pk parasites invade mature red blood cells (RBCs, i.e., normocytes) and reticulocytes, changes in the red blood cell (RBC) surface can result in life-threatening cytoadherence, sequestration, and reduced RBC deformability. Since molecular mechanisms involving the erythrocytic stage are responsible for onset of severe disease and lethal outcomes, it is biologically plausible that manual exchange transfusion (ET) or automated RBC exchange (RBCX) could be highly beneficial by replacing "sticky" parasitized RBCs with uninfected, deformable, healthy donor RBCs. Here we suggest use of special Pk-resistant donor RBCs to optimize adjunctive manual ET/RBCX for malaria. "Therapeutically-rational exchange transfusion" (T-REX) is proposed in which Pk-resistant RBCs are transfused (instead of disease-promoting RBCs). Because expression of the Duffy antigen on the surface of human RBCs is essential for parasite invasion, T-REX of Duffy-negative RBCs-also known as Fy(a-b-) RBCs-could replace the majority of the patient's circulating normocytes with Pk invasion-resistant RBCs (in a single procedure lasting about 2 h). When sequestered or non-sequestered iRBCs rupture-in a 24 h Pk asexual life cycle-the released merozoites cannot invade Fy(a-b-) RBCs. When Fy(a-b-) RBC units are scarce (e.g., in Malaysia), clinicians can consider the risks and benefits of transfusing plausibly Pk-resistant RBCs, such as glucose-6-phosphate dehydrogenase deficient (G6PDd) RBCs and Southeast Asian ovalocytes (SAO). Patients typically require a very short recovery time (<1 h) after the procedure. Fy(a-b-) RBCs should have a normal lifespan, while SAO and G6PDd RBCs may have mildly reduced half-lives. Because SAO and G6PDd RBCs come from screened blood donors who are healthy and not anemic, these RBCs have a low-risk for hemolysis and do not need to be removed after the patient recovers from malaria. T-REX could be especially useful if (1) antimalarial medications are not readily available, (2) patients are likely to progress to severe disease, or (3) drug-resistant strains emerge. In conclusion, T-REX is a proposed optimization of manual ET/RBCX that has not yet been utilized but can be considered by physicians to treat Pk malaria patients.
Collapse
Affiliation(s)
- Ryan Philip Jajosky
- Joint Program in Transfusion Medicine, Brigham and Women’s Hospital, Harvard Medical School, 630E New Research Building, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; (S.-C.W.)
- Biconcavity Inc., Lilburn, GA 30047, USA
| | - Shang-Chuen Wu
- Joint Program in Transfusion Medicine, Brigham and Women’s Hospital, Harvard Medical School, 630E New Research Building, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; (S.-C.W.)
| | | | - Sean R. Stowell
- Joint Program in Transfusion Medicine, Brigham and Women’s Hospital, Harvard Medical School, 630E New Research Building, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; (S.-C.W.)
| |
Collapse
|
9
|
Fasano RM, Doctor A, Stowell SR, Spinella PC, Carson JL, Maier CL, Josephson CD, Triulzi DJ. Optimizing RBC Transfusion Outcomes in Patients with Acute Illness and in the Chronic Transfusion Setting. Transfus Med Rev 2023; 37:150758. [PMID: 37743191 DOI: 10.1016/j.tmrv.2023.150758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023]
Abstract
Red blood cell (RBC) transfusion is a common clinical intervention used to treat patients with acute and chronic anemia. The decision to transfuse RBCs in the acute setting is based on several factors but current clinical studies informing optimal RBC transfusion decision making (TDM) are largely based upon hemoglobin (Hb) level. In contrast to transfusion in acute settings, chronic RBC transfusion therapy has several different purposes and is associated with distinct transfusion risks such as iron overload and RBC alloimmunization. Consequently, RBC TDM in the chronic setting requires optimizing the survival of transfused RBCs in order to reduce transfusion exposure over the lifespan of an individual and the associated transfusion complications mentioned. This review summarizes the current medical literature addressing optimal RBC-TDM in the acute and chronic transfusion settings and discusses the current gaps in knowledge which need to be prioritized in future national and international research initiatives.
Collapse
Affiliation(s)
- Ross M Fasano
- Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, USA.
| | - Allan Doctor
- Division of Pediatric Critical Care Medicine and Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sean R Stowell
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philip C Spinella
- Departments of Surgery and Critical Care Medicine, Pittsburgh University, Pittsburgh, PA, USA
| | - Jeffrey L Carson
- Division of General Internal Medicine, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Cheryl L Maier
- Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, USA
| | - Cassandra D Josephson
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Darrell J Triulzi
- Vitalant and Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
10
|
Zheng Y, Gossett JM, Chen PL, Barton M, Ryan M, Yu J, Kang G, Hankins JS, Chou ST. Proinflammatory state promotes red blood cell alloimmunization in pediatric patients with sickle cell disease. Blood Adv 2023; 7:4799-4808. [PMID: 37023228 PMCID: PMC10469551 DOI: 10.1182/bloodadvances.2022008647] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/09/2023] [Accepted: 02/27/2023] [Indexed: 04/08/2023] Open
Abstract
We examined risk factors for red blood cell (RBC) alloimmunization in pediatric patients with sickle cell disease, focusing on the recipients' inflammatory state at the time of transfusion and anti-inflammatory role of hydroxyurea (HU). Among 471 participants, 55 (11.70%) participants were alloimmunized and formed 59 alloantibodies and 17 autoantibodies with an alloimmunization rate of 0.36 alloantibodies per 100 units. Analysis of 27 participants in whom alloantibodies were formed with specificities showed 23.8% (30/126) of units transfused during a proinflammatory event resulting in alloantibody formation compared with 2.8% (27/952) of units transfused at steady state. Therefore, transfusion during proinflammatory events increased the risk for alloimmunization (odds ratio [OR], 4.22; 95% confidence interval [CI], 1.64-10.85; P = .003). Further analysis of all the 471 participants showed that alloimmunization of patients who received episodic transfusion, mostly during proinflammatory events, was not reduced with HU therapy (OR, 6.52; 95% CI, 0.85-49.77; P = .071), HU therapy duration (OR, 1.13; 95% CI, 0.997-1.28; P = .056), or HU dose (OR, 1.06; 95% CI, 0.96-1.16; P = .242). The analysis also identified high transfusion burden (OR, 1.02; 95% CI, 1.003-1.04; P = .020) and hemoglobin S (HbSS) and HbSβ0-thalassemia genotypes (OR, 11.22, 95% CI, 1.51-83.38; P = .018) as additional risk factors for alloimmunization. In conclusion, the inflammatory state of transfusion recipients affects the risk of RBC alloimmunization, which is not modified by HU therapy. Judicious use of transfusion during proinflammatory events is critical for preventing alloimmunization.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jeffrey M. Gossett
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Pei-Lin Chen
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Martha Barton
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Missy Ryan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jing Yu
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Guolian Kang
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jane S. Hankins
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Stella T. Chou
- Departments of Pediatrics and Pathology, The Children’s Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA
| |
Collapse
|
11
|
Salarvand S, Moeini Nasab S, Abdollahi A, Nozarian Z, Nazar E. Frequency and Underlying Causes of Alloimmunization Against Red Blood Cell Antigens in Patients Referred to the Blood Bank of the Tertiary Referral Hospital of Tehran from 2018 to 2020. ARCHIVES OF IRANIAN MEDICINE 2023; 26:499-503. [PMID: 38310405 PMCID: PMC10862052 DOI: 10.34172/aim.2023.75] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 07/03/2023] [Indexed: 02/05/2024]
Abstract
BACKGROUND Alloimmunization against blood group antigens is an important non-infectious complication of blood transfusion, and early detection of these alloantibodies by antibody screening before transfusion is crucial. Identifying which underlying factors will affect the occurrence of alloimmunization will be necessary to manage this event as accurately as possible. We aimed to assess the prevalence rate and main determinants of RBC alloimmunization among patients referred to a large referral blood bank in Iran. METHODS This retrospective cross-sectional study was conducted on all patients referred to a blood bank at Imam Khomeini Hospital between October 2018 and September 2020. Information was collected by referring to the archives of the hospital information system as well as the documents recorded at the blood bank ward and reviewed by two pathologists and completed documents. RESULTS In total, 39270 cases were cross-matched. Accordingly, the frequency of alloimmunization cases was equal to 220 cases, which indicated a prevalence of 0.56%. The most common alloantibodies were anti-K (43.2%, 95% CI: 36.8‒49.5), anti-E (34%, 95% CI: 27.7‒40.5), and anti-C (16.3%, 95% CI: 11.4‒21.4). Among patients with positive alloimmunization, the most common blood groups were blood group B (34.6%), followed by blood group A (34.1%). Most of these patients were Rh-positive (77.3%). In patients with positive alloimmunization, the frequency of hemoglobinopathy was estimated to be 37.7%. Frequent blood transfusions were found in 42.2%, a history of malignancy in 17.3%, graft history in 11.3%, and a history of pregnancy in 35.0%. CONCLUSION Alloimmunization was more prevalent and more predictable among patients with hemoglobinopathies and those receiving recurrent transfusions. Therefore, a history of repeated blood transfusions should be regarded as a risk factor contributing to alloimmunization.
Collapse
Affiliation(s)
- Samaneh Salarvand
- Department of Pathology, Imam Khomeini Complex Hospital (IKHC), Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Moeini Nasab
- Department of Pathology, Imam Khomeini Complex Hospital (IKHC), Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Abdollahi
- Department of Pathology, Imam Khomeini Complex Hospital (IKHC), Tehran University of Medical Sciences, Tehran, Iran
| | - Zohreh Nozarian
- Department of Pathology, Farabi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Nazar
- Department of Pathology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Chornenkyy Y, Yamamoto T, Hara H, Stowell SR, Ghiran I, Robson SC, Cooper DKC. Future prospects for the clinical transfusion of pig red blood cells. Blood Rev 2023; 61:101113. [PMID: 37474379 PMCID: PMC10968389 DOI: 10.1016/j.blre.2023.101113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/23/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023]
Abstract
Transfusion of allogeneic human red blood cell (hRBCs) is limited by supply and compatibility between individual donors and recipients. In situations where the blood supply is constrained or when no compatible RBCs are available, patients suffer. As a result, alternatives to hRBCs that complement existing RBC transfusion strategies are needed. Pig RBCs (pRBCs) could provide an alternative because of their abundant supply, and functional similarities to hRBCs. The ability to genetically modify pigs to limit pRBC immunogenicity and augment expression of human 'protective' proteins has provided major boosts to this research and opens up new therapeutic avenues. Although deletion of expression of xenoantigens has been achieved in genetically-engineered pigs, novel genetic methods are needed to introduce human 'protective' transgenes into pRBCs at the high levels required to prevent hemolysis and extend RBC survival in vivo. This review addresses recent progress and examines future prospects for clinical xenogeneic pRBC transfusion.
Collapse
Affiliation(s)
- Yevgen Chornenkyy
- Department of Pathology, McGaw Medical Center of Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Takayuki Yamamoto
- Center for Transplantation Science, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA; Division of Transplantation, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.
| | - Hidetaka Hara
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Sean R Stowell
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ionita Ghiran
- Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Simon C Robson
- Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - David K C Cooper
- Center for Transplantation Science, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Wabnitz H, Cruz-Leal Y, Lazarus AH. Antigen-specific IgG subclass composition in recipient mice can indicate the degree of red blood cell alloimmunization as well as discern between primary and secondary immunization. Transfusion 2023; 63:619-628. [PMID: 36591986 DOI: 10.1111/trf.17232] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND Despite the vast antigen disparity between donor and recipient red blood cells (RBCs), only 2%-6% of transfusion patients mount an alloantibody response. Recently, RBC antigen density has been proposed as one of the factors that can influence alloimmunization, however, there has been no characterization of the role of antigen density along with RBC dose in primary and secondary immunization. STUDY DESIGN AND METHODS To generate RBCs that express distinct antigen copy numbers, different quantities of hen egg lysozyme (HEL) were coupled to murine RBCs. The HEL-RBCs were subsequently transfused into recipient mice at different RBC doses and their HEL-specific IgM, IgG, and IgG subclass response was evaluated. RESULTS Productive immune responses could be generated through a high copy number antigen transfused at low RBC doses or a low copy number transfused at high RBC doses. Further, primary but submaximal humoral immunization predominantly induced the IgG2b and IgG3 subclasses. In contrast, a maximal primary immunization or a secondary immunization induced all four IgG subclasses. DISCUSSION Our results confirm the existence of an antigen threshold for productive immune responses but indicate that a high antigen copy number alone might not be enough to induce a response, but rather a combination of both antigen copy number and cell dosage may determine the outcome of immunization. Further, this study provides a proof of concept that the IgG subclass composition can be an indicator of the level of RBC alloimmunization as well as discern between primary and secondary immunization at least in this murine model.
Collapse
Affiliation(s)
- Hanna Wabnitz
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Yoelys Cruz-Leal
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Alan H Lazarus
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
14
|
S. Abdullah SZ, Hassan MN, Ramli M, Abdullah M, Mohd Noor NH. Red Blood Cell Alloimmunization and Its Associated Factors among Chronic Liver Disease Patients in a Teaching Hospital in Northeastern Malaysia. Diagnostics (Basel) 2023; 13:diagnostics13050886. [PMID: 36900030 PMCID: PMC10001194 DOI: 10.3390/diagnostics13050886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 03/02/2023] Open
Abstract
Red blood cell (RBC) alloimmunization is an important complication of blood transfusion. Variations in the frequency of alloimmunization have been noted among different patient populations. We aimed to determine the prevalence of RBC alloimmunization and associated factors among chronic liver disease (CLD) patients in our center. This is a case-control study involving 441 patients with CLD who were being treated at Hospital Universiti Sains Malaysia and subjected to pre-transfusion testing from April 2012 until April 2022. Clinical and laboratory data were retrieved and statistically analyzed. A total of 441 CLD patients were included in our study, with the majority being elderly, with the mean age of patients 57.9 (SD ± 12.1) years old, male (65.1%) and Malays (92.1%). The most common causes of CLD in our center are viral hepatitis (62.1%) and metabolic liver disease (25.4%). Twenty-four patients were reported to have RBC alloimmunization, resulting in an overall prevalence of 5.4%. Higher rates of alloimmunization were seen in females (7.1%) and patients with autoimmune hepatitis (11.1%). Most patients developed a single alloantibody (83.3%). The most common alloantibody identified belonged to the Rh blood group, anti-E (35.7%) and anti-c (14.3%), followed by the MNS blood group, anti-Mia (17.9%). There was no significant factor association of RBC alloimmunization among CLD patients identified. Our center has a low prevalence of RBC alloimmunization among CLD patients. However, the majority of them developed clinically significant RBC alloantibodies, mostly from the Rh blood group. Therefore, phenotype matching for Rh blood groups should be provided for CLD patients requiring blood transfusions in our center to prevent RBC alloimmunization.
Collapse
Affiliation(s)
- Siti Zaleha S. Abdullah
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Mohd Nazri Hassan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
- Transfusion Medicine Unit, Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
- Correspondence: (M.N.H.); (N.H.M.N.); Tel.: +60-9767-6198 (M.N.H.); +60-9767-6196 (N.H.M.N.)
| | - Marini Ramli
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
- Transfusion Medicine Unit, Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Marne Abdullah
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
- Transfusion Medicine Unit, Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Noor Haslina Mohd Noor
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
- Transfusion Medicine Unit, Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
- Correspondence: (M.N.H.); (N.H.M.N.); Tel.: +60-9767-6198 (M.N.H.); +60-9767-6196 (N.H.M.N.)
| |
Collapse
|
15
|
Arthur CM, Stowell SR. The Development and Consequences of Red Blood Cell Alloimmunization. ANNUAL REVIEW OF PATHOLOGY 2023; 18:537-564. [PMID: 36351365 PMCID: PMC10414795 DOI: 10.1146/annurev-pathol-042320-110411] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
While red blood cell (RBC) transfusion is the most common medical intervention in hospitalized patients, as with any therapeutic, it is not without risk. Allogeneic RBC exposure can result in recipient alloimmunization, which can limit the availability of compatible RBCs for future transfusions and increase the risk of transfusion complications. Despite these challenges and the discovery of RBC alloantigens more than a century ago, relatively little has historically been known regarding the immune factors that regulate RBC alloantibody formation. Through recent epidemiological approaches, in vitro-based translational studies, and newly developed preclinical models, the processes that govern RBC alloimmunization have emerged as more complex and intriguing than previously appreciated. Although common alloimmunization mechanisms exist, distinct immune pathways can be engaged, depending on the target alloantigen involved. Despite this complexity, key themes are beginning to emerge that may provide promising approaches to not only actively prevent but also possibly alleviate the most severe complications of RBC alloimmunization.
Collapse
Affiliation(s)
- Connie M Arthur
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, ,
| | - Sean R Stowell
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, ,
| |
Collapse
|
16
|
An HH, Gagne AL, Maguire JA, Pavani G, Abdulmalik O, Gadue P, French DL, Westhoff CM, Chou ST. The use of pluripotent stem cells to generate diagnostic tools for transfusion medicine. Blood 2022; 140:1723-1734. [PMID: 35977098 PMCID: PMC9707399 DOI: 10.1182/blood.2022015883] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
Red blood cell (RBC) transfusion is one of the most common medical treatments, with more than 10 million units transfused per year in the United States alone. Alloimmunization to foreign Rh proteins (RhD and RhCE) on donor RBCs remains a challenge for transfusion effectiveness and safety. Alloantibody production disproportionately affects patients with sickle cell disease who frequently receive blood transfusions and exhibit high genetic diversity in the Rh blood group system. With hundreds of RH variants now known, precise identification of Rh antibody targets is hampered by the lack of appropriate reagent RBCs with uncommon Rh antigen phenotypes. Using a combination of human-induced pluripotent stem cell (iPSC) reprogramming and gene editing, we designed a renewable source of cells with unique Rh profiles to facilitate the identification of complex Rh antibodies. We engineered a very rare Rh null iPSC line lacking both RHD and RHCE. By targeting the AAVS1 safe harbor locus in this Rh null background, any combination of RHD or RHCE complementary DNAs could be reintroduced to generate RBCs that express specific Rh antigens such as RhD alone (designated D--), Goa+, or DAK+. The RBCs derived from these iPSCs (iRBCs) are compatible with standard laboratory assays used worldwide and can determine the precise specificity of Rh antibodies in patient plasma. Rh-engineered iRBCs can provide a readily accessible diagnostic tool and guide future efforts to produce an alternative source of rare RBCs for alloimmunized patients.
Collapse
Affiliation(s)
- Hyun Hyung An
- Division of Hematology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Alyssa L. Gagne
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Jean Ann Maguire
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Giulia Pavani
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Osheiza Abdulmalik
- Division of Hematology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Paul Gadue
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Deborah L. French
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | - Stella T. Chou
- Division of Hematology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Division of Transfusion Medicine, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
17
|
Rollins MR, Chou ST. Adverse events of red blood cell transfusions in patients with sickle cell disease. Transfus Apher Sci 2022; 61:103557. [PMID: 36064527 PMCID: PMC10149091 DOI: 10.1016/j.transci.2022.103557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Blood transfusion is a common medical intervention for patients with sickle cell disease (SCD) and disease related complications. While patients with SCD are at risk for all transfusion related adverse events defined by the National Healthcare Safety Network (NHSN) Biovigilance Component Hemovigilance Module Surveillance Protocol, they are uniquely susceptible to certain adverse events. This review discusses risk factors, mitigation strategies, and management recommendations for alloimmunization, hemolytic transfusion reactions, hyperviscosity and transfusion-associated iron overload in the context of SCD.
Collapse
Affiliation(s)
- Margo R Rollins
- Children's Healthcare of Atlanta, Department of Pathology and Laboratory Medicine, 1405 Clifton Rd NE, 1st Floor, Atlanta, GA 30322, USA; Emory University School of Medicine, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, 1405 Clifton Rd NE, Atlanta, GA 30322, USA
| | - Stella T Chou
- The Children's Hospital of Philadelphia, Departments of Pediatrics and Pathology and Laboratory Medicine, The School of Medicine at the University of Pennsylvania, 3615 Civic Center Boulevard, Abramson Research Building Room 316D, Philadelphia, PA 19104, USA.
| |
Collapse
|
18
|
Buban KR, Lawrence CE, Zhu XJ, Tobian AAR, Gehrie EA, Vozniak S, Shrestha R, Lokhandwala PM, Bloch EM. Algorithm‐based selection of automated red blood cell exchange procedure goals reduces blood utilization in chronically transfused adults with sickle cell disease. J Clin Apher 2022; 37:468-475. [DOI: 10.1002/jca.22004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 05/11/2022] [Accepted: 06/22/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Kristen R. Buban
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| | - Courtney E. Lawrence
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| | - Xianming Joshua Zhu
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| | - Aaron A. R. Tobian
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| | | | - Sonja Vozniak
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| | - Ruchee Shrestha
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| | | | - Evan M. Bloch
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| |
Collapse
|
19
|
Waldron E, Tanhehco YC. Under the Hood: The Molecular Biology Driving Gene Therapy for the Treatment of Sickle Cell Disease. Transfus Apher Sci 2022; 61:103566. [DOI: 10.1016/j.transci.2022.103566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
20
|
Covington ML, Cone-Sullivan JK, Andrzejewski C, Lu W, Thomasson RR, O'Brien K, Brunker PAR, Stowell SR. Unmasking delayed hemolytic transfusion reactions in patients with sickle-cell disease: Challenges and opportunities for improvement. Transfusion 2022; 62:1662-1670. [PMID: 35778994 DOI: 10.1111/trf.16967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/30/2022] [Accepted: 05/17/2022] [Indexed: 12/25/2022]
Affiliation(s)
- Mischa L Covington
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jensyn K Cone-Sullivan
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Chester Andrzejewski
- Transfusion Medicine Service, Baystate Medical Center, Baystate Health, Springfield, Massachusetts, USA
| | - Wen Lu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Reggie R Thomasson
- Department of Pathology and Laboratory Medicine, Boston Medical Center and Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kerry O'Brien
- Department of Pathology, Beth Israel Deaconess, Harvard Medical School, Boston, Massachusetts, USA
| | - Patricia A R Brunker
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sean R Stowell
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Rankin A, Webb J, Nickel RS. Preventing antibody positive delayed hemolytic transfusion reactions in sickle cell disease: Lessons learned from a case. Transfus Med 2022; 32:433-436. [PMID: 35318744 DOI: 10.1111/tme.12862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/02/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Red blood cell (RBC) transfusions are important in the management of patients with sickle cell disease (SCD). However, a potentially catastrophic complication of transfusion in this population is the delayed hemolytic transfusion reaction (DHTR). The pathophysiology of all DHTRs is not understood, but some are known to be caused by an anamnestic resurgence of RBC alloantibodies. CASE PRESENTATION A child with SCD transfused for acute chest syndrome re-presented a week after hospital discharge with severe anaemia, hemolysis, and a newly detected anti-E. This patient had been previously transfused years ago at an outside institution and the anti-E had not been previously documented. DISCUSSION The presented case of an antibody positive DHTR illustrates several concepts critical to the prevention of this complication. RBC alloantibodies must be detected and this information must be shared. Prophylactic C/c, E/e, K antigen matching is helpful for patients with SCD, but systems must be in place to identify these patients. Patients transfused at multiple different hospitals are especially at risk for this complication and efforts are needed to prevent them from suffering a DHTR.
Collapse
Affiliation(s)
- Alexander Rankin
- Children's National Hospital, Washington, District of Columbia, USA.,Children's Hospital Colorado, Aurora, Colorado, USA
| | - Jennifer Webb
- Children's National Hospital, Washington, District of Columbia, USA.,The George Washington University School of Medicine & Health Sciences, Washington, District of Columbia, USA
| | - Robert Sheppard Nickel
- Children's National Hospital, Washington, District of Columbia, USA.,The George Washington University School of Medicine & Health Sciences, Washington, District of Columbia, USA
| |
Collapse
|
22
|
Soldatenko A, Hoyt LR, Xu L, Calabro S, Lewis SM, Gallman AE, Hudson KE, Stowell SR, Luckey CJ, Zimring JC, Liu D, Santhanakrishnan M, Hendrickson JE, Eisenbarth SC. Innate and Adaptive Immunity to Transfused Allogeneic RBCs in Mice Requires MyD88. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:991-997. [PMID: 35039331 PMCID: PMC10107373 DOI: 10.4049/jimmunol.2100784] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/09/2021] [Indexed: 01/02/2023]
Abstract
RBC transfusion therapy is essential for the treatment of anemia. A serious complication of transfusion is the development of non-ABO alloantibodies to polymorphic RBC Ags; yet, mechanisms of alloantibody formation remain unclear. Storage of mouse RBCs before transfusion increases RBC immunogenicity through an unknown mechanism. We previously reported that sterile, stored mouse RBCs activate splenic dendritic cells (DCs), which are required for alloimmunization. Here we transfused mice with allogeneic RBCs to test whether stored RBCs activate pattern recognition receptors (PRRs) on recipient DCs to induce adaptive immunity. TLRs are a class of PRRs that regulate DC activation, which signal through two adapter molecules: MyD88 and TRIF. We show that the inflammatory cytokine response, DC activation and migration, and the subsequent alloantibody response to transfused RBCs require MyD88 but not TRIF, suggesting that a restricted set of PRRs are responsible for sensing RBCs and triggering alloimmunization.
Collapse
Affiliation(s)
- Arielle Soldatenko
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Laura R Hoyt
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Lan Xu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Samuele Calabro
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Steven M Lewis
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Antonia E Gallman
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Krystalyn E Hudson
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Sean R Stowell
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
| | - Chance J Luckey
- Department of Pathology, University of Virginia, Charlottesville, VA; and
| | - James C Zimring
- Department of Pathology, University of Virginia, Charlottesville, VA; and
| | - Dong Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Manjula Santhanakrishnan
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT.,Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Jeanne E Hendrickson
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT.,Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT; .,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
23
|
Hendrickson JE. Management of hemolytic transfusion reactions. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2021; 2021:704-709. [PMID: 34889404 PMCID: PMC8791106 DOI: 10.1182/hematology.2021000308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Delayed hemolytic transfusion reactions (DHTRs) in patients with sickle cell disease are underappreciated and potentially fatal. Patients with DHTRs typically have symptoms of pain or dark urine days to weeks following a red blood cell (RBC) transfusion. In instances of DHTRs with hyperhemolysis, the patient's hemoglobin (Hgb) may be significantly lower than it was pretransfusion, and the Hgb A may drop by more than 50%. In most cases, at least 1 RBC alloantibody and sometimes multiple RBC alloantibodies can be identified during the DHTR, with those antibodies presumably having fallen below the level of detection at the time of the implicated transfusion. However, in up to one-third of cases, no new RBC alloantibodies can be identified posttransfusion. Complement is increasingly being appreciated to play a role in DHTRs and hyperhemolysis, not only due to classic pathway activation (with complement fixed antibody bound to RBCs) but also due to alternative pathway activation (resulting in part from plasma free heme). As such, anti-C5 inhibition has recently been reported to be effective at mitigating hemolysis in the setting of some severe DHTRs. Transfusion avoidance during DHTRs is recommended if possible, with long-term transfusion support advice being less clear; for example, a history of a severe DHTR may lead to questions regarding the safety of transfusions prior to curative therapies such as stem cell transplantation or gene therapy. A better understanding of antibody-positive and antibody-negative DHTRs, including patient- or disease-specific risk factors, is necessary to improve transfusion safety.
Collapse
Affiliation(s)
- Jeanne E. Hendrickson
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
- Correspondence Jeanne E. Hendrickson, Yale University Department of Laboratory Medicine, 330 Cedar Street, Clinic Building 405, PO Box 208035, New Haven, CT 06520-0835; e-mail:
| |
Collapse
|
24
|
van Sambeeck JHJ, van der Schoot CE, van Dijk NM, Schonewille H, Janssen MP. Extended red blood cell matching for all transfusion recipients is feasible. Transfus Med 2021; 32:221-228. [PMID: 34845765 DOI: 10.1111/tme.12831] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 10/08/2021] [Accepted: 11/02/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To demonstrate the feasibility and effectiveness of extended matching of red blood cells (RBC) in practice. BACKGROUND At present, alloimmunisation preventing matching strategies are only applied for specific transfusion recipient groups and include a limited number of RBC antigens. The general assumption is that providing fully matched RBC units to all transfusion recipients is not feasible. In this article we refute this assumption and compute the proportion of alloimmunisation that can be prevented, when all donors and transfusion recipients are typed for A, B, D plus twelve minor blood group antigens (C, c, E, e, K, Fya , Fyb , Jka , Jkb , M, S and s). METHODS We developed a mathematical model that determines the optimal sequence for antigen matching. The model allows for various matching strategies, issuing policies and inventory sizes. RESULTS For a dynamic inventory composition (accounting for randomness in the phenotypes supplied and requested) and an antigen identical issuing policy 97% and 94% of alloimmunisation events can be prevented, when respectively one and two RBC units per recipient are requested from an inventory of 1000 units. Although this proportion decreases with smaller inventory sizes, even for an inventory of 60 units almost 50% of all alloimmunisation events can be prevented. CONCLUSION In case antigen of both donors and recipients are comprehensively typed, extended preventive matching is feasible for all transfusion recipients in practice and will significantly reduce transfusion-induced alloimmunisation and (alloantibody-induced) haemolytic transfusion reactions.
Collapse
Affiliation(s)
- Joost H J van Sambeeck
- Department of Donor Medicine Research, Sanquin Research, Amsterdam, The Netherlands.,Center for Healthcare Operations Improvement and Research, University of Twente, Enschede, The Netherlands.,Department of Stochastic Operations Research, University of Twente, Enschede, The Netherlands
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands.,Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Nico M van Dijk
- Center for Healthcare Operations Improvement and Research, University of Twente, Enschede, The Netherlands.,Department of Stochastic Operations Research, University of Twente, Enschede, The Netherlands
| | - Henk Schonewille
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands
| | - Mart P Janssen
- Department of Donor Medicine Research, Sanquin Research, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Madany E, Lee J, Halprin C, Seo J, Baca N, Majlessipour F, Hendrickson JE, Pepkowitz SH, Hayes C, Klapper E, Gibb DR. Altered type 1 interferon responses in alloimmunized and nonalloimmunized patients with sickle cell disease. EJHAEM 2021; 2:700-710. [PMID: 35128535 PMCID: PMC8813163 DOI: 10.1002/jha2.270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 01/05/2023]
Abstract
Patients with sickle cell disease (SCD) have a high prevalence of RBC alloimmunization. However, underlying mechanisms are poorly understood. Given that proinflammatory type 1 interferons (IFNα/β) and interferon stimulated genes (ISGs) promote alloimmunization in mice, we hypothesized that IFNα/β may contribute to the increased frequency of alloimmunization in patients with SCD. To investigate this, expression of ISGs in blood leukocytes and peripheral blood mononuclear cells (PBMCs) of previously transfused SCD patients with or without alloimmunization and race-matched healthy controls were quantified, and IFNα/β gene scores were calculated. IFNα/β gene scores of SCD leukocytes and plasma cytokines were elevated, compared to controls (gene score, p < 0.01). Upon stimulation with IFNβ, isolated PBMCs from patients with SCD had elevated ISGs and IFNα/β gene scores (p < 0.05), compared to stimulated PBMCs from controls. However, IFNβ-stimulated and unstimulated ISG expression did not significantly differ between alloimmunized and non-alloimmunized patients. These findings indicate that patients with SCD express an IFNα/β gene signature, and larger studies are needed to fully determine its role in alloimmunization. Further, illustration of altered IFNα/β responses in SCD has potential implications for IFNα/β-mediated viral immunity, responses to IFNα/β-based therapies, and other sequelae of SCD.
Collapse
Affiliation(s)
- Emaan Madany
- Cedars‐Sinai Medical CenterDepartment of Pathology and Laboratory MedicineLos AngelesCaliforniaUnited States
| | - June Lee
- Cedars‐Sinai Medical CenterDepartment of Pathology and Laboratory MedicineLos AngelesCaliforniaUnited States
| | - Chelsea Halprin
- Cedars‐Sinai Medical CenterDepartment of Pathology and Laboratory MedicineLos AngelesCaliforniaUnited States
| | - Jina Seo
- Cedars‐Sinai Medical CenterDepartment of Pathology and Laboratory MedicineLos AngelesCaliforniaUnited States
| | - Nicole Baca
- Cedars‐Sinai Medical CenterDepartment of PediatricsLos AngelesCaliforniaUnited States
| | - Fataneh Majlessipour
- Cedars‐Sinai Medical CenterDepartment of PediatricsLos AngelesCaliforniaUnited States
| | - Jeanne E. Hendrickson
- Department of Laboratory MedicineYale University School of MedicineNew HavenConnecticutUnited States
- Department of PediatricsYale University School of MedicineNew HavenConnecticutUnited States
| | - Samuel H. Pepkowitz
- Cedars‐Sinai Medical CenterDepartment of Pathology and Laboratory MedicineLos AngelesCaliforniaUnited States
| | - Chelsea Hayes
- Cedars‐Sinai Medical CenterDepartment of Pathology and Laboratory MedicineLos AngelesCaliforniaUnited States
| | - Ellen Klapper
- Cedars‐Sinai Medical CenterDepartment of Pathology and Laboratory MedicineLos AngelesCaliforniaUnited States
| | - David R. Gibb
- Cedars‐Sinai Medical CenterDepartment of Pathology and Laboratory MedicineLos AngelesCaliforniaUnited States
| |
Collapse
|
26
|
Juskewitch JE, Tauscher CD, Moldenhauer SK, Schieber JE, Jacob EK, DiGuardo MA. To Give or Not to Give: RhD Immunoglobulin for an RHD * 39 Pregnant Woman with Sickle Cell Disease. Transfus Med Hemother 2021; 48:244-249. [PMID: 34539319 DOI: 10.1159/000512644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/27/2020] [Indexed: 11/19/2022] Open
Abstract
Introduction Patients with sickle cell disease (SCD) have repeated episodes of red blood cell (RBC) sickling and microvascular occlusion that manifest as pain crises, acute chest syndrome, and chronic hemolysis. These clinical sequelae usually increase during pregnancy. Given the racial distribution of SCD, patients with SCD are also more likely to have rarer RBC antigen genotypes than RBC donor populations. We present the management and clinical outcome of a 21-year-old pregnant woman with SCD and an RHD*39 (RhD[S103P], G-negative) variant. Case Presentation Ms. S is B positive with a reported history of anti-D, anti-C, and anti-E alloantibodies (anti-G testing unknown). Genetic testing revealed both an RHD*39 and homozygous partial RHCE*ceVS.02 genotype. Absorption/elution testing confirmed the presence of anti-G, anti-C, and anti-E alloantibodies but could not definitively determine the presence/absence of an anti-D alloantibody. Ms. S desired to undergo elective pregnancy termination and the need for postprocedural RhD immunoglobulin (RhIG) was posed. Given that only the G antigen site is changed in an RHD*39 genotype and the potential risk of RhIG triggering a hyperhemolytic episode in an SCD patient, RhIG was not administered. There were no procedural complications. Follow-up testing at 10 weeks showed no increase in RBC alloantibody strength. Discussion/Conclusion Ms. S represents a rare RHD*39 and partial RHCE*ceVS.02 genotype which did not further alloimmunize in the absence of RhIG administration. Her case also highlights the importance of routine anti-G alloantibody testing in women of childbearing age with apparent anti-D and anti-C alloantibodies.
Collapse
Affiliation(s)
| | - Craig D Tauscher
- Division of Transfusion Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Eapen K Jacob
- Division of Transfusion Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
27
|
Proske P, Distelmaier L, Aramayo-Singelmann C, Koliastas N, Iannaccone A, Papathanasiou M, Temme C, Klump H, Lenz V, Koldehoff M, Carpinteiro A, Reinhardt HC, Köninger A, Röth A, Yamamoto R, Dührsen U, Alashkar F. Pregnancies and Neonatal Outcomes in Patients with Sickle Cell Disease (SCD): Still a (High-)Risk Constellation? J Pers Med 2021; 11:jpm11090870. [PMID: 34575647 PMCID: PMC8464744 DOI: 10.3390/jpm11090870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 11/24/2022] Open
Abstract
Background: This monocentric study conducted at the University Hospital of Essen aims to describe maternal and fetal/neonatal outcomes in sickle cell disease (SCD) documented between 1996 to 2021 (N = 53), reflecting the largest monocentric analysis carried out in Germany. Methods/Results: 46 pregnancies in 22 patients were followed. None of the patients died. In total, 35% (11/31) of pregnancies were preterm. 15 pregnancies in eight patients were conceived on hydroxycarbamide (HC), of which nine had a successful outcome and three were terminated prematurely. There was no difference regarding the rate of spontaneous abortions in patients receiving HC compared to HC-naive patients prior to conception. In patients other than HbS/C disease, pregnancies were complicated by vaso-occlusive crises (VOCs)/acute pain crises (APCs) (96%, 23/24); acute chest syndrome (ACS) (13%, 3/24), transfusion demand (79%, 19/24), urinary tract infections (UTIs) (42%, 10/24) and thromboembolic events (8%, 2/24). In HbS/C patients complications included: VOCs/APCs (43%, 3/7; ACS: 14%, 1/7), transfusion demand (14%, 1/7), and UTIs (14%, 1/7). Independent of preterm deliveries, a significant difference with respect to neonatal growth in favor of neonates from HbS/C mothers was observed. Conclusion: Our data support the results of previous studies, highlighting the high rate of maternal and fetal/neonatal complications in pregnant SCD patients.
Collapse
Affiliation(s)
- Pia Proske
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (P.P.); (L.D.); (M.K.); (A.C.); (H.C.R.); (A.R.); (U.D.)
| | - Laura Distelmaier
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (P.P.); (L.D.); (M.K.); (A.C.); (H.C.R.); (A.R.); (U.D.)
- Vivantes, MVZ Neukölln, 12351 Berlin, Germany
| | - Carmen Aramayo-Singelmann
- Department of Pediatrics III, University Children’s Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany;
| | - Nikolaos Koliastas
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, 45147 Essen, Germany; (N.K.); (A.I.); (A.K.)
| | - Antonella Iannaccone
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, 45147 Essen, Germany; (N.K.); (A.I.); (A.K.)
| | - Maria Papathanasiou
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, 45147 Essen, Germany;
| | - Christian Temme
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (C.T.); (H.K.); (V.L.)
| | - Hannes Klump
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (C.T.); (H.K.); (V.L.)
| | - Veronika Lenz
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (C.T.); (H.K.); (V.L.)
| | - Michael Koldehoff
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (P.P.); (L.D.); (M.K.); (A.C.); (H.C.R.); (A.R.); (U.D.)
| | - Alexander Carpinteiro
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (P.P.); (L.D.); (M.K.); (A.C.); (H.C.R.); (A.R.); (U.D.)
- Institute for Molecular Biology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (P.P.); (L.D.); (M.K.); (A.C.); (H.C.R.); (A.R.); (U.D.)
| | - Angela Köninger
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, 45147 Essen, Germany; (N.K.); (A.I.); (A.K.)
- Hospital of the Order of St. John of God Regensburg, Clinic for Gynaecology and Obstetrics, 93049 Regensburg, Germany
| | - Alexander Röth
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (P.P.); (L.D.); (M.K.); (A.C.); (H.C.R.); (A.R.); (U.D.)
| | | | - Ulrich Dührsen
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (P.P.); (L.D.); (M.K.); (A.C.); (H.C.R.); (A.R.); (U.D.)
| | - Ferras Alashkar
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (P.P.); (L.D.); (M.K.); (A.C.); (H.C.R.); (A.R.); (U.D.)
- Correspondence:
| |
Collapse
|
28
|
Zerra PE, Patel SR, Jajosky RP, Arthur CM, McCoy JW, Allen JWL, Chonat S, Fasano RM, Roback JD, Josephson CD, Hendrickson JE, Stowell SR. Marginal zone B cells mediate a CD4 T-cell-dependent extrafollicular antibody response following RBC transfusion in mice. Blood 2021; 138:706-721. [PMID: 33876205 PMCID: PMC8394907 DOI: 10.1182/blood.2020009376] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/30/2021] [Indexed: 01/07/2023] Open
Abstract
Red blood cell (RBC) transfusions can result in alloimmunization toward RBC alloantigens that can increase the probability of complications following subsequent transfusion. An improved understanding of the immune mechanisms that underlie RBC alloimmunization is critical if future strategies capable of preventing or even reducing this process are to be realized. Using the HOD (hen egg lysozyme [HEL] and ovalbumin [OVA] fused with the human RBC antigen Duffy) model system, we aimed to identify initiating immune factors that may govern early anti-HOD alloantibody formation. Our findings demonstrate that HOD RBCs continuously localize to the marginal sinus following transfusion, where they colocalize with marginal zone (MZ) B cells. Depletion of MZ B cells inhibited immunoglobulin M (IgM) and IgG anti-HOD antibody formation, whereas CD4 T-cell depletion only prevented IgG anti-HOD antibody development. HOD-specific CD4 T cells displayed similar proliferation and activation following transfusion of HOD RBCs into wild-type or MZ B-cell-deficient recipients, suggesting that IgG formation is not dependent on MZ B-cell-mediated CD4 T-cell activation. Moreover, depletion of follicular B cells failed to substantially impact the anti-HOD antibody response, and no increase in antigen-specific germinal center B cells was detected following HOD RBC transfusion, suggesting that antibody formation is not dependent on the splenic follicle. Despite this, anti-HOD antibodies persisted for several months following HOD RBC transfusion. Overall, these data suggest that MZ B cells can initiate and then contribute to RBC alloantibody formation, highlighting a unique immune pathway that can be engaged following RBC transfusion.
Collapse
Affiliation(s)
- Patricia E Zerra
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, and
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA and
| | - Seema R Patel
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA and
| | - Ryan Philip Jajosky
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, and
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; and
| | - Connie M Arthur
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, and
| | - James W McCoy
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, and
| | - Jerry William Lynn Allen
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; and
| | - Satheesh Chonat
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA and
| | - Ross M Fasano
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, and
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA and
| | - John D Roback
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, and
| | - Cassandra D Josephson
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, and
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA and
| | | | - Sean R Stowell
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; and
| |
Collapse
|
29
|
Medved J, Knott BM, Tarrah SN, Li AN, Shah N, Moscovich TC, Boscia AR, Salazar JE, Santhanakrishnan M, Hendrickson JE, Fu X, Zimring JC, Luckey CJ. The lysophospholipid-binding molecule CD1D is not required for the alloimmunization response to fresh or stored RBCs in mice despite RBC storage driving alterations in lysophospholipids. Transfusion 2021; 61:2169-2178. [PMID: 34181769 DOI: 10.1111/trf.16554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Despite the significant adverse clinical consequences of RBC alloimmunization, our understanding of the signals that induce immune responses to transfused RBCs remains incomplete. Though RBC storage has been shown to enhance alloimmunization in the hen egg lysozyme, ovalbumin, and human Duffy (HOD) RBC alloantigen mouse model, the molecular signals leading to immune activation in this system remain unclear. Given that the nonclassical major histocompatibility complex (MHC) Class I molecule CD1D can bind to multiple different lysophospholipids and direct immune activation, we hypothesized that storage of RBCs increases lysophospholipids known to bind CD1D, and further that recipient CD1D recognition of these altered lipids mediates storage-induced alloimmunization responses. STUDY DESIGN AND METHODS We used a mass spectrometry-based approach to analyze the changes in lysophospholipids that are induced during storage of mouse RBCs. CD1D knockout (CD1D-KO) and wild-type (WT) control mice were transfused with stored HOD RBCs to measure the impact of CD1D deficiency on RBC alloimmunization. RESULTS RBC storage results in alterations in multiple lysophospholipid species known to bind to CD1D and activate the immune system. Prior to transfusion, CD1D-deficient mice had lower baseline levels of polyclonal immunoglobulin (IgG) relative to WT mice. In response to stored RBC transfusion, CD1D-deficient mice generated similar levels of anti-HOD IgM and anti-HOD IgG. CONCLUSION Although storage of RBCs leads to alteration of several lysophospholipids known to be capable of binding CD1D, storage-induced RBC alloimmunization responses are not impacted by recipient CD1D deficiency.
Collapse
Affiliation(s)
- Jelena Medved
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Brittney M Knott
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Soraya N Tarrah
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Andria N Li
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Neha Shah
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Tamara C Moscovich
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Alexis R Boscia
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Juan E Salazar
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | | | - Jeanne E Hendrickson
- Departments of Laboratory Medicine and Pediatrics, Yale University, New Haven, Connecticut, USA
| | - Xiaoyun Fu
- Bloodworks NW Research Institute, and Department of Internal Medicine, Division of Hematology, University of Washington School of Medicine, Seattle, Washington, USA
| | - James C Zimring
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Chance John Luckey
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
30
|
Ejaz K, Roback JD, Stowell SR, Sullivan HC. Daratumumab: Beyond Multiple Myeloma. Transfus Med Rev 2021; 35:36-43. [PMID: 34312046 DOI: 10.1016/j.tmrv.2021.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/01/2022]
Abstract
Daratumumab (DARA) is the biological name of an Immunoglobulin G1k human monoclonal antibody. DARA the first-in-class therapy targeting CD38 expressing- plasma cells (PC) and plasma blasts. It has been approved for the treatment of multiple myeloma. It is also being examined in the setting of other hematologic malignancies. As DARA targets PCs, it could potentially be used to treat many other disease processes that are antibody mediated. In fact, several case reports and case series report experiences of using DARA to treat a variety of antibody-mediated disorderss. The aim of this review is to present a summary of the literature thus far regarding the application of DARA beyond its uses in multiple myeloma and other hematologic diseases. Specifically, we address uses of DARA as an immunologic modulator in various antibody mediated processes.
Collapse
Affiliation(s)
- Kiran Ejaz
- Department of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - John D Roback
- Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sean R Stowell
- Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Harold C Sullivan
- Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
31
|
Muniz JG, Arnoni C, Medeiros R, Vendrame T, Cortez A, S Afonso J, Latini F, Castilho L, Girão M. Antigen matching for transfusion support in Brazilian female patients with sickle cell disease to reduce RBC alloimmunization. Transfusion 2021; 61:2458-2467. [PMID: 34121200 DOI: 10.1111/trf.16544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/12/2021] [Accepted: 04/05/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Red blood cell (RBC) alloimmunization is a complication of patients with sickle cell disease (SCD) and it has a greater impact on pregnancy, leading to a risk of hemolytic disease of the newborn and reducing blood availability for pregnant women. This study proposed to evaluate antigen matching transfusion protocols, aiming to reduce RBC alloimmunization in Brazilian female patients with SCD. METHODS Samples from female patients with SCD (153) and self-declared Afro-Brazilian donors (307) were genotyped for RBC antigens and RH variants were investigated. The transfusion needs of patients during 1-year period and the number of compatible donors were assessed using three antigen-matching transfusion protocols: prophylactic CEK antigen-matched RBCs, prophylactic extended antigen-matched RBCs, and extended-matched red blood cells (RBCs) only for alloimmunized patients. In addition, RH molecular matching has been proposed for patients carrying variant RHCE. RESULTS Provision of CEK antigen-matched donors would have been possible in 92.4% of transfusion events while provision of prophylactic extended antigen-matched RBCs would cover 88.7% of the transfusion events. Extended antigen matching for alloimmunized patients would be efficient in 99% of the cases. The presence of partial D in 10 patients increased the need of D-negative donors. Compatible donors could be enough for four of the five patients with altered RHCE genotypes in both alleles. CONCLUSION In Brazilians, screening African descent donors allows the implementation of prophylactic CEK and extended antigen-matching transfusion protocols to female patients with SCD to reduce RBC alloimmunization; however, the supply of compatible blood can be impaired for patients with Rh variants.
Collapse
Affiliation(s)
- Janaína Guilhem Muniz
- Technical and Scientific Department, Colsan-Associação Beneficente de Coleta de Sangue, São Paulo, Brazil
| | - Carine Arnoni
- Technical and Scientific Department, Colsan-Associação Beneficente de Coleta de Sangue, São Paulo, Brazil
| | - Rosangela Medeiros
- Technical and Scientific Department, Colsan-Associação Beneficente de Coleta de Sangue, São Paulo, Brazil
| | - Tatiane Vendrame
- Technical and Scientific Department, Colsan-Associação Beneficente de Coleta de Sangue, São Paulo, Brazil
| | - Afonso Cortez
- Technical and Scientific Department, Colsan-Associação Beneficente de Coleta de Sangue, São Paulo, Brazil
| | - José S Afonso
- Departamento de Ginecologia, Unifesp (Universidade Federal de São Paulo), São Paulo, Brazil.,Department of Gynecology, UEA - University of the State of Amazonas, Brazil
| | - Flavia Latini
- Technical and Scientific Department, Colsan-Associação Beneficente de Coleta de Sangue, São Paulo, Brazil
| | - Lilian Castilho
- Hemocentro Campinas, University of Campinas - Unicamp, Campinas, Brazil
| | - Manoel Girão
- Departamento de Ginecologia, Unifesp (Universidade Federal de São Paulo), São Paulo, Brazil
| |
Collapse
|
32
|
Yuan Z, Wei Y, Chen X, He S, Cai K, Zhong M, Huang H, Tong X, Liu Z, Yang X. Anti-JMH alloantibody in inherited JMH-negative patients leads to immunogenic destruction of JMH-positive RBCs. Clin Exp Immunol 2021; 205:182-197. [PMID: 34021913 DOI: 10.1111/cei.13622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/25/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
The clinical significance of the specific anti-John Milton Hagen (JMH) alloantibody in inherited JMH-negative patients remains unclear. During clinical blood transfusion, it is often classified as an anti-JMH autoantibody in acquired JMH-negative patients, which might further lead to the occurrence of haemolysis events. In this study, we found that the proportion of inherited JMH-negative people in the Guangzhou population was 0.41%, based on the study of 243 blood samples by flow cytometry. Gene sequencing analysis revealed two novel variants located in exon 11 (c.1348G>A, p.Ala449Thr) and exon 14 (c.1989G>T, p.Leu663Phe). Specific antigen presentation showed that JMH-positive RBCs (red blood cells) could be internalized by SEMA7A-/- dendritic cells (DCs) and that SEMA7A-/- DCs activated by the semaphorin 7a (Sema7a) protein or JMH-positive erythrocytes further induced activation of CD4+ T cells to secrete interferon (IFN)-γ. Transfusion of JMH-positive RBCs could lead to the production of the specific anti-JMH alloantibody in Sema7a knock-out (KO) C57 mice. After erythrocyte sensitization, complement C3 was specifically fixed, causing the destruction of JMH-positive erythrocytes. The anti-JMH alloantibody caused immunological destruction of JMH-positive erythrocytes and promoted the clearance of JMH-positive RBCs. We should be cautious when making conclusions about the clinical significance of the anti-JMH alloantibody.
Collapse
Affiliation(s)
- Zhaohu Yuan
- Department of Blood Transfusion, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou, Guangdong, China
| | - Yaming Wei
- Department of Blood Transfusion, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou, Guangdong, China
| | - Xiaojie Chen
- Department of Blood Transfusion, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou, Guangdong, China
| | - Shufei He
- Department of Blood Transfusion, Third People's Hospital of Huizhou, Huizhou, Guangdong, China
| | - Kui Cai
- Department of Blood Transfusion, Foshan First People's Hospital, Foshan, Guangdong, China
| | - Minglu Zhong
- Department of Blood Transfusion, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou, Guangdong, China
| | - Huiying Huang
- Department of Blood Transfusion, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou, Guangdong, China
| | - Xinxin Tong
- Department of Blood Transfusion, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou, Guangdong, China
| | - Zhen Liu
- Department of Blood Transfusion, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou, Guangdong, China
| | - Xuexin Yang
- Department of Blood Transfusion, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou, Guangdong, China
| |
Collapse
|
33
|
Rankin A, Darbari D, Campbell A, Webb J, Mo YD, Jacquot C, Delaney M, Luban NLC, Nickel RS. Screening for new red blood cell alloantibodies after transfusion in patients with sickle cell disease. Transfusion 2021; 61:2255-2264. [PMID: 34002408 DOI: 10.1111/trf.16444] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Patients with sickle cell disease (SCD) are frequent recipients of red blood cell (RBC) transfusions and are at risk for RBC alloimmunization. RBC alloimmunization is diagnosed by identifying RBC alloantibodies as part of pre-transfusion testing, but this testing fails to detect alloantibodies that have evanesced. It may be beneficial to screen for new RBC alloantibody development after transfusion before possible antibody evanescence. STUDY DESIGN AND METHODS Our institution started a new initiative for episodically transfused patients with SCD to obtain at least one antibody screen 2-6 months after transfusion as part of their clinical care. A database was created to prospectively track all transfused patients for 1 year and their post-transfusion antibody screen results. Patients received prophylactically CEK-matched RBC units. RESULTS During the study year, 138 patients with SCD received a total of 242 RBC transfusions. Patients with a history of an RBC alloantibody (n = 13, 9.4%) had previously received more RBC units than non alloimmunized patients (median 11 vs. 2 RBC units, p = .0002). A total of 337 post-transfusion antibody screens were obtained in 127 patients (92.0%) with 110 patients (79.7%) having at least one antibody screen 2-6 months post-transfusion. With this prospective testing, two new RBC alloantibodies (anti-C and -M) were identified in two patients. CONCLUSION It is feasible to test for new RBC alloantibody development in most episodically transfused patients with SCD as part of their routine care. The yield of this screening appears low with CEK matching, but it could still provide important information for individual patients.
Collapse
Affiliation(s)
- Alexander Rankin
- Divisions of Hematology and Laboratory Medicine, Children's National Hospital, Washington, District of Columbia, USA.,Pediatric Hematology-Oncology, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Deepika Darbari
- Divisions of Hematology and Laboratory Medicine, Children's National Hospital, Washington, District of Columbia, USA.,Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Andrew Campbell
- Divisions of Hematology and Laboratory Medicine, Children's National Hospital, Washington, District of Columbia, USA.,Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Jennifer Webb
- Divisions of Hematology and Laboratory Medicine, Children's National Hospital, Washington, District of Columbia, USA.,Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Yunchuan Delores Mo
- Divisions of Hematology and Laboratory Medicine, Children's National Hospital, Washington, District of Columbia, USA.,Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Cyril Jacquot
- Divisions of Hematology and Laboratory Medicine, Children's National Hospital, Washington, District of Columbia, USA.,Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Meghan Delaney
- Divisions of Hematology and Laboratory Medicine, Children's National Hospital, Washington, District of Columbia, USA.,Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Naomi L C Luban
- Divisions of Hematology and Laboratory Medicine, Children's National Hospital, Washington, District of Columbia, USA.,Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Robert Sheppard Nickel
- Divisions of Hematology and Laboratory Medicine, Children's National Hospital, Washington, District of Columbia, USA.,Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| |
Collapse
|
34
|
A novel algorithm comprehensively characterizes human RH genes using whole-genome sequencing data. Blood Adv 2021; 4:4347-4357. [PMID: 32915977 DOI: 10.1182/bloodadvances.2020002148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/10/2020] [Indexed: 11/20/2022] Open
Abstract
RHD and RHCE genes encode Rh blood group antigens and exhibit extensive single-nucleotide polymorphisms and chromosome structural changes in patients with sickle cell disease (SCD). RH variation can drive loss of antigen epitopes or expression of new epitopes, predisposing patients with SCD to Rh alloimmunization. Serologic antigen typing is limited to common Rh antigens, necessitating a genetic approach to detect variant antigen expression. We developed a novel algorithm termed RHtyper for RH genotyping from existing whole-genome sequencing (WGS) data. RHtyper determined RH genotypes in an average of 3.4 and 3.3 minutes per sample for RHD and RHCE, respectively. In a validation cohort consisting of 57 patients with SCD, RHtyper achieved 100% accuracy for RHD and 98.2% accuracy for RHCE, when compared with genotypes obtained by RH BeadChip and targeted molecular assays and after verification by Sanger sequencing and independent next-generation sequencing assays. RHtyper was next applied to WGS data from an additional 827 patients with SCD. In the total cohort of 884 patients, RHtyper identified 38 RHD and 28 RHCE distinct alleles, including a novel RHD DAU allele, RHD* 602G, 733C, 744T 1136T. RHtyper provides comprehensive and high-throughput RH genotyping from WGS data, facilitating deconvolution of the extensive RH genetic variation among patients with SCD. We have implemented RHtyper as a cloud-based public access application in DNAnexus (https://platform.dnanexus.com/app/RHtyper), enabling clinicians and researchers to perform RH genotyping with next-generation sequencing data.
Collapse
|
35
|
Antigen density dictates RBC clearance, but not antigen modulation, following incompatible RBC transfusion in mice. Blood Adv 2021; 5:527-538. [PMID: 33496748 DOI: 10.1182/bloodadvances.2020002695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/17/2020] [Indexed: 12/17/2022] Open
Abstract
Incompatible red blood cell (RBC) transfusion can result in life-threatening transfusion complications that can be challenging to manage in patients with transfusion-dependent anemia. However, not all incompatible RBC transfusions result in significant RBC removal. One factor that may regulate the outcome of incompatible RBC transfusion is the density of the incompatible antigen. Despite the potential influence of target antigen levels during incompatible RBC transfusion, a model system capable of defining the role of antigen density in this process has not been developed. In this study, we describe a novel model system of incompatible transfusion using donor mice that express different levels of the KEL antigen and recipients with varying anti-KEL antibody concentrations. Transfusion of KEL+ RBCs that express high or moderate KEL antigen levels results in rapid antibody-mediated RBC clearance. In contrast, relatively little RBC clearance was observed following the transfusion of KEL RBCs that express low KEL antigen levels. Intriguingly, unlike RBC clearance, loss of the KEL antigen from the transfused RBCs occurred at a similar rate regardless of the KEL antigen density following an incompatible transfusion. In addition to antigen density, anti-KEL antibody levels also regulated RBC removal and KEL antigen loss, suggesting that antigen density and antibody levels dictate incompatible RBC transfusion outcomes. These results demonstrate that antibody-induced antigen loss and RBC clearance can occur at distinct antigen density thresholds, providing important insight into factors that may dictate the outcome of an incompatible RBC transfusion.
Collapse
|
36
|
Abstract
Red blood cell (RBC) transfusion is critical in managing acute and chronic complications of sickle cell disease. Alloimmunization and iron overload remain significant complications of transfusion therapy and are minimized with prophylactic Rh and K antigen RBC matching and iron chelation. Matched sibling donor hematopoietic stem cell transplant (HSCT) is a curative therapeutic option. Autologous hematopoietic stem cell (HSC)-based gene therapy has recently shown great promise, for which obtaining sufficient HSCs is essential for success. This article discusses RBC transfusion indications and complications, transfusion support during HSCT, and HSC mobilization and collection for autologous HSCT with gene therapy.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Pathology, St. Jude Children's Research Hospital, MS 342, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Stella T Chou
- Department of Pediatrics, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, 3615 Civic Center Boulevard, Abramson Research Center Room 316D, Philadelphia, PA 19010, USA.
| |
Collapse
|
37
|
Pediatric Hemovigilance and Adverse Transfusion Reactions. Clin Lab Med 2020; 41:51-67. [PMID: 33494885 DOI: 10.1016/j.cll.2020.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Some types of transfusion reactions occur more frequently in the pediatric than the adult population. Allergic reactions are the most common, followed by nonhemolytic transfusion reactions; male children seem most susceptible to such reactions. Platelets are often implicated and pulmonary reactions are understudied in children. Clinical sequelae in neonates, such as bronchopulmonary dysplasia/chronic lung disease and intraventricular hemorrhage, have received increasing attention in relation to transfusion. There is a need to better understand the pathophysiology of transfusion reactions in neonatal and pediatric populations so preventive strategies can be undertaken. There is also a need for robust hemovigilance systems.
Collapse
|
38
|
Hendrickson JE. Red blood cell alloimmunization and sickle cell disease: a narrative review on antibody induction. ANNALS OF BLOOD 2020; 5:33. [PMID: 33554044 PMCID: PMC7861514 DOI: 10.21037/aob-2020-scd-01] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The high prevalence of red blood cell (RBC) alloantibodies in people with sickle cell disease (SCD) cannot be debated. Why people with SCD are so likely to form RBC alloantibodies, however, remains poorly understood. Over the past decade, a better understanding of non-ABO blood group antigen variants has emerged; RH genetic diversity and the role this diversity plays in RBC alloimmunization is discussed elsewhere. Outside of antigen variants, the immune systems of people with SCD are known to be different than those of people without SCD. Some of these differences are due to effects of free heme, whereas others are impacted by hyposplenism. Descriptive studies of differences in white blood cell (WBC) subsets, platelet counts and function, and complement activation between people with SCD and race-matched controls exist. Studies comparing the immune systems of alloimmunized people with SCD to non-alloimmunized people with SCD to race-matched controls without SCD have uncovered differences in T-cell subsets, monocytes, Fcγ receptor polymorphisms, and responses to free heme. Studies in murine models have documented the role that recipient inflammation plays in RBC alloantibody formation, with human studies reporting a similar association. Murine studies have also reported the importance of type 1 interferon (IFNα/β), known to play a pivotal role in autoimmunity, in RBC alloantibody formation. The goal of this manuscript is to review existing data on factors influencing RBC alloantibody induction in people with SCD with a focus on inflammation and other immune system considerations, from the bench to the bedside.
Collapse
Affiliation(s)
- Jeanne E. Hendrickson
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
39
|
Lee JY, Madany E, El Kadi N, Pandya S, Ng K, Yamashita M, Jefferies CA, Gibb DR. Type 1 Interferon Gene Signature Promotes RBC Alloimmunization in a Lupus Mouse Model. Front Immunol 2020; 11:584254. [PMID: 33101313 PMCID: PMC7546415 DOI: 10.3389/fimmu.2020.584254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/03/2020] [Indexed: 11/28/2022] Open
Abstract
Red blood cell (RBC) transfusion exposes recipients to hundreds of unmatched minor RBC antigens. This exposure can lead to production of alloantibodies that promote clinically significant hemolytic events. Multiple studies have reported an increased frequency of RBC alloimmunization in patients with autoimmunity. However, cellular and molecular mechanisms that underlie autoimmunity-induced alloimmunization have not been reported. Patients with systemic lupus erythematosus (SLE) have a high frequency of alloimmunization and express a type 1 interferon (IFNα/β) gene signature. Thus, we utilized the pristane-induced lupus mouse model to test the hypothesis that inflammation in lupus promotes RBC alloimmunization, and to examine the potential role of IFNα/β. Intraperitoneal injection of pristane, a hydrocarbon oil, led to autoantibody production, glomerulonephritis, and pulmonary hemorrhage in wild type (WT) mice. Pristane treatment significantly induced serum IFNα and expression of multiple interferon-stimulated genes (ISGs) in peripheral blood and peritoneal fluid cells, including inflammatory macrophages. Following transfusion with allogeneic RBCs expressing the KEL glycoprotein, pristane-treated WT mice produced significantly elevated levels of anti-KEL IgM and anti-KEL IgG, compared to untreated mice. Pristane induced comparable levels of inflammatory cells and cytokines in mice lacking the IFNα/β receptor (IFNAR1-/-) or the IFNα/β-inducing transcriptions factors (IRF3/7-/-), compared to WT mice. However, pristane-treated IFNAR1-/- and IRF3/7-/- mice failed to produce ISGs and produced significantly lower levels of transfusion-induced anti-KEL IgG, compared to WT mice. Thus, pristane induction of a lupus-like phenotype promoted alloimmunization to the KEL RBC antigen in an IFNα/β-dependent manner. To our knowledge, this is the first examination of molecular mechanisms contributing to RBC alloimmunization in a model of autoimmunity. These results warrant further investigation of the role of IFNα/β in alloimmunization to other RBC antigens and the contribution of the IFNα/β gene signature to the elevated frequency of alloimmunization in patients with SLE.
Collapse
Affiliation(s)
- June Young Lee
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Emaan Madany
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Najwa El Kadi
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Sumaarg Pandya
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Kessandra Ng
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Michifumi Yamashita
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Caroline A. Jefferies
- Department of Internal Medicine, Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - David R. Gibb
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Division of Transfusion Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
40
|
Torres-Aguilar H, Sosa-Luis SA, Ríos-Ríos WDJ, Romero-Tlalolini MDLÁ, Aguilar-Ruiz SR. Silent red blood cell autoantibodies: Are they naturally occurring or an effect of tolerance loss for a subsequent autoimmune process? Autoimmunity 2020; 53:367-375. [PMID: 32815426 DOI: 10.1080/08916934.2020.1799989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Unexpected anti-red blood cell (RBC) alloantibodies are routinely investigated in immunohematology and blood banking since their existence in pregnant women may induce haemolytic disease of the foetus and newborn, and their presence in donors may induce haemolytic transfusion reactions or hyperacute rejection in solid organ transplantation. Unexpected anti-RBC alloantibodies may target antigens of the most blood types excluding the expected antibodies targeting the ABO antigens. Their incidence in humans was originally linked to alloimmunization events such as blood transfusions, transplants, or pregnancies. But later, many findings revealed their existence in pathogenic processes such as malignancies, infections, and autoimmune diseases; and usually (but not always) associated to autoimmune haemolytic anaemia (AIHA). Nevertheless, unexpected anti-RBC autoantibodies are also occasionally found in healthy individuals in the absence of AIHA and with no history of alloimmunization or the associated pathologic processes. Hence, they are generally known as non-clinically significant, are excluded for typification and called "silent red blood cell autoantibodies (SRBCAA)". This review highlights evidence related to genetic predisposition, molecular mimicry, immune dysregulation, and immune tolerance loss surrounding the existence of anti-RBC antibodies, describing the presence of SRBCAA as possible early witnesses of the development of autoimmune diseases.
Collapse
Affiliation(s)
- Honorio Torres-Aguilar
- Clinical Immunology Research Department of Biochemical Sciences Faculty, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca City, Mexico
| | - Sorely Adelina Sosa-Luis
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - William de Jesús Ríos-Ríos
- Clinical Immunology Research Department of Biochemical Sciences Faculty, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca City, Mexico
| | | | - Sergio Roberto Aguilar-Ruiz
- Molecular Immunology Research Department of Medicine and Surgery Faculty, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca City, Mexico
| |
Collapse
|
41
|
Nickel RS, Flegel WA, Adams SD, Hendrickson JE, Liang H, Tisdale JF, Hsieh MM. 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. [PMID: 32637902 PMCID: PMC7327930 DOI: 10.1016/j.eclinm.2020.100432] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Hematopoietic stem cell transplantation (HSCT) is curative for patients with sickle cell disease (SCD). Prior to HSCT, patients with SCD commonly receive RBC transfusions with some becoming RBC or HLA alloimmunized. This alloimmunization may impact post-HSCT transfusion requirements and donor engraftment. METHODS The study population included patients with SCD transplanted on a single-center nonmyeloablative, HLA-matched sibling HSCT trial at the National Heart, Lung, and Blood Institute (NHLBI) who had a pre-HSCT sample available for HLA class I antibody testing. We evaluated transfusion requirements and engraftment outcomes comparing patients with and without pre-existing HLA and RBC antibodies. FINDINGS Of 36 patients studied, 10 (28%) had HLA class I antibodies and 11 (31%) had a history of RBC alloantibodies. Up to day +45 post-HSCT, patients with HLA antibodies received more platelet transfusions (median 2.5 vs 1, p = 0.042) and those with RBC alloantibodies received more RBC units (median 7 vs 4, p = 0.0059) compared to respective non-alloimmunized patients. HLA alloimmunization was not associated with neutrophil engraftment, donor chimerism, or graft rejection. However, RBC alloimmunization correlated with a decreased donor T cell chimerism at 1 year (median 24% vs 55%, p = 0.035). INTERPRETATION Pre-existing HLA and RBC alloantibodies are clinically significant for patients undergoing HLA-matched nonmyeloablative HSCT. Testing for both HLA and RBC antibodies is important to help estimate transfusion needs peri‑HSCT. The association of lower donor T cell chimerism and pre-existing RBC alloantibodies needs further investigation. FUNDING NIH Clinical Center and NHLBI Intramural Research Program (Z99 CL999999, HL006007-11) and the Thrasher Research Fund.
Collapse
Affiliation(s)
- Robert Sheppard Nickel
- Children's National Hospital, Division of Hematology, 111 Michigan Ave NW, Washington, DC 20010, United States
- The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
- Corresponding author at: Children's National Hospital, Division of Hematology, 111 Michigan Ave NW, Washington, DC 20010, United States.
| | - Willy A. Flegel
- Department of Transfusion Medicine, NIH Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Sharon D. Adams
- Department of Transfusion Medicine, NIH Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Jeanne E. Hendrickson
- Departments of Laboratory Medicine and Pediatrics, Yale School of Medicine, New Haven, CT, United States
| | - Hua Liang
- The George Washington University, Department of Statistics, Washington, DC, United States
| | - John F. Tisdale
- Cellular and Molecular Therapeutics Branch, National Institutes of Health, Bethesda, MD, United States
| | - Matthew M. Hsieh
- Cellular and Molecular Therapeutics Branch, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
42
|
Zerra PE, Arthur CM, Chonat S, Maier CL, Mener A, Shin S, Allen JWL, Baldwin WH, Cox C, Verkerke H, Jajosky RP, Tormey CA, Meeks SL, Stowell SR. Fc Gamma Receptors and Complement Component 3 Facilitate Anti-fVIII Antibody Formation. Front Immunol 2020; 11:905. [PMID: 32582142 PMCID: PMC7295897 DOI: 10.3389/fimmu.2020.00905] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/20/2020] [Indexed: 01/02/2023] Open
Abstract
Anti-factor VIII (fVIII) alloantibodies, which can develop in patients with hemophilia A, limit the therapeutic options and increase morbidity and mortality of these patients. However, the factors that influence anti-fVIII antibody development remain incompletely understood. Recent studies suggest that Fc gamma receptors (FcγRs) may facilitate recognition and uptake of fVIII by recently developed or pre-existing naturally occurring anti-fVIII antibodies, providing a mechanism whereby the immune system may recognize fVIII following infusion. However, the role of FcγRs in anti-fVIII antibody formation remains unknown. In order to define the influence of FcγRs on the development of anti-fVIII antibodies, fVIII was injected into WT or FcγR knockout recipients, followed by evaluation of anti-fVIII antibodies. Anti-fVIII antibodies were readily observed following fVIII injection into FcγR knockouts, with similar anti-fVIII antibody levels occurring in FcγR knockouts as detected in WT mice injected in parallel. As antibodies can also fix complement, providing a potential mechanism whereby anti-fVIII antibodies may influence anti-fVIII antibody formation independent of FcγRs, fVIII was also injected into complement component 3 (C3) knockout recipients in parallel. Similar to FcγR knockouts, C3 knockout recipients developed a robust response to fVIII, which was likewise similar to that observed in WT recipients. As FcγRs or C3 may compensate for each other in recipients only deficient in FcγRs or C3 alone, we generated mice deficient in both FcγRs and C3 to test for potential antibody effector redundancy in anti-fVIII antibody formation. Infusion of fVIII into FcγRs and C3 (FcγR × C3) double knockouts likewise induced anti-fVIII antibodies. However, unlike individual knockouts, anti-fVIII antibodies in FcγRs × C3 knockouts were initially lower than WT recipients, although anti-fVIII antibodies increased to WT levels following additional fVIII exposure. In contrast, infusion of RBCs expressing distinct alloantigens into FcγRs, C3 or FcγR × C3 knockout recipients either failed to change anti-RBC levels when compared to WT recipients or actually increased antibody responses, depending on the target antigen. Taken together, these results suggest FcγRs and C3 can differentially impact antibody formation following exposure to distinct alloantigens and that FcγRs and C3 work in concert to facilitate early anti-fVIII antibody formation.
Collapse
Affiliation(s)
- Patricia E Zerra
- Department of Pathology and Laboratory Medicine, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States.,Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Connie M Arthur
- Department of Pathology and Laboratory Medicine, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| | - Satheesh Chonat
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Cheryl L Maier
- Department of Pathology and Laboratory Medicine, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| | - Amanda Mener
- Department of Pathology and Laboratory Medicine, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| | - Sooncheon Shin
- Department of Pathology and Laboratory Medicine, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| | - Jerry William L Allen
- Department of Pathology and Laboratory Medicine, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| | - W Hunter Baldwin
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Courtney Cox
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Hans Verkerke
- Department of Pathology and Laboratory Medicine, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| | - Ryan P Jajosky
- Department of Pathology and Laboratory Medicine, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| | - Christopher A Tormey
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, United States.,Pathology and Laboratory Medicine Service, VA Conneciticut Healthcare System, West Haven, CT, United States
| | - Shannon L Meeks
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Sean R Stowell
- Department of Pathology and Laboratory Medicine, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
43
|
Shih AW, Yan MTS, Elahie AL, Barty RL, Liu Y, Berardi P, Azzam M, Siddiqui R, Parvizian MK, Mcdougall T, Heddle NM, Al-Habsi KS, Goldman M, Cote J, Athale U, Verhovsek MM. Utilising red cell antigen genotyping and serological phenotyping in sickle cell disease patients to risk-stratify patients for alloimmunisation risk. Transfus Med 2020; 30:263-274. [PMID: 32432400 DOI: 10.1111/tme.12685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 03/23/2020] [Accepted: 04/25/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND Alloimmunisation and haemolytic transfusion reactions (HTRs) can occur in patients with sickle cell disease (SCD) despite providing phenotype-matched red blood cell (RBC) transfusions. Variant RBC antigen gene alleles/polymorphisms can lead to discrepancies in serological phenotyping. We evaluated differences between RBC antigen genotyping and phenotyping methods and retrospectively assessed if partial antigen expression may lead to increased risk of alloimmunisation and HTRs in SCD patients at a tertiary centre in Canada. METHODS RBC antigen phenotyping and genotyping were performed by a reference laboratory on consenting SCD patients. Patient demographic, clinical and transfusion-related data were obtained from a local transfusion registry and chart review after research ethics board approval. RESULTS A total of 106 SCD patients were enrolled, and 91% (n = 96) showed additional clinically relevant genotyping information when compared to serological phenotyping alone. FY*02N.01 (FY*B GATA-1) (n = 95; 90%) and RH variant alleles (n = 52, 49%; majority accompanied by FY*02N.01) were common, the latter with putative partial antigen expression in 25 patients. Variability in genotype-phenotype antigen prediction occurred mostly in the Rh system, notably with the e antigen (kappa: 0.17). Fifteen (14.2%) patients had a history of alloimmunisation, with five having HTR documented; no differences in clinical outcomes were found in patients with partial antigen expression. Genotype/extended-phenotype matching strategies may have prevented alloimmunisation events. CONCLUSION We show a high frequency of variant alleles/polymorphisms in the SCD population, where genotyping may complement serological phenotyping. Genotyping SCD patients before transfusion may prevent alloimmunisation and HTRs, and knowledge of the FY*02N.01 variant allele increases feasibility of finding compatible blood.
Collapse
Affiliation(s)
- Andrew W Shih
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Matthew T S Yan
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Medical Services and Hospital Relations, Canadian Blood Services, Vancouver, British Columbia, Canada
| | - Allahna L Elahie
- Hamilton Regional Laboratory Medicine Program, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Rebecca L Barty
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,McMaster Centre for Transfusion Research, McMaster University, Hamilton, Ontario, Canada
| | - Yang Liu
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,McMaster Centre for Transfusion Research, McMaster University, Hamilton, Ontario, Canada
| | - Philip Berardi
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Mona Azzam
- Department of Pediatrics, Suez Canal University, Ismailia, Egypt
| | - Reda Siddiqui
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Michael K Parvizian
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,McMaster Centre for Transfusion Research, McMaster University, Hamilton, Ontario, Canada
| | - Tara Mcdougall
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Nancy M Heddle
- Hamilton Regional Laboratory Medicine Program, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Centre for Innovation, Canadian Blood Services, Hamilton, Ontario, Canada
| | - Khalid S Al-Habsi
- Department of Blood Banks Services, Directorate General of Specialized Medical Care, Ministry of Health, Muscat, Oman
| | - Mindy Goldman
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Jacqueline Cote
- National Immunohematology Reference Laboratory, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Uma Athale
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Madeleine M Verhovsek
- Hamilton Regional Laboratory Medicine Program, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,McMaster Centre for Transfusion Research, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
44
|
Yamada C, Takeshita A, Ohto H, Ishimaru K, Kawabata K, Nomaguchi Y, Haraguchi Y, Abe M, Sobue K, Takenouchi H, Takadate J, Kamimura M, Katai A, Kasai D, Minami Y, Sugimoto T, Michino J, Nagai K, Kumagai M, Hasegawa Y, Ishizuka K, Ohtomo N, Yamada N, Muroi K, Matsushita T, Takahashi K. A Japanese multi‐institutional collaborative study of antigen‐positive red blood cell (RBC) transfusions in patients with corresponding RBC antibodies. Vox Sang 2020; 115:456-465. [DOI: 10.1111/vox.12906] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Chiaki Yamada
- Transfusion and Cell Therapy Hamamatsu University School of Medicine Hamamatsu Japan
| | - Akihiro Takeshita
- Transfusion and Cell Therapy Hamamatsu University School of Medicine Hamamatsu Japan
| | - Hitoshi Ohto
- Department of Blood Transfusion and Transplantation Immunology Fukushima Medical University Fukushima Japan
| | - Ken Ishimaru
- Blood Service Headquarters Japanese Red Cross Society Minato‐ku Japan
| | - Kinuyo Kawabata
- Department of Blood Transfusion and Transplantation Immunology Fukushima Medical University Fukushima Japan
| | - Yuriko Nomaguchi
- Division of Transfusion Medicine Fukuoka University Fukuoka Japan
| | - Yasue Haraguchi
- Department of Blood Transfusion Medicine and Cell Therapy Kagoshima University Kagoshima Japan
| | - Misao Abe
- Blood Transfusion & Cell Therapy Kansai Medical University Moriguchi Japan
| | - Koki Sobue
- Division of Blood Transfusion Toho University Ota‐ku Japan
| | - Hiroyuki Takenouchi
- Department of Transfusion and Cell Therapy University of Miyazaki Miyazaki Japan
| | - Junko Takadate
- Division of Central Clinical Laboratory Iwate Medical University Morioka Japan
| | - Masami Kamimura
- Division of Blood Transfusion Niigata University Niigata Japan
| | - Akiko Katai
- Department of Transfusion Medicine Aichi Medical University Aichi‐gun Japan
| | - Daisuke Kasai
- Department of Clinical Laboratory Nagano Municipal Hospital Nagano Japan
| | - Yumiko Minami
- Division of Transfusion Medicine Osaka Medical College Takatsuki Japan
| | - Tatsuya Sugimoto
- Division of Medical Technology and Department of Blood Transfusion Service Tokai University Isehara Japan
| | - Junko Michino
- Division of Clinical Laboratory, Transfusion Medicine and Cell Therapy University of Toyama Toyama Japan
| | - Kazuhiro Nagai
- Transfusion and Cell Therapy Unit Nagasaki University Nagasaki Japan
| | - Mikako Kumagai
- Division of Blood Transfusion Akita University Akita Japan
| | - Yuichi Hasegawa
- Department of Transfusion Medicine University of Tsukuba Tsukuba Japan
| | - Keiko Ishizuka
- Transfusion and Cell Therapy Hamamatsu University School of Medicine Hamamatsu Japan
| | - Naoki Ohtomo
- Center for Transfusion Medicine and Cell Therapy Tokyo Medical and Dental University Bunkyo‐ku Japan
| | - Naotomo Yamada
- Department of Transfusion Medicine Saga University Saga Japan
| | - Kazuo Muroi
- Division of Cell Transplantation and Transfusion Jichi Medical University Shimotsuke Japan
| | | | - Koki Takahashi
- Blood Service Headquarters Japanese Red Cross Society Minato‐ku Japan
| |
Collapse
|
45
|
Thein SL, Pirenne F, Fasano RM, Habibi A, Bartolucci P, Chonat S, Hendrickson JE, Stowell SR. Hemolytic transfusion reactions in sickle cell disease: underappreciated and potentially fatal. Haematologica 2020; 105:539-544. [PMID: 32029505 PMCID: PMC7049330 DOI: 10.3324/haematol.2019.224709] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/18/2019] [Indexed: 12/20/2022] Open
Affiliation(s)
- Swee Lay Thein
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MA, USA
| | - France Pirenne
- Etablissement Français du Sang, INSERM U955, Université Paris Est Créteil, Créteil, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| | - Ross M Fasano
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA, USA.,Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Anoosha Habibi
- Laboratoire d'Excellence GR-Ex, Paris, France.,Sickle Cell Referral Center, Department of Internal Medicine, Henri-Mondor University Hospital- UPEC, AP-HP, Créteil, France
| | - Pablo Bartolucci
- Sickle Cell Referral Center, Department of Internal Medicine, Henri-Mondor University Hospital- UPEC, AP-HP, Créteil, France
| | - Satheesh Chonat
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Jeanne E Hendrickson
- Departments of Laboratory Medicine and Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Sean R Stowell
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
46
|
Chou ST, Alsawas M, Fasano RM, Field JJ, Hendrickson JE, Howard J, Kameka M, Kwiatkowski JL, Pirenne F, Shi PA, Stowell SR, Thein SL, Westhoff CM, Wong TE, Akl EA. American Society of Hematology 2020 guidelines for sickle cell disease: transfusion support. Blood Adv 2020; 4:327-355. [PMID: 31985807 PMCID: PMC6988392 DOI: 10.1182/bloodadvances.2019001143] [Citation(s) in RCA: 248] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/20/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Red cell transfusions remain a mainstay of therapy for patients with sickle cell disease (SCD), but pose significant clinical challenges. Guidance for specific indications and administration of transfusion, as well as screening, prevention, and management of alloimmunization, delayed hemolytic transfusion reactions (DHTRs), and iron overload may improve outcomes. OBJECTIVE Our objective was to develop evidence-based guidelines to support patients, clinicians, and other healthcare professionals in their decisions about transfusion support for SCD and the management of transfusion-related complications. METHODS The American Society of Hematology formed a multidisciplinary panel that was balanced to minimize bias from conflicts of interest and that included a patient representative. The panel prioritized clinical questions and outcomes. The Mayo Clinic Evidence-Based Practice Research Program supported the guideline development process. The Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach was used to form recommendations, which were subject to public comment. RESULTS The panel developed 10 recommendations focused on red cell antigen typing and matching, indications, and mode of administration (simple vs red cell exchange), as well as screening, prevention, and management of alloimmunization, DHTRs, and iron overload. CONCLUSIONS The majority of panel recommendations were conditional due to the paucity of direct, high-certainty evidence for outcomes of interest. Research priorities were identified, including prospective studies to understand the role of serologic vs genotypic red cell matching, the mechanism of HTRs resulting from specific alloantigens to inform therapy, the role and timing of regular transfusions during pregnancy for women, and the optimal treatment of transfusional iron overload in SCD.
Collapse
Affiliation(s)
- Stella T Chou
- Division of Hematology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Mouaz Alsawas
- Mayo Clinic Evidence-Based Practice Research Program, Mayo Clinic, Rochester, MN
| | - Ross M Fasano
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Joshua J Field
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Jeanne E Hendrickson
- Department of Laboratory Medicine and
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Jo Howard
- Department of Haematological Medicine, King's College London, London, United Kingdom
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Michelle Kameka
- Nicole Wertheim College of Nursing and Health Sciences, Florida International University, Miami, FL
| | - Janet L Kwiatkowski
- Division of Hematology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - France Pirenne
- INSERM-U955, Laboratory of Excellence, French Blood Establishment, Créteil, France
| | | | - Sean R Stowell
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Connie M Westhoff
- Laboratory of Immunohematology and Genomics, New York Blood Center, New York, NY
| | - Trisha E Wong
- Division of Hematology/Oncology, Department of Pediatrics, Oregon Health and Science University, Portland, OR; and
| | - Elie A Akl
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
47
|
Jariwala K, Mishra K, Ghosh K. Comparative study of alloimmunization against red cell antigens in sickle cell disease & thalassaemia major patients on regular red cell transfusion. Indian J Med Res 2019; 149:34-40. [PMID: 31115372 PMCID: PMC6507543 DOI: 10.4103/ijmr.ijmr_940_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Background & objectives: Sickle cell disease (SCD) patients require red cell transfusion during different clinical complications of the disease. Such patients are at a high risk for developing alloantibody against red cell antigens. From India, there are limited data available on alloantibody formation in multiply transfused SCD patients. The present study was thus undertaken to fill up this lacunae by looking at the development of red cell alloantibodies in SCD and β-thalassaemia patients on regular transfusion. Methods: All sickle cell disease patients undergoing red cell transfusion between 2008 and 2016, were included. During this period, a large number of β-thalassaemia major patients also underwent regular red cell transfusion. These thalassaemia patients were also included to compare the tendency of antibody formation between SCD and β-thalassaemia major patients. All patients before regular transfusion were regularly assessed for the development of red cell antibody. Red cell antigen, antibody screen crossmatch and antibody identification were done using the standard technique. Results: A total of 138 patients with SCD aged between 4 and 53 yr (mean 17.6 yr) consisting of 83 males and 55 females (male:female, 1.5:1) along with 333 transfusion-dependent β-thalassaemia patients were studied. Over the last eight years, 15 patients with SCD and four patients with thalassaemia developed alloantibody (P<0.001). Antibody specificity of their alloantibodies was against Rhc, RhE, Kell, Fya and Fyb only. Sickle cell disease patients with and without alloantibody required on the average 11.8 and 8.6 units of red cell concentrate, respectively (P<0.05). Interpretation & conclusions: About 11 per cent of the transfused sickle cells patients developed alloantibodies. The antibody specificity was restricted to Rh, Kell and Duffy blood group systems. Extended antigen matching involving Rh, Kell and Duffy antigens may prevent alloantibody in such patients.
Collapse
|
48
|
Catastrophic Delayed Hemolytic Transfusion Reaction in a Patient With Sickle Cell Disease Without Alloantibodies: Case Report and Review of Literature. J Pediatr Hematol Oncol 2019; 41:624-626. [PMID: 30179992 DOI: 10.1097/mph.0000000000001307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
While packed red blood cell (PRBC) transfusion therapy is a mainstay in the treatment of certain patients with sickle cell disease (SCD) and the standard of care for preoperative management, there are associated risks. Delayed hemolytic transfusion reaction (DHTR) is a risk of PRBC transfusion occurring 2 to 20 days from transfusion and typically presents with severe pain characteristic of vaso-occlusive crisis, fever, and hemolytic anemia. DHTRs are uncommon, occurring in only 4% to 11% of transfused patients with SCD, but may be catastrophic in nature with progression to multiorgan failure within hours. Here, we describe a case of a 20-year-old female with sickle cell SS disease who developed a severe DHTR 5 days following an elective preoperative PRBC transfusion, and rapidly progressed to multiorgan failure and death. This is the first reported case of a catastrophic DHTR in a patient with SCD without any detectable known or new alloantibodies.
Collapse
|
49
|
Tang A, Branscomb J, Zhou M, Snyder A, Eckman J. Characterizing complication risk from multisite, intermittent transfusions for the treatment of sickle cell disease. Pediatr Blood Cancer 2019; 66:e27921. [PMID: 31318157 DOI: 10.1002/pbc.27921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/22/2019] [Accepted: 06/24/2019] [Indexed: 11/11/2022]
Abstract
Blood transfusions are indicated for some acute complications of sickle cell disease (SCD). To characterize the SCD population at increased risk of transfusion-associated complications, Georgia hospital discharge data were used to estimate the frequency of intermittent transfusions and the proportion of patients receiving them at multiple institutions. Ten years of data (2007-2016) showed almost 19% of patients with SCD (1585/8529) received transfusions at more than one hospital. The likelihood of multisite transfusions increased from ages 18 through 40 and with the number of transfusions received. The results support the need to track and share transfusion histories in order to reduce complication risks.
Collapse
Affiliation(s)
- Amy Tang
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia.,Department of Pediatrics, Division of Hematology/Oncology, Emory University School of Medicine, Atlanta, Georgia
| | - Jane Branscomb
- Georgia Health Policy Center, Andrew Young School of Policy Studies, Georgia State University, Atlanta, Georgia
| | - Mei Zhou
- Georgia Health Policy Center, Andrew Young School of Policy Studies, Georgia State University, Atlanta, Georgia
| | - Angela Snyder
- Georgia Health Policy Center, Andrew Young School of Policy Studies, Georgia State University, Atlanta, Georgia
| | - James Eckman
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
50
|
Arthur CM, Chonat S, Fasano R, Yee MEM, Josephson CD, Roback JD, Stowell SR. Examining the Role of Complement in Predicting, Preventing, and Treating Hemolytic Transfusion Reactions. Transfus Med Rev 2019; 33:217-224. [PMID: 31679762 PMCID: PMC7147990 DOI: 10.1016/j.tmrv.2019.09.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/24/2022]
Abstract
Red blood cell (RBC) transfusion is a critical component of optimal management for a broad range of conditions. Regardless of the indication, pretransfusion testing is required to appropriately match RBC donors and recipients to provide immunologically compatible blood. Although this approach is effective in the vast majority of situations, occasionally, patients will inadvertently receive an incompatible RBC transfusion, which can result in a hemolytic transfusion reaction (HTR). In addition, patients with life-threatening anemia and a complex alloantibody profile, which precludes rapid procurement of compatible RBCs, may also receive incompatible RBCs, placing them at risk for an HTR. Despite the rarity of these clinical situations, when incompatible blood transfusion results in an HTR, the consequences can be devastating. In this review, we will explore the challenges associated with actively preventing and treating acute HTRs following incompatible RBC transfusion. In doing so, we will focus primarily on the role of complement, not only as a key player in HTRs, but also as a potential target for the prevention and treatment of HTRs.
Collapse
Affiliation(s)
- Connie M Arthur
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Satheesh Chonat
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA
| | - Ross Fasano
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA; Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA
| | - Marianne E M Yee
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA; Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA
| | - Cassandra D Josephson
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA; Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA
| | - John D Roback
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA
| | - Sean R Stowell
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA.
| |
Collapse
|