Informacja

Drogi użytkowniku, aplikacja do prawidłowego działania wymaga obsługi JavaScript. Proszę włącz obsługę JavaScript w Twojej przeglądarce.

Tytuł pozycji:

Autoimmune cytopenias following allogeneic hematopoietic stem cell transplant in pediatric patients: Response to therapy and late effects.

Tytuł:
Autoimmune cytopenias following allogeneic hematopoietic stem cell transplant in pediatric patients: Response to therapy and late effects.
Autorzy:
Koo J; Children's Hospital Colorado, Division of Pediatric Hematology/Oncology/Bone Marrow Transplant, Colorado, University of Colorado Anschutz Medical Campus, Aurora.
Giller RH; Children's Hospital Colorado, Division of Pediatric Hematology/Oncology/Bone Marrow Transplant, Colorado, University of Colorado Anschutz Medical Campus, Aurora.
Quinones R; Children's Hospital Colorado, Division of Pediatric Hematology/Oncology/Bone Marrow Transplant, Colorado, University of Colorado Anschutz Medical Campus, Aurora.
McKinney CM; Children's Hospital Colorado, Division of Pediatric Hematology/Oncology/Bone Marrow Transplant, Colorado, University of Colorado Anschutz Medical Campus, Aurora.
Verneris MR; Children's Hospital Colorado, Division of Pediatric Hematology/Oncology/Bone Marrow Transplant, Colorado, University of Colorado Anschutz Medical Campus, Aurora.
Knight-Perry J; Children's Hospital Colorado, Division of Pediatric Hematology/Oncology/Bone Marrow Transplant, Colorado, University of Colorado Anschutz Medical Campus, Aurora.
Źródło:
Pediatric blood & cancer [Pediatr Blood Cancer] 2020 Sep; Vol. 67 (9), pp. e28591. Date of Electronic Publication: 2020 Jul 13.
Typ publikacji:
Journal Article
Język:
English
Imprint Name(s):
Original Publication: Hoboken, N.J. : John Wiley, c 2004-
MeSH Terms:
Anemia, Hemolytic, Autoimmune/*mortality
Graft vs Host Disease/*mortality
Hematologic Neoplasms/*mortality
Hematopoietic Stem Cell Transplantation/*mortality
Adolescent ; Adult ; Anemia, Hemolytic, Autoimmune/etiology ; Anemia, Hemolytic, Autoimmune/pathology ; Case-Control Studies ; Child ; Child, Preschool ; Female ; Follow-Up Studies ; Graft vs Host Disease/etiology ; Graft vs Host Disease/pathology ; Hematologic Neoplasms/pathology ; Hematologic Neoplasms/therapy ; Hematopoietic Stem Cell Transplantation/adverse effects ; Humans ; Infant ; Male ; Prognosis ; Retrospective Studies ; Survival Rate ; Young Adult
References:
Chang TY, Jaing TH, Wen YC, Huang IA, Chen SH, Tsay PK. Risk factor analysis of autoimmune hemolytic anemia after allogeneic hematopoietic stem cell transplantation in children. Medicine (Baltimore). 2016;95(46):e5396.
Holbro A, Abinun M, Daikeler T. Management of autoimmune diseases after haematopoietic stem cell transplantation. Br J Haematol. 2012;157(3):281-290.
Wang M, Wang W, Abeywardane A, et al. Autoimmune hemolytic anemia after allogeneic hematopoietic stem cell transplantation: analysis of 533 adult patients who underwent transplantation at King's College Hospital. Biol Blood Marrow Transplant. 2015;21(1):60-66.
Faraci M, Zecca M, Pillon M, et al. Autoimmune hematological diseases after allogeneic hematopoietic stem cell transplantation in children: an Italian multicenter experience. Biol Blood Marrow Transplant. 2014;20(2):272-278.
Neely JA, Dvorak CC, Pantell MS, Melton A, Huang JN, Shimano KA. Autoimmune cytopenias in pediatric hematopoietic cell transplant patients. Front Pediatr. 2019;7:171.
Even-Or E, Naser Eddin A, Shadur B, et al. Successful treatment with daratumumab for post-HSCT refractory hemolytic anemia. Pediatr Blood Cancer. 2019;67(1):e28010.
Miller PDE, Snowden JA, De Latour RP, et al. Autoimmune cytopenias (AIC) following allogeneic haematopoietic stem cell transplant for acquired aplastic anaemia: a joint study of the Autoimmune Diseases and Severe Aplastic Anaemia Working Parties (ADWP/SAAWP) of the European Society for Blood and Marrow Transplantation (EBMT). Bone Marrow Transplant. 2019;55(2):441-451.
Szanto CL, Langenhorst J, de Koning C, et al. Predictors for autoimmune cytopenias after allogeneic hematopoietic cell transplantation in children. Biol Blood Marrow Transplant. 2019;26(1):114-122.
Neunert CE, Despotovic JM. Autoimmune hemolytic anemia and immune thrombocytopenia following hematopoietic stem cell transplant: a critical review of the literature. Pediatr Blood Cancer. 2019;66(4):e27569.
Waespe N, Zeilhofer U, Gungor T. Treatment-refractory multi-lineage autoimmune cytopenia after unrelated cord blood transplantation: remission after combined bortezomib and vincristine treatment. Pediatr Blood Cancer. 2014;61(11):2112-2114.
Raj A, Bertolone S, Cheerva A. Successful treatment of refractory autoimmune hemolytic anemia with monthly rituximab following nonmyeloablative stem cell transplantation for sickle cell disease. J Pediatr Hematol Oncol. 2004;26(5):312-314.
Barmettler S, Ong MS, Farmer JR, Choi H, Walter J. Association of immunoglobulin levels, infectious risk, and mortality with rituximab and hypogammaglobulinemia. JAMA Netw Open. 2018;1(7):e184169.
Roberts DM, Jones RB, Smith RM, et al. Rituximab-associated hypogammaglobulinemia: incidence, predictors and outcomes in patients with multi-system autoimmune disease. J Autoimmun. 2015;57:60-65.
Chiou FK, Beath SV, Patel M, Gupte GL. Hypogammaglobulinemia and bacterial infections following pediatric post-transplant lymphoproliferative disorder in the rituximab era. Pediatr Transplant. 2019;23(6):e13519.
Kado R, Sanders G, McCune WJ. Diagnostic and therapeutic considerations in patients with hypogammaglobulinemia after rituximab therapy. Curr Opin Rheumatol. 2017;29(3):228-233.
Sanz J, Arriaga F, Montesinos P, et al. Autoimmune hemolytic anemia following allogeneic hematopoietic stem cell transplantation in adult patients. Bone Marrow Transplant. 2007;39(9):555-561.
Neunert C, Lim W, Crowther M, et al. The American Society of Hematology 2011 evidence-based practice guideline for immune thrombocytopenia. Blood. 2011;117(16):4190-4207.
Wang WC. Evans syndrome in childhood: pathophysiology, clinical course, and treatment. Am J Pediatr Hematol Oncol. 1988;10(4):330-338.
Michel M, Chanet V, Dechartres A, et al. The spectrum of Evans syndrome in adults: new insight into the disease based on the analysis of 68 cases. Blood. 2009;114(15):3167-3172.
Kruizinga MD, van Tol MJD, Bekker V, et al. Risk factors, treatment, and immune dysregulation in autoimmune cytopenia after allogeneic hematopoietic stem cell transplantation in pediatric patients. Biol Blood Marrow Transplant. 2018;24(4):772-778.
Hwang-Bo S, Kim SK, Lee JW, et al. Treatment and response of autoimmune cytopenia occurring after allogeneic hematopoietic cell transplantation in children. Blood Res. 2017;52(2):119-124.
Khalil A, Zaidman I, Bergman R, Elhasid R, Ben-Arush MW. Autoimmune complications after hematopoietic stem cell transplantation in children with nonmalignant disorders. Scientific World Journal. 2014;2014:581657.
Dvorak CC, Bollard CM, El-Bietar J, Filipovich A. Complications of transplant for nonmalignant disorders: autoimmune cytopenias, opportunistic infections, and PTLD. Biol Blood Marrow Transplant. 2012;18(1 Suppl):S101-110.
Page KM, Mendizabal AM, Prasad VK, et al. Posttransplant autoimmune hemolytic anemia and other autoimmune cytopenias are increased in very young infants undergoing unrelated donor umbilical cord blood transplantation. Biol Blood Marrow Transplant. 2008;14(10):1108-1117.
Szanto CL, Langenhorst J, de Koning C, et al. Predictors for autoimmune cytopenias after allogeneic hematopoietic cell transplantation in children. Biol Blood Marrow Transplant. 2020;26(1):114-122.
Pierini A, Nishikii H, Baker J, et al. Foxp3(+) regulatory T cells maintain the bone marrow microenvironment for B cell lymphopoiesis. Nat Commun. 2017;8:15068.
Nguyen VH, Shashidhar S, Chang DS, et al. The impact of regulatory T cells on T-cell immunity following hematopoietic cell transplantation. Blood. 2008;111(2):945-953.
Kohm AP, Carpentier PA, Anger HA, Miller SD. Cutting edge: CD4+CD25+ regulatory T cells suppress antigen-specific autoreactive immune responses and central nervous system inflammation during active experimental autoimmune encephalomyelitis. J Immunol. 2002;169(9):4712-4716.
Trenado A, Sudres M, Tang Q, et al. Ex vivo-expanded CD4+CD25+ immunoregulatory T cells prevent graft-versus-host-disease by inhibiting activation/differentiation of pathogenic T cells. J Immunol. 2006;176(2):1266-1273.
Xiao F, Ma L, Zhao M, et al. Ex vivo expanded human regulatory T cells delay islet allograft rejection via inhibiting islet-derived monocyte chemoattractant protein-1 production in CD34+ stem cells-reconstituted NOD-scid IL2rgammanull mice. PLoS One. 2014;9(3):e90387.
Battaglia M, Stabilini A, Migliavacca B, Horejs-Hoeck J, Kaupper T, Roncarolo MG. Rapamycin promotes expansion of functional CD4+CD25+FOXP3+ regulatory T cells of both healthy subjects and type 1 diabetic patients. J Immunol. 2006;177(12):8338-8347.
Stallone G, Infante B, Di Lorenzo A, Rascio F, Zaza G, Grandaliano G. mTOR inhibitors effects on regulatory T cells and on dendritic cells. J Transl Med. 2016;14(1):152.
Schuetz C, Hoenig M, Moshous D, et al. Daratumumab in life-threatening autoimmune hemolytic anemia following hematopoietic stem cell transplantation. Blood Adv. 2018;2(19):2550-2553.
de Montalembert M, Ribeil JA, Brousse V, et al. Cardiac iron overload in chronically transfused patients with thalassemia, sickle cell anemia, or myelodysplastic syndrome. PLoS One. 2017;12(3):e0172147.
Maximova N, Gregori M, Boz G, et al. MRI-based evaluation of multiorgan iron overload is a predictor of adverse outcomes in pediatric patients undergoing allogeneic hematopoietic stem cell transplantation. Oncotarget. 2017;8(45):79650-79661.
Khojah AM, Miller ML, Klein-Gitelman MS, et al. Rituximab-associated hypogammaglobulinemia in pediatric patients with autoimmune diseases. Pediatr Rheumatol Online J. 2019;17(1):61.
Ottaviano G, Marinoni M, Graziani S, et al. Rituximab unveils hypogammaglobulinemia and immunodeficiency in children with autoimmune cytopenia. J Allergy Clin Immunol Pract. 2019;8(1):273-282.
Even-Or E, Naser Eddin A, Shadur B, et al. Successful treatment with daratumumab for post-HSCT refractory hemolytic anemia. Pediatr Blood Cancer. 2020;67(1):e28010.
Bathini S, Holtzman NG, Koka R, et al. Refractory postallogeneic stem cell transplant pure red cell aplasia in remission after treatment with daratumumab. Am J Hematol. 2019;94(8):E216-E219.
Barcellini W, Fattizzo B, Zaninoni A. Management of refractory autoimmune hemolytic anemia after allogeneic hematopoietic stem cell transplantation: current perspectives. J Blood Med. 2019;10:265-278.
Blennerhassett R, Sudini L, Gottlieb D, Bhattacharyya A. Post-allogeneic transplant Evans syndrome successfully treated with daratumumab. Br J Haematol. 2019;187(2):e48-e51.
Salas MQ, Alahmari A, Lipton JH. Successful treatment of refractory red cell aplasia after allogeneic hematopoietic cell transplantation with daratumumab. Eur J Haematol. 2020;104(2):145-147.
Cartron G, Watier H, Golay J, Solal-Celigny P. From the bench to the bedside: ways to improve rituximab efficacy. Blood. 2004;104(9):2635-2642.
Cardarelli PM, Quinn M, Buckman D, et al. Binding to CD20 by anti-B1 antibody or F(ab')(2) is sufficient for induction of apoptosis in B-cell lines. Cancer Immunol Immunother. 2002;51(1):15-24.
Contributed Indexing:
Keywords: allogeneic HSCT; autoimmune cytopenia; hypogammaglobulinemia; late effects
Entry Date(s):
Date Created: 20200714 Date Completed: 20201207 Latest Revision: 20201214
Update Code:
20240105
DOI:
10.1002/pbc.28591
PMID:
32658382
Czasopismo naukowe
Background: Autoimmune cytopenias (AICs) are rare, but serious complications of allogeneic hematopoietic cell transplantation (allo-HSCT).
Procedure: We performed a case-control study using 20 pediatric AIC cases and 40 controls, matched by stem cell source and primary indication comparing clinical and transplant characteristics, treatment, outcomes, and late effects.
Results: Cases were more likely to be human leukocyte antigen mismatched (P = 0.04). There was no difference in conditioning regimen, serotherapy use, graft-versus-host disease (GVHD) prophylaxis, incidence of acute or chronic GVHD, ABO compatibility, infections, and donor engraftment. The median time to AIC onset was 219 days (range, 97-1205 days) and AIC resolution was 365 days (range, 10 days to 2737.5 days). First-line therapies for AIC patients most commonly included corticosteroids (75%) and rituximab (55%). Only 25% of patients responded to first-line treatment. At a median of 611.5 days from last rituximab dose, 82.5% patients were still receiving intravenous immune globulin for hypogammaglobulinemia compared with 2.5% of controls (P < 0.0001). Iron overload was higher in AIC patients (P = 0.0004), as was avascular necrosis (P = 0.04). There was no difference in overall survival at one year after HSCT (85% vs 82.5%). Two patients with refractory autoimmune hemolytic anemia responded to daratumumab and had resolution of B-cell aplasia.
Conclusions: In this study, we find poor initial responses to AIC-directed therapies and significant late effects.
(© 2020 Wiley Periodicals LLC.)

Ta witryna wykorzystuje pliki cookies do przechowywania informacji na Twoim komputerze. Pliki cookies stosujemy w celu świadczenia usług na najwyższym poziomie, w tym w sposób dostosowany do indywidualnych potrzeb. Korzystanie z witryny bez zmiany ustawień dotyczących cookies oznacza, że będą one zamieszczane w Twoim komputerze. W każdym momencie możesz dokonać zmiany ustawień dotyczących cookies