Assessment of Patient-Specific Human Leukocyte Antigen Genomic Loss at Relapse After Antithymocyte Globulin-Based T-Cell-Replete Haploidentical Hematopoietic Stem Cell Transplant

Hengwei Wu, Jimin Shi, Yi Luo, Jian Yu, Xiaoyu Lai, Lizhen Liu, Huarui Fu, Guifang Ouyang, Xiaojun Xu, Haowen Xiao, He Huang, Yanmin Zhao, Hengwei Wu, Jimin Shi, Yi Luo, Jian Yu, Xiaoyu Lai, Lizhen Liu, Huarui Fu, Guifang Ouyang, Xiaojun Xu, Haowen Xiao, He Huang, Yanmin Zhao

Abstract

Importance: Patient-specific human leukocyte antigen (HLA) genomic loss (HLA loss) is one of the reputed mechanisms of leukemia immune escape and relapse after haploidentical hematopoietic stem cell transplant (HSCT). However, clinical characteristics and prognosis of this distinct relapse type in the setting of haploidentical HSCT based on antithymocyte globulin (ATG) T-cell-replete conditioning are still unknown, especially for patients with lymphoid leukemia.

Objective: To identify the incidence of and patient characteristics associated with HLA loss at hematologic cancer relapse after ATG-based haploidentical HSCT and to assess overall survival after HLA loss at relapse.

Design, setting, and participants: This retrospective and multicenter case series study used data from medical records to identify patients who experienced relapse of hematologic cancer after receipt of ATG-based haploidentical HSCT. The study included 788 consecutive patients aged 8 to 70 years with lymphoid or myeloid leukemia who received ATG T-cell-replete haploidentical HSCT at the Zhejiang Cooperative Group for Blood and Marrow Transplantation between May 1, 2012, and May 31, 2021.

Exposures: Relapse after ATG-based haploidentical HSCT.

Main outcomes and measures: Incidence, risk factors, and postrelapse overall survival among patients with HLA loss at hematologic cancer relapse after receipt of haploidentical HSCT. Logistic regression analysis was used to identify characteristics associated with the likelihood of HLA loss, and Kaplan-Meier and Cox regression analyses were performed to evaluate postrelapse survival.

Results: A total of 788 patients who received haploidentical HSCT for hematologic cancer were identified, 180 of whom experienced relapse after HSCT. Of those, 106 evaluable patients (median age, 30.9 years [range, 8.3-64.6 years]; 54 female [50.9%] and 52 male [49.1%]) were screened for HLA loss, which was detected in 54 patients (50.9%). Patients with HLA loss experienced relapse later than those without HLA loss (lymphoid group: median, 323 days [range, 98-2056 days] vs 151 days [range, 57-2544 days]; P = .01; myeloid group: median, 321 days [range, 55-1574 days] vs 223 days [range, 68-546 days]; P = .03). Among patients with lymphoid leukemia, those with minimal residual disease positivity before hematologic relapse (odds ratio [OR], 28.47; 95% CI, 1.99-407.98; P = .01), those with chronic graft-vs-host disease (OR, 27.68; 95% CI, 1.40-546.88; P = .03), and those with more than 180 days between HSCT and relapse (OR, 6.91; 95% CI, 1.32-36.22; P = .02) were more likely to lose unshared HLA at relapse, whereas male patients (OR, 0.03; 95% CI, 0.003-0.32; P = .04) were more likely to preserve their HLA genome at relapse. Patients with myeloid leukemia had different factors associated with HLA loss, including underweight status (OR, 0.10; 95% CI, 0.02-0.60; P = .01) and acute graft-vs-host disease (OR, 4.84; 95% CI, 1.14-20.53; P = .03). The receipt of preemptive donor lymphocyte infusion among patients with minimal residual disease recurrence did not postpone hematologic cancer relapse in those with HLA loss (median, 322 days [range, 204-1030 days]) compared with no receipt of donor lymphocyte infusion (median, 340 days [range, 215 days to not available]; P > .99).

Conclusions and relevance: In this study, HLA loss at leukemia relapse occurred frequently after receipt of ATG-based haploidentical HSCT. The identification of risk factors associated with HLA loss would help to prompt screening, avoid potentially harmful infusions of donor T cells, and develop alternative therapeutic strategies.

Conflict of interest statement

Conflict of Interest Disclosures: None reported.

Figures

Figure 1.. Study Flow Diagram
Figure 1.. Study Flow Diagram
Initial records search was performed for patients who had (1) hematologic cancer, (2) age between 8 and 70 years, (3) a low-dose antithymocyte globulin–based haploidentical hematopoietic stem cell transplant (HSCT), and (4) a transplant date between May 2012 and May 2021. A total of 788 records were identified and screened for patients with (1) minimal residual disease–negative during complete remission after haploidentical HSCT, (2) engraftment with 100% donor chimerism after haploidentical HSCT, (3) bone marrow hematologic relapse of minimal residual disease–positive, and (4) donor chimerism less than 97%. HLA indicates human leukocyte antigen.
Figure 2.. Independent Risk Factors Associated With…
Figure 2.. Independent Risk Factors Associated With Human Leukocyte Antigen Loss at Relapse
AUROC indicates area under the receiver operating characteristic curve; GVHD, graft-vs-host disease; HLA, human leukocyte antigen; HSCT, hematopoietic stem cell transplant; and MRD, minimal residual disease.
Figure 3.. Postrelapse Overall Survival
Figure 3.. Postrelapse Overall Survival
Plus signs indicate deaths occurring at specific time points. Boxes outlined by dashes indicate patients who were alive in each group at different time points. HLA indicates human leukocyte antigen; and OS, overall survival.

References

    1. Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood. 2008;112(12):4371-4383. doi:10.1182/blood-2008-03-077974
    1. Janelle V, Rulleau C, Del Testa S, Carli C, Delisle JS. T-cell immunotherapies targeting histocompatibility and tumor antigens in hematological malignancies. Front Immunol. 2020;11:276. doi:10.3389/fimmu.2020.00276
    1. Kanakry CG, Fuchs EJ, Luznik L. Modern approaches to HLA-haploidentical blood or marrow transplantation. Nat Rev Clin Oncol. 2016;13(1):10-24. doi:10.1038/nrclinonc.2015.128
    1. Horowitz M, Schreiber H, Elder A, et al. . Epidemiology and biology of relapse after stem cell transplantation. Bone Marrow Transplant. 2018;53(11):1379-1389. doi:10.1038/s41409-018-0171-z
    1. Marincola FM, Jaffee EM, Hicklin DJ, Ferrone S. Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance. Adv Immunol. 2000;74:181-273. doi:10.1016/s0065-2776(08)60911-6
    1. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331(6024):1565-1570. doi:10.1126/science.1203486
    1. Garrido F, Perea F, Bernal M, Sanchez-Palencia A, Aptsiauri N, Ruiz-Cabello F. The escape of cancer from T cell–mediated immune surveillance: HLA class I loss and tumor tissue architecture. Vaccines (Basel). 2017;5(1):7. doi:10.3390/vaccines5010007
    1. Garrido F, Cabrera T, Concha A, Glew S, Ruiz-Cabello F, Stern PL. Natural history of HLA expression during tumour development. Immunol Today. 1993;14(10):491-499. doi:10.1016/0167-5699(93)90264-L
    1. Masuda K, Hiraki A, Fujii N, et al. . Loss or down-regulation of HLA class I expression at the allelic level in freshly isolated leukemic blasts. Cancer Sci. 2007;98(1):102-108. doi:10.1111/j.1349-7006.2006.00356.x
    1. Booman M, Douwes J, Glas AM, et al. . Mechanisms and effects of loss of human leukocyte antigen class II expression in immune-privileged site-associated B-cell lymphoma. Clin Cancer Res. 2006;12(9):2698-2705. doi:10.1158/1078-0432.CCR-05-2617
    1. Schwindt H, Vater I, Kreuz M, et al. . Chromosomal imbalances and partial uniparental disomies in primary central nervous system lymphoma. Leukemia. 2009;23(10):1875-1884. doi:10.1038/leu.2009.120
    1. Crucitti L, Crocchiolo R, Toffalori C, et al. . Incidence, risk factors and clinical outcome of leukemia relapses with loss of the mismatched HLA after partially incompatible hematopoietic stem cell transplantation. Leukemia. 2015;29(5):1143-1152. doi:10.1038/leu.2014.314
    1. Vago L, Toffalori C, Ahci M, et al. . Incidence of HLA loss in a global multicentric cohort of post-transplantation relapses: results from the HLAloss Collaborative Study. Blood. 2018;132(suppl 1):818. doi:10.1182/blood-2018-99-112142
    1. World Medical Association. World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310(20):2191-2194. doi:10.1001/jama.2013.281053
    1. National Health and Family Planning Commission of the People's Republic of China. [Screening standard for malnutrition of school-age children and adolescents (WS/ T 456-2014)]. Standards Press of China; 2014. Accessed March 3, 2022.
    1. Jagasia MH, Greinix HT, Arora M, et al. . National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. the 2014 Diagnosis and Staging Working Group report. Biol Blood Marrow Transplant. 2015;21(3):389-401. doi:10.1016/j.bbmt.2014.12.001
    1. Harris AC, Young R, Devine S, et al. . International, multicenter standardization of acute graft-versus-host disease clinical data collection: a report from the Mount Sinai Acute GVHD International Consortium. Biol Blood Marrow Transplant. 2016;22(1):4-10. doi:10.1016/j.bbmt.2015.09.001
    1. Ahci M, Toffalori C, Bouwmans E, et al. . A new tool for rapid and reliable diagnosis of HLA loss relapses after HSCT. Blood. 2017;130(10):1270-1273. doi:10.1182/blood-2017-05-784306
    1. Huang XJ. Haploidentical hematopoietic stem cell transplantation without in vitro T-cell–depletion for the treatment of hematologic diseases. Chimerism. 2013;4(1):26-28. doi:10.1038/sj.bmt.1705445
    1. Shen Y, Qi J, Chen J, et al. Allogeneic hematopoietic stem cell transplantation from non-sibling 10/10 HLA-matched related donors: a single-center experience. Haematologica. 2021;106(11):3017-3020. doi:10.3324/haematol.2021.278933
    1. Casucci M, Perna SK, Falcone L, et al. . Graft-versus-leukemia effect of HLA-haploidentical central-memory T-cells expanded with leukemic APCs and modified with a suicide gene. Mol Ther. 2013;21(2):466-475. doi:10.1038/mt.2012.227
    1. Brissot E, Labopin M, Ehninger G, et al. . Haploidentical versus unrelated allogeneic stem cell transplantation for relapsed/refractory acute myeloid leukemia: a report on 1578 patients from the Acute Leukemia Working Party of the EBMT. Haematologica. 2019;104(3):524-532. doi:10.3324/haematol.2017.187450
    1. Ruggeri A, Sun Y, Labopin M, et al. . Post-transplant cyclophosphamide versus anti-thymocyte globulin as graft- versus-host disease prophylaxis in haploidentical transplant. Haematologica. 2017;102(2):401-410. doi:10.3324/haematol.2016.151779
    1. Luo Y, Xiao H, Lai X, et al. . T-cell–replete haploidentical HSCT with low-dose anti–T-lymphocyte globulin compared with matched sibling HSCT and unrelated HSCT. Blood. 2014;124(17):2735-2743. doi:10.1182/blood-2014-04-571570
    1. Rovatti PE, Gambacorta V, Lorentino F, Ciceri F, Vago L. Mechanisms of leukemia immune evasion and their role in relapse after haploidentical hematopoietic cell transplantation. Front Immunol. 2020;11:147. doi:10.3389/fimmu.2020.00147
    1. Muniz P, Kwon M, Carbonell D, et al. . Clinical utility of the detection of the loss of the mismatched HLA in relapsed hematological patients after haploidentical stem cell transplantation with high-dose cyclophosphamide. Front Immunol. 2021;12:642087. doi:10.3389/fimmu.2021.642087
    1. Schneider-Hohendorf T, Gorlich D, Savola P, et al. . Sex bias in MHC I-associated shaping of the adaptive immune system. Proc Natl Acad Sci U S A. 2018;115(9):2168-2173. doi:10.1073/pnas.1716146115
    1. Yachi PP, Ampudia J, Gascoigne NRJ, Zal T. Nonstimulatory peptides contribute to antigen-induced CD8-T cell receptor interaction at the immunological synapse. Nat Immunol. 2005;6(8):785-792. doi:10.1038/ni1220
    1. Bachireddy P, Ennis C, Nguyen VN, et al. . Distinct evolutionary paths in chronic lymphocytic leukemia during resistance to the graft-versus-leukemia effect. Sci Transl Med. 2020;12(561):eabb7661. doi:10.1126/scitranslmed.abb7661
    1. Lindsey AM. Cancer cachexia: effects of the disease and its treatment. Semin Oncol Nurs. 1986;2(1):19-29. doi:10.1016/0749-2081(86)90005-7
    1. Murry DJ, Riva L, Poplack DG. Impact of nutrition on pharmacokinetics of anti-neoplastic agents. Int J Cancer Suppl. 1998;11:48-51. doi:10.1002/(sici)1097-0215(1998)78:11+<48::aid-ijc14>;2-3
    1. Boullata JI. Drug disposition in obesity and protein-energy malnutrition. Proc Nutr Soc. 2010;69(4):543-550. doi:10.1017/S0029665110001990
    1. Greiner J, Gotz M, Bunjes D, Hofmann S, Wais V. Immunological and clinical impact of manipulated and unmanipulated DLI after allogeneic stem cell transplantation of AML patients. J Clin Med. 2019;9(1):39. doi:10.3390/jcm9010039

Source: PubMed

3
S'abonner