1
|
Shanmugasundaram K, Napier S, Dimitrova D, Stokes A, Wilder J, Chai A, Lisco A, Anderson MV, Sereti I, Uzel G, Freeman AF, McKeown C, Sponaugle J, Sabina R, Rechache K, Hyder MA, Kanakry JA, Kanakry CG. Safety but limited efficacy of donor lymphocyte infusion for post-transplantation cyclophosphamide-treated patients. Bone Marrow Transplant 2024; 59:1513-1524. [PMID: 39134710 PMCID: PMC11530367 DOI: 10.1038/s41409-024-02312-4] [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: 01/04/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 11/03/2024]
Abstract
The therapeutic efficacy of donor lymphocyte infusions (DLIs) given after allogeneic hematopoietic cell transplantation (HCT) is limited by risk of graft-versus-host disease (GVHD). Post-transplantation cyclophosphamide (PTCy) effectively prevents severe GVHD, but there are limited data on outcomes of DLIs given to PTCy-treated patients. We reviewed 162 consecutive PTCy-treated patients transplanted between 2015-2022 within the Center for Immuno-Oncology at the National Cancer Institute. Of 38 DLIs given to 21 patients after 22 HCTs, few DLIs were associated with toxicities of acute GVHD (7.8%), cytokine release syndrome (CRS, 7.8%), or chronic GVHD (2.6%), and all occurred in those receiving serotherapy-containing pre-HCT conditioning (50% of HCTs). Seven DLIs resulted in complete response (18.4%), with 5 of these given after HCTs using serotherapy-containing conditioning. Excluding infectious indications, complete response to DLIs given after transplants with versus without serotherapy-containing pre-HCT conditioning were 30% and 4.3%, respectively. Two patients received DLI for infection and experienced complete resolution without GVHD or CRS, although the efficacy cannot be definitively attributable to the DLI. DLIs given to PTCy-treated patients had low toxicity but limited efficacy, although pre-HCT serotherapy may modulate both toxicity and response. Novel strategies are needed to enhance the therapeutic efficacy of post-transplant cellular therapies without aggravating GVHD.
Collapse
Affiliation(s)
- Krithika Shanmugasundaram
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Scott Napier
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dimana Dimitrova
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anita Stokes
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer Wilder
- Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Amy Chai
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrea Lisco
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute for Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Megan V Anderson
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute for Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Irini Sereti
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute for Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Gulbu Uzel
- Laboratory of Clinical Immunology and Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christi McKeown
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer Sponaugle
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ruby Sabina
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kamil Rechache
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mustafa A Hyder
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer A Kanakry
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Christopher G Kanakry
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
2
|
Ionete A, Bardas A, Varady Z, Vasilica M, Szegedi O, Coriu D. Modified Prophylactic Donor Lymphocyte Infusion (DLI) in an Adult T Cell Lymphoma/Leukemia (ATLL) Patient-Modality of Relapse Prevention. Diseases 2024; 12:210. [PMID: 39329879 PMCID: PMC11431229 DOI: 10.3390/diseases12090210] [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: 07/29/2024] [Revised: 08/21/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Adult T-cell Leukemia/Lymphoma (ATLL) is a rare but aggressive malignancy associated with the human T-cell lymphotropic virus type 1 (HTLV-1). ATLL is a challenging malignancy characterized by its aggressive nature and poor prognosis. Despite advancements in treatment, relapse rates remain high. Donor lymphocyte infusion (DLI) is a promising therapeutic option post-hematopoietic stem cell transplantation (HSCT) to prevent relapse. However, the prophylactic use of DLI in ATLL patients remains underexplored. We report the case of a 45-year-old female diagnosed with ATLL. Following induction chemotherapy and successful HSCT, a modified prophylactic DLI regimen was administered, consisting of gradually increasing doses of donor lymphocytes. The patient demonstrated a favorable response with no significant graft-versus-host disease (GVHD) and maintained remission over a 40-month follow-up period, suggesting a potential benefit of this approach. This case highlights the potential efficacy and safety of modified prophylactic DLI in ATLL patients, warranting further investigation. Our findings suggest that modified prophylactic DLI is a viable option for ATLL patients post-HSCT, offering a balance between efficacy and safety. Future research should focus on optimizing DLI protocols and exploring biomarkers for response prediction.
Collapse
Affiliation(s)
- Alexandra Ionete
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (Z.V.); (M.V.); (D.C.)
- Faculty of General Medicine, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania;
| | - Alexandru Bardas
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (Z.V.); (M.V.); (D.C.)
- Faculty of General Medicine, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania;
| | - Zsofia Varady
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (Z.V.); (M.V.); (D.C.)
| | - Madalina Vasilica
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (Z.V.); (M.V.); (D.C.)
| | - Orsolya Szegedi
- Faculty of General Medicine, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania;
| | - Daniel Coriu
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (Z.V.); (M.V.); (D.C.)
- Faculty of General Medicine, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania;
| |
Collapse
|
3
|
Fitch T, Lane A, McDonnell J, Bleesing J, Jordan M, Kumar A, Khandelwal P, Khoury R, Marsh R, Chandra S. Age Impacts Risk of Mixed Chimerism Following Reduced-Intensity Conditioning Hematopoietic Cell Transplantation for Non-Severe Combined Immune Deficiency Inborn Errors of Immunity. Transplant Cell Ther 2024; 30:101.e1-101.e12. [PMID: 37821080 DOI: 10.1016/j.jtct.2023.09.024] [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: 06/27/2023] [Revised: 09/19/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
Alemtuzumab, fludarabine, and melphalan containing-reduced intensity conditioning (RIC) is commonly used in patients undergoing allogeneic hematopoietic cell transplantation (HCT) for definitive treatment of high-risk inborn errors of immunity (IEI). Although survival is favorable, there is an increased risk of mixed chimerism leading to secondary graft failure. This study evaluated factors associated with the risk of developing mixed chimerism, particularly the influence of age in patients undergoing allogeneic HCT for non-severe combined immune deficiency (SCID) IEI who received a uniform RIC regimen that included intermediate schedule alemtuzumab, fludarabine, and melphalan. We hypothesized that age would impact the incidence of mixed chimerism. We retrospectively reviewed records of patients who underwent HCT for non-SCID IEI with a uniform RIC regimen that included intermediate schedule alemtuzumab (1 mg/kg divided over days -14 to -10), fludarabine (150 mg/m2 or 5 mg/kg if weight <10 kg divided over days -9 to -4), and melphalan (140 mg/m2 or 4.7 mg/kg if weight <10 kg on day -3) between 2010 and 2020 at our institution. Mixed chimerism was defined as <95% donor chimerism on 2 or more consecutive occasions in whole blood. Ninety-three patients who underwent RIC-HCT for non-SCID IEI using intermediate schedule alemtuzumab, fludarabine, and melphalan were categorized into 3 groups: age <1 year, age 1 to 5 years, and age >5 years. Forty-nine patients (52.7%) developed mixed chimerism, at a median of 34 days post-HCT (range, 10 to 1396 days). Mixed chimerism developed in 88.9% (n = 16/18) of the age <1 year group, in 57.1% (n = 20/35) of the age 1 to 5 years group, and in 35% (n =14/40) of the age >5 years group. Patients age <5 years were significantly more likely to develop mixed chimerism (χ2 (3, N = 93) = 14.8; P = .001). We observed a significantly increased cumulative incidence of developing mixed chimerism associated with age <1 year (P = .0002). Competing risk regression analysis showed a 3-fold higher risk of developing mixed chimerism for age <1 year (subdistribution hazard ratio (HR), 3.05; 95% confidence interval [CI], 1.11 to 8.38; P = .031,) compared to age >5 years and a significantly decreased risk of mixed chimerism in patients who developed acute GVHD prior to any intervention (OR, .24; 95% CI, .09 to .65; P = .005) There were no significant associations between mixed chimerism and graft source, graft type, CD34+ or CD3+ cell dose, HLA match, or underlying disease (hemophagocytic lymphohistiocytosis [HLH] versus non-HLH). Additionally, the need for secondary intervention was evaluated; 27 patients (29.0%) required 1 or more secondary interventions (donor lymphocyte infusion, CD34 boost, or second HCT). Patients age <1 year with mixed chimerism were significantly more likely than patients age >5 years to require secondary intervention for mixed chimerism (P = .004). Our study demonstrates that age <5 years, especially age <1 year, is associated with an increased risk of developing mixed chimerism in patients undergoing RIC-HCT for non-SCID IEI using intermediate-schedule alemtuzumab, fludarabine, and melphalan. Our data suggest tailoring regimen intensity based on age to reduce the incidence of mixed chimerism. Children age <5 years, particularly those age <1 year, require a higher-intensity regimen. Possible strategies include adding thiotepa or using a busulfan-based reduced toxicity regimen.
Collapse
Affiliation(s)
- Taylor Fitch
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Adam Lane
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - John McDonnell
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jack Bleesing
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Michael Jordan
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ashish Kumar
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Pooja Khandelwal
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ruby Khoury
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Rebecca Marsh
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Sharat Chandra
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
4
|
Bowman S, Stanek J, Bajwa R, Polishchuk V, Abu-Arja R, Rangarajan HG. CD34 Stem Cell Boost in Pediatric Allogeneic Stem Cell Transplant Recipients: A Case Series and Review of Literature. Clin Hematol Int 2023:10.1007/s44228-023-00042-w. [PMID: 37027103 DOI: 10.1007/s44228-023-00042-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 03/10/2023] [Indexed: 04/08/2023] Open
Abstract
Patients with poor graft function (PGF) or declining donor chimerism (DC) post allogeneic hematopoietic cell transplantation (HCT) may benefit from a CD34-selected stem cell boost (SCB). We retrospectively studied outcomes of fourteen pediatric patients (PGF: 12 and declining DC: 2), with a median age of 12.8 (range 0.08-20.6) years at HCT, who received a SCB. Primary and secondary endpoints included resolution of PGF or improvement in DC (≥ 15% increase), overall survival (OS) and transplant-related mortality (TRM), respectively. The median CD34 dose infused was 7.47 × 106/kg (range 3.51 × 106-3.39 × 107/kg). Among patients with PGF who survived ≥ 3 months post-SCB (n = 8), we observed a non-significant decrease in the cumulative median number of red cell transfusions, platelet transfusions, and GCSF but not intravenous immunoglobulin doses in the 3 months before and after SCB. Overall response rate (ORR) was 50%, with 29% complete and 21% partial responses. ORR was better in recipients who received lymphodepletion (LD) pre-SCB versus none (75% versus 40%; p = 0.56). The incidence of acute and chronic graft-versus-host-disease was 7% and 14%, respectively. The 1-year OS was 50% (95% CI 23-72%) and TRM was 29% (95% CI 8-58%). SCB was effective in half of our cohort with possible benefit of LD pre-SCB.
Collapse
Affiliation(s)
- Sara Bowman
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA
| | - Joe Stanek
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA
- Biostatistics Resource at Nationwide Children's Hospital, Columbus, OH, USA
| | - Rajinder Bajwa
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Veronika Polishchuk
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Rolla Abu-Arja
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Hemalatha G Rangarajan
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA.
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Scheiermann J, Künkele A, von Stackelberg A, Eggert A, Lang P, Zirngibl F, Martin L, Schulte JH, von Bernuth H. Case report: HLA-haploidentical HSCT rescued with donor lymphocytes infusions in a patient with X-linked chronic granulomatous disease. Front Immunol 2023; 14:1042650. [PMID: 36875143 PMCID: PMC9978143 DOI: 10.3389/fimmu.2023.1042650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 02/02/2023] [Indexed: 02/18/2023] Open
Abstract
Chronic granulomatous disease is an inborn error of immunity due to disrupted function of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex. This results in impaired respiratory burst of phagocytes and insufficient killing of bacteria and fungi. Patients with chronic granulomatous disease are at increased risk for infections, autoinflammation and autoimmunity. Allogeneic hematopoietic stem cell transplantation (HSCT) is the only widely available curative therapy. While HSCT from human leukocyte antigen (HLA) matched siblings or unrelated donors are standard of care, transplantation from HLA-haploidentical donors or gene therapy are considered alternative options. We describe a 14-month-old male with X-linked chronic granulomatous disease who underwent a paternal HLA-haploidentical HSCT using T-cell receptor (TCR) alpha/beta+/CD19+ depleted peripheral blood stem cells followed by mycophenolate graft versus host disease prophylaxis. Decreasing donor fraction of CD3+ T cells was overcome by repeated infusions of donor lymphocytes from the paternal HLA-haploidentical donor. The patient achieved normalized respiratory burst and full donor chimerism. He remained disease-free off any antibiotic prophylaxis for more than three years after HLA-haploidentical HSCT. In patients with x-linked chronic granulomatous disease without a matched donor paternal HLA-haploidentical HSCT is a treatment option worth to consider. Administration of donor lymphocytes can prevent imminent graft failure.
Collapse
Affiliation(s)
- Julia Scheiermann
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Center, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Annette Künkele
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Center, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium [Deutsches Konsortium für Transnationale Krebsforschung (DKTK)], Berlin, Germany.,German Cancer Research Center [Deutsches Krebsforschungszentrum (DKFZ)], Heidelberg, Germany
| | - Arend von Stackelberg
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Center, Berlin, Germany
| | - Angelika Eggert
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Center, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium [Deutsches Konsortium für Transnationale Krebsforschung (DKTK)], Berlin, Germany.,German Cancer Research Center [Deutsches Krebsforschungszentrum (DKFZ)], Heidelberg, Germany
| | - Peter Lang
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Center, Berlin, Germany.,Department of Pediatric Hematology and Oncology, University Hospital, Tübingen, Germany
| | - Felix Zirngibl
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Center, Berlin, Germany
| | - Luise Martin
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, University Hospital Center, Berlin, Germany
| | - Johannes Hubertus Schulte
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Center, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium [Deutsches Konsortium für Transnationale Krebsforschung (DKTK)], Berlin, Germany.,German Cancer Research Center [Deutsches Krebsforschungszentrum (DKFZ)], Heidelberg, Germany
| | - Horst von Bernuth
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, University Hospital Center, Berlin, Germany.,Department of Immunology, Labor Berlin GmbH, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany
| |
Collapse
|
6
|
Incremental donor lymphocyte infusion to treat mixed chimerism after allogeneic stem cell transplantation in children with non-malignant diseases. Bone Marrow Transplant 2023; 58:109-111. [PMID: 36261706 DOI: 10.1038/s41409-022-01844-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/21/2022] [Accepted: 10/03/2022] [Indexed: 01/07/2023]
|
7
|
Ali T, Behfar M, Mohseni R, Salajegheh P, Kheder M, Abou-Fakher F, Nikfetrat Z, Jafari F, Naji P, Hamidieh AA. Escalated Dose Donor Lymphocyte Infusion Treatment in Patients with Primary Immune Deficiencies After HSCT with Reduced-Intensity Conditioning Regimen. Hematol Oncol Stem Cell Ther 2022; 15:272-278. [PMID: 34242597 DOI: 10.1016/j.hemonc.2021.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 06/06/2021] [Accepted: 06/13/2021] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE/BACKGROUND Mixed chimerism is a major concern after allogenic hematopoietic stem cell transplantation (HSCT) using a reduced-intensity conditioning (RIC) regimen in primary immunodeficiencies (PIDs). A donor lymphocyte infusion (DLI) escalating dose regimen has been developed with the aim of reducing toxicity while preserving efficacy. However, the graft-versus-host disease (GvHD) development remains the most common and adverse effect of DLI and continues to be a limiting factor in its application, especially nonmalignant diseases such as PIDs. We prospectively evaluated PID patients after HSCT using RIC in Childrens Medical Center, who were candidates for an escalating dose of DLI for MC from 2016 to 2018. METHODS With the median follow-up of 16.4 months, 12 patients (nine males and three females) with a median age of 3.72 years received DLI. The median number of DLI was 3.2 (range, 1-5), the maximum and total dose of DLIs administered per patient were 3.6 × 107 (range, 1-5) cells/kg CD3+ and 9.3 × 107 (range, 1-15) cells/kg CD3+ cells, respectively. RESULTS Median donor chimerism at baseline before the DLIs was 41% (range, 11-73%), patients received DLIs at a median of 105 (range, 37-230) days and 52 (range, 3-168) days after the HSCT and onset of the MC, respectively. At the final assessment, six (54.5%) patients improved after DLIs at a median of 47.3 days. CONCLUSION PID patients may benefit from DLI with an escalating dose regimen, but the GvHD development remains a concern during the DLI, and the optimum dose and frequency must be standardized.
Collapse
Affiliation(s)
- Tahani Ali
- Pediatric Cell and Gene Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Pediatric Hematopoietic Stem Cell Transplant Center, Children's Hospital, Damascus University, Damascus, Syria
| | - Maryam Behfar
- Pediatric Cell and Gene Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Pediatric Hematopoietic Stem Cell Transplant Department, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Rashin Mohseni
- Pediatric Cell and Gene Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Pourya Salajegheh
- Department of Pediatric, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maged Kheder
- Pediatric Cell and Gene Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Pediatric Hematopoietic Stem Cell Transplant Center, Children's Hospital, Damascus University, Damascus, Syria
| | - Faihaa Abou-Fakher
- Pediatric Cell and Gene Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Pediatric Hematopoietic Stem Cell Transplant Center, Children's Hospital, Damascus University, Damascus, Syria
| | - Zeynab Nikfetrat
- Pediatric Hematopoietic Stem Cell Transplant Department, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Jafari
- Pediatric Hematopoietic Stem Cell Transplant Department, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Naji
- Pediatric Cell and Gene Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Ngwube A, Franay C, Shah N. Post-transplant CD34+ selected stem cell boost as an intervention for declining mixed chimerism following reduced intensity conditioning allogeneic stem cell transplant in children and young adults with sickle cell disease: A case series. Pediatr Hematol Oncol 2022; 39:475-480. [PMID: 35147476 DOI: 10.1080/08880018.2021.2013369] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
| | - Cara Franay
- Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Niketa Shah
- Yale University and Yale New Haven Children's Hospital, New Haven, CT, USA
| |
Collapse
|
9
|
CD34-selected stem cell boost as therapy for late graft rejection following allogeneic transplantation for sickle cell disease. Bone Marrow Transplant 2022; 57:1592-1594. [PMID: 35798858 DOI: 10.1038/s41409-022-01749-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 11/08/2022]
|
10
|
Guidotti S, Bassi S, Rossi A, Borotti E, Cortellazzi P, Ferrarese D, Ronda E, Scarpa A, Schiro A, Merli F, Prezioso L, Bonifazi F, Narni F, Fabrizi E, Vallisa D. Mixed T cell CHIMERISM in bone marrow at day+30 after allogeneic stem cell transplantation: Correlation with acute GVHD grades III-IV and no impact on relapse and overall survival. Eur J Haematol 2022; 109:83-89. [PMID: 35389543 DOI: 10.1111/ejh.13772] [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: 12/15/2021] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Early chimerism analysis is important to assess engraftment in allogeneic hematopoietic stem cell transplantations. METHODS We retrospectively investigated the impact of T-cell chimerism at day 30 in bone marrow on acute graft-versus-host disease (aGVHD), relapse, and overall survival in 142 adult allo-transplanted patients. RESULTS The majority of patients (89%) received myeloablative conditioning and 90% have undergone T-cell replete donor graft. At day 30, 103 patients showed T-complete chimerism with prevalence in haploidentical transplants, whereas 39 cases had CD3+ mixed chimerism, including 30 patients transplanted with HLA identical donors, and 21 with T-cell donors<90%. T-cell chimerism at day 30 was weakly inversely related to aGVHD grades II-IV (p = .078) with no cases of grades III-IV aGVHD in patients with CD3+ <95%. Mixed T-cell chimerism did not impact on relapse (p = .448) and five of the seven patients who relapsed had T-cell chimerism ≤90%. Older age and active disease at transplant had a statistically significant negative effect on overall survival (p = .01 and p = .0001, respectively), whereas mixed CD3+ chimerism did not. CONCLUSIONS T lymphocyte chimerism analysis at day +30 in bone marrow could identify allo-transplanted patients at major risk of aGVHD grades III-IV (CD3+ donors >95%) mainly post-myeloablative conditioning regimen.
Collapse
Affiliation(s)
- Samantha Guidotti
- Bone Marrow Transplant Laboratory, Azienda USL di Piacenza, Piacenza, Italy
| | - Simona Bassi
- Department of Hematology, Azienda USL di Piacenza, Piacenza, Italy
| | - Angela Rossi
- Bone Marrow Transplant Laboratory, Azienda USL di Piacenza, Piacenza, Italy
| | - Elena Borotti
- Bone Marrow Transplant Laboratory, Azienda USL di Piacenza, Piacenza, Italy
| | - Pilade Cortellazzi
- Bone Marrow Transplant Laboratory, Azienda USL di Piacenza, Piacenza, Italy
| | - Diego Ferrarese
- Bone Marrow Transplant Laboratory, Azienda USL di Piacenza, Piacenza, Italy
| | - Elena Ronda
- Bone Marrow Transplant Laboratory, Azienda USL di Piacenza, Piacenza, Italy
| | - Alice Scarpa
- Bone Marrow Transplant Laboratory, Azienda USL di Piacenza, Piacenza, Italy
| | - Alice Schiro
- Bone Marrow Transplant Laboratory, Azienda USL di Piacenza, Piacenza, Italy
| | - Francesco Merli
- Hematology, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | | | - Francesca Bonifazi
- Department of Hematology, IRCCS Azienda-Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Franco Narni
- Department of Hematology, AOU di Modena, Modena, Italy
| | - Enrico Fabrizi
- Dipartimento di Scienze Economiche e Sociale e Dipartimento di Scienze Statistiche, Università Cattolica del Sacro Cuore, Italy
| | - Daniele Vallisa
- Department of Hematology, Azienda USL di Piacenza, Piacenza, Italy
| |
Collapse
|
11
|
Yabe H. Allogeneic hematopoietic stem cell transplantation for inherited metabolic disorders. Int J Hematol 2022; 116:28-40. [PMID: 35594014 DOI: 10.1007/s12185-022-03383-z] [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: 04/03/2022] [Revised: 05/01/2022] [Accepted: 05/01/2022] [Indexed: 11/26/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) has been used to treat patients with inherited metabolic disorders (IMDs) for more than 40 years. In the first two decades, various IMDs were treated by HSCT with a wide variety of donor sources and conditioning regimens selected at the institutional level. However, HSCT was not always successful due to post-transplant complications such as graft failure. In the third decade, myeloablative conditioning with targeted busulfan-based pharmacokinetic monitoring was established as an optimal conditioning regimen, and unrelated cord blood was recognized as an excellent donor source. During the fourth decade, further improvements were made to transplant procedures, including modification of the conditioning regimen, and the survival rate after HSCT markedly improved. Simultaneously, several long-term observational studies for patients after HSCT clarified its therapeutic effects on growth and development of cognitive function, fine motor skills, and activities of daily living when compared with enzyme replacement therapy. Although immune-mediated cytopenia was newly highlighted as a problematic morbidity after HSCT for IMDs, especially in younger patients who received unrelated cord blood, a recent study with rituximab added to the conditioning raised expectations that this issue can be overcome.
Collapse
Affiliation(s)
- Hiromasa Yabe
- Department of Innovative Medical Science, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan.
| |
Collapse
|
12
|
Day JW, Elfeky R, Nicholson B, Goodman R, Pearce R, Fox TA, Worth A, Booth C, Veys P, Carpenter B, Hough R, Gaspar HB, Titman P, Ridout D, Workman S, Hernandes F, Sandford K, Laurence A, Campbell M, Burns SO, Morris EC. Retrospective, Landmark Analysis of Long-term Adult Morbidity Following Allogeneic HSCT for Inborn Errors of Immunity in Infancy and Childhood. J Clin Immunol 2022; 42:1230-1243. [PMID: 35579633 PMCID: PMC9537214 DOI: 10.1007/s10875-022-01278-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/22/2022] [Indexed: 11/28/2022]
Abstract
Purpose
Allogeneic hematopoietic stem cell transplant (HSCT) remains the treatment of choice for patients with inborn errors of immunity (IEI). There is little published medical outcome data assessing late medical complications following transition to adult care. We sought to document event-free survival (EFS) in transplanted IEI patients reaching adulthood and describe common late-onset medical complications and factors influencing EFS. Methods In this landmark analysis, 83 adults surviving 5 years or more following prior HSCT in childhood for IEI were recruited. The primary endpoint was event-free survival, defined as time post-first HSCT to graft failure, graft rejection, chronic infection, life-threatening or recurrent infections, malignancy, significant autoimmune disease, moderate to severe GVHD or major organ dysfunction. All events occurring less than 5 years post-HSCT were excluded. Results EFS was 51% for the whole cohort at a median of 20 years post HSCT. Multivariable analysis identified age at transplant and whole blood chimerism as independent predictors of long-term EFS. Year of HSCT, donor, conditioning intensity and underlying diagnosis had no significant impact on EFS. 59 events occurring beyond 5 years post-HSCT were documented in 37 patients (45% cohort). A total of 25 patients (30% cohort) experienced ongoing significant complications requiring active medical intervention at last follow-up. Conclusion Although most patients achieved excellent, durable immune reconstitution with infrequent transplant-related complications, very late complications are common and associated with mixed chimerism post-HSCT. Early intervention to correct mixed chimerism may improve long-term outcomes and adult health following HSCT for IEI in childhood. Supplementary Information The online version contains supplementary material available at 10.1007/s10875-022-01278-6.
Collapse
Affiliation(s)
- James W Day
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK.,University College London Hospitals NHS Foundation Trust, London, UK
| | - Reem Elfeky
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK
| | - Bethany Nicholson
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK
| | - Rupert Goodman
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK
| | | | - Thomas A Fox
- University College London Hospitals NHS Foundation Trust, London, UK.,UCL Institute of Immunity & Transplantation, London, UK
| | - Austen Worth
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Claire Booth
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK.,UCL Great Ormond Street Institute of Child Health, London, UK
| | - Paul Veys
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Ben Carpenter
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Rachael Hough
- University College London Hospitals NHS Foundation Trust, London, UK
| | - H Bobby Gaspar
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK.,UCL Great Ormond Street Institute of Child Health, London, UK
| | - Penny Titman
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Deborah Ridout
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Sarita Workman
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK
| | - Fernando Hernandes
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK
| | | | - Arian Laurence
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK.,University College London Hospitals NHS Foundation Trust, London, UK
| | - Mari Campbell
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK.,UCL Institute of Immunity & Transplantation, London, UK
| | - Siobhan O Burns
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK.,UCL Institute of Immunity & Transplantation, London, UK
| | - Emma C Morris
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK. .,University College London Hospitals NHS Foundation Trust, London, UK. .,UCL Institute of Immunity & Transplantation, London, UK.
| |
Collapse
|
13
|
You Y, Wang J, Wang Z. Programmed death 1 monoclonal antibody helped to treat mixed chimeric and reactivation of Epstein-Barr virus in a patient with adult-onset chronic active Epstein-Barr virus infection after allogeneic hematopoietic stem cell transplantation: A case report. Medicine (Baltimore) 2022; 101:e28542. [PMID: 35029211 PMCID: PMC8758036 DOI: 10.1097/md.0000000000028542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 12/21/2021] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Systemic forms of chronic active Epstein-Barr virus infection (CAEBV) can predispose a patient to a protracted course of fulminant hemophagocytic lymphohistiocytosis, which has a poor prognosis. Epstein-Barr virus (EBV) infection may persist even after theoretically curative hematopoietic stem cell transplantation. PATIENT CONCERNS A female patient with CAEBV underwent chemotherapy followed by allogeneic hematopoietic stem cell transplantation from her human leukocyte antigen-matched sister. Neutrophil and platelet engraftment was observed on day +12 and +10. Full donor chimerism (DC) was achieved on Day +21. DIAGNOSES From day +38, EBV-DNA in the blood was persistently positive, and DC declined. We attempted empirical interventions such as withdrawal of immune suppression, multiple donor lymphocyte infusion, stem cell boost, and interferon-α treatment. However, EBV-DNA copies continued to increase aggressively, whereas DC decreased rapidly and then reached a nadir of 63.27%. INTERVENTIONS Salvage programmed death 1 (PD-1) antibody treatment was administered as salvage therapy at +69 and +84. OUTCOMES EBV-DNA was negative on day +97 and was ultimately undetectable. Equivalently, a full and stable DC was obtained at +97. LESSONS We summarize a case of PD-1 antibody used as salvage treatment in a post-transplant patient with CAEBV, which was eradicated and full DC was obtained. This case suggests that the PD-1 antibody appears to be a promising option for fighting EBV and mixed DCs.
Collapse
|
14
|
Outcomes following DLI in patients with haematological malignancies: Donor characteristics matter. Transplant Cell Ther 2022; 28:183.e1-183.e8. [DOI: 10.1016/j.jtct.2022.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/16/2022] [Accepted: 01/21/2022] [Indexed: 11/18/2022]
|
15
|
Delie A, Verlinden A, Beel K, Deeren D, Mazure D, Baron F, Breems D, De Becker A, Graux C, Lewalle P, Maertens J, Poire X, Schoemans H, Selleslag D, Van Obbergh F, Kerre T. Use of chimerism analysis after allogeneic stem cell transplantation: Belgian guidelines and review of the current literature. Acta Clin Belg 2021; 76:500-508. [PMID: 32362204 DOI: 10.1080/17843286.2020.1754635] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative treatment option in both adult and pediatric patients with malignant and non-malignant hematological diseases. Chimerism analysis, which determines the donor or recipient origin of hematopoietic cells in HSCT recipients, is an essential aspect of post-HSCT follow-up.Objectives: To review the current literature and develop Belgian consensus guidelines for the use of chimerism analysis in the standard of care after allogeneic HSCT.Methods: Non-systematic review of the literature in consultancy with the members of the BHS transplantation committee.Results: Clinical application with regards to prediction of graft failure or relapse as well as cell source are reviewed. A consensus guideline on the use of chimerism analysis after HSCT is presented.Conclusion: Monitoring of the dynamics or kinetics of a patient's chimerism status by serial analysis at fixed time points, as well as on suspicion of relapse or graft failure, is needed to monitor engraftment levels, as well as disease control and possible relapse.
Collapse
Affiliation(s)
- Anke Delie
- Department of Hematology, University Hospital, Ghent University, Ghent, Belgium
| | - Anke Verlinden
- Department of Hematology, University Hospital, University of Antwerp, Antwerp, Belgium
| | - Karolien Beel
- Department of Hematology, Ziekenhuis Netwerk, Antwerpen, Belgium
| | - Dries Deeren
- Department of Hematology, AZ Delta, Roeselare, Belgium
| | - Dominiek Mazure
- Department of Hematology, University Hospital, Ghent University, Ghent, Belgium
| | - Frédéric Baron
- Department of Hematology, University Hospital, University of Liège, Liège, Belgium
| | - Dimitri Breems
- Department of Hematology, Ziekenhuis Netwerk, Antwerpen, Belgium
| | - Ann De Becker
- Department of Hematology, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium
| | - Carlos Graux
- Department of Hematology, University Hospital Namur, Université Catholique de Louvain, Belgium
| | - Philippe Lewalle
- Department of Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Johan Maertens
- Department of Hematology, University Hospital, KU Leuven, Leuven, Belgium
| | - Xavier Poire
- Department of Hematology, University Hospital Saint Luc, Univeristé Catholique de Louvain, Brussels, Belgium
| | - Helene Schoemans
- Department of Hematology, University Hospital, KU Leuven, Leuven, Belgium
| | | | | | - Tessa Kerre
- Department of Hematology, University Hospital, Ghent University, Ghent, Belgium
| |
Collapse
|
16
|
Llaurador G, Nicoletti E, Prockop SE, Hsu S, Fuller K, Mauguen A, O'Reilly RJ, Boelens JJ, Boulad F. Donor-Host Lineage-Specific Chimerism Monitoring and Analysis in Pediatric Patients Following Allogeneic Stem Cell Transplantation: Influence of Pretransplantation Variables and Correlation with Post-Transplantation Outcomes. Transplant Cell Ther 2021; 27:780.e1-780.e14. [PMID: 34082161 DOI: 10.1016/j.jtct.2021.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 11/19/2022]
Abstract
The impact of donor-host chimerism in post-hematopoietic stem cell transplantation (HSCT) outcomes is poorly understood. We were interested in studying whether pre-HSCT variables influenced lineage-specific donor-host chimerism and how lineage-specific chimerism impacts post-HSCT outcomes. Our main objective was to study pre-HSCT variables as predictors of lineage-specific donor-host chimerism patterns and to better characterize the relationship between post-HSCT lineage-specific chimerism and adverse outcomes, including graft failure and disease relapse. We conducted a retrospective data analysis of all patients who underwent allogeneic HSCT at the Pediatric Transplantation and Cellular Therapy service at Memorial Sloan Kettering Cancer Center between January 2010 and June 2015 and had at least 2 measurements of split-lineage chimerism. The trend of lineage-specific donor-host chimerism post-HSCT and the impact of age, disease, graft type, and pretransplantation conditioning regimen on chimerism at 3 months and 12 months post-HSCT were studied. The Wilcoxon signed-rank test, Mann-Whitney-Wilcoxon test, and Cox proportional hazard models were used for statistical analyses. A total of 137 patients were included (median age, 11.3 years). Most patients had a hematologic malignancy (n = 95), and fewer had a nonmalignant disorder (n = 27) or primary immune deficiency (n = 15). Myeloablative conditioning regimens (n = 126) followed by T cell-depleted (TCD) peripheral blood stem cell or bone marrow grafts (n = 101) were most commonly used. Mixed chimerism (MC) of total peripheral blood leukocytes (PBLs) did not predict loss of donor chimerism in all lineages and when stable was not associated with graft failure or rejection in this analyses. Split chimerism with complete donor chimerism (CC) of myeloid, B, and natural killer cells, but not T cells, occurred early post-HSCT, but full donor T cell chimerism was achieved at 12 months post-HSCT by most patients. MC within the T cell lineage was the major contributor to PBL MC, with lower median donor T cell chimerism at 3 months than at 12 months (91%) post-HSCT (51% versus 91%; P < .0001). Predictors of MC at 3 and 12 months were (1) age <3 years (P = .01 for PBLs and P = .003 for myeloid lineage); (2) nonmalignant disorder (P = .007 for PBLs); and (3) the use of reduced-intensity conditioning regimens. TCD grafts produced lower donor T cell chimerism at 3 months post-HSCT compared with unmodified grafts (P < .0001), where T cell lineage CC was achieved early post-HSCT. The donor T cell chimerism was similar at 12 months in the 2 types of grafts. Umbilical cord blood grafts had CC in all lineages at all time points post-HSCT. Loss of donor B cell chimerism was associated with increased risk of relapse in hematologic malignancies (hazard ratio, 1.33; P = .05). Age, underlying disease, conditioning regimen, and graft manipulation can impact post-HSCT donor-host chimerism and be predictors for early MC. MC in total PBLs and T cells was not related to graft failure or disease relapse. Whole-blood PBL chimerism analysis is not sufficient to assess the significance of post-HSCT donor-host status; rather, lineage-specific chimerism, particularly for myeloid, T, and B cells, should be analyzed to guide interventions and inform outcomes.
Collapse
Affiliation(s)
- Gabriela Llaurador
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pediatrics, Weill Cornell Medical College, New York, New York
| | | | - Susan E Prockop
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pediatrics, Weill Cornell Medical College, New York, New York
| | - Susan Hsu
- Histocompatibility/Molecular Genetics Laboratory, American Red Cross Penn Jersey Region, Philadelphia, Pennsylvania
| | - Kirsten Fuller
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard J O'Reilly
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jaap J Boelens
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Farid Boulad
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
17
|
Xu ZL, Cheng YF, Zhang YY, Mo XD, Han TT, Wang FR, Yan CH, Sun YQ, Chen YH, Tang FF, Han W, Wang Y, Zhang XH, Liu KY, Huang XJ, Xu LP. The incidence, clinical outcome, and protective factors of mixed chimerism following hematopoietic stem cell transplantation for severe aplastic anemia. Clin Transplant 2021; 35:e14160. [PMID: 33222318 DOI: 10.1111/ctr.14160] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 10/21/2020] [Accepted: 11/10/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The aim of our study was to determine possible predictors and clinical course of mixed chimerism (MC) in aplastic anemia after transplantation. METHODS A total of 207 transplants were obtained from haploidentical donors (HID) using busulfan (Bu), cyclophosphamide (Cy), and anti-thymocyte globulin (ATG) regimens, and 69 transplants from matched related donors (MRD) and 29 transplants from unrelated donors (URD) using Cy/ATG regimens were obtained. RESULTS Incidences of MC were 1.93 ± 0.01%, 20.29 ± 0.01%, and 35.71 ± 0.01% in HID, MRD, and URD transplantation (p < .001). In multivariate analysis, incidence of MC was significantly higher in patients without adding Bu in conditioning (p < .001) and receiving a lower number of CD3 + cells in graft (p = .042). MC was associated with significantly lower II-IV aGvHD (3.70% vs. 27.7%, p = .007), but higher secondary graft rejection rates (14.8% vs. 0.4%, p < .001) and poorer overall survival (72.7 ± 8.9% vs. 89.6 ± 2.0%, p = .011) than those of donor chimerism cohort. CONCLUSIONS Mixed chimerism was an unsettling status even in non-malignancy. Haploidentical transplantation with more intense regimen by adding Bu to Cy and ATG was associated with reduced MC following HSCT for SAA. An intensified regimen should be explored in matched related or unrelated donors.
Collapse
Affiliation(s)
- Zheng-Li Xu
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yi-Fei Cheng
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yuan-Yuan Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Dong Mo
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Ting-Ting Han
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Feng-Rong Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Chen-Hua Yan
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yu-Qian Sun
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yu-Hong Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Fei-Fei Tang
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Wei Han
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Kai-Yan Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, Peking, China.,National Clinical Research Center for Hematologic Disease, Peking, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| |
Collapse
|
18
|
Chen H, Li XY, Zhan LP, Fang JP, Huang K, Li Y, Weng WJ, Xu LH, Xu HG, Zhou DH. Prediction, management, and prognosis of mixed chimerism after hematopoietic stem cell transplantation in transfusion-dependent pediatric thalassemia patients. Pediatr Transplant 2020; 24:e13876. [PMID: 33098346 DOI: 10.1111/petr.13876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 07/18/2020] [Accepted: 09/06/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Early-onset mixed chimerism (MC) with a high proportion of residual host cells is considered a signal of graft rejection in patients undergoing allogenic hematopoietic stem cell transplantation for transfusion-dependent thalassemia. In order to prevent graft rejection and minimize the risk of treatment-related graft-versus-host disease (GVHD), we established a hierarchical management system based on chimerism analysis. METHOD This retrospective study provides a comprehensive review of the characteristics, interventions, and outcomes of the 38 patients who developed MC after transplantation among the 144 pediatric thalassemia patients between July 2007 and January 2019 at our center. RESULTS A sibling donor, a blood type-matched donor, conditioning regimens without fludarabine, and transplants containing <10 × 108 total nucleated cells/kg were identified to be associated with the development of MC. Among the 38 patients developing MC, only four patients rejected the grafts. The response rate to donor lymphocyte infusion (DLI, only for patients receiving sibling donor transplantation) and cytokine immunomodulation without DLI was 70.6% and 42.9%, respectively. Patients that developed GVHD after DLI or cytokine therapy had a more significant increase in donor cell chimerism (16%, range 0%-35%) than those without (8.5%, range -21% to 40%, P = .049). However, even when treatment-related GVHD was included, patients with MC had a lower cumulative incidence of total acute GVHD than patients with complete donor chimerism (29.2% vs 48.0%, P = .030). CONCLUSIONS Interventions based on chimerism analysis were effective in preventing graft rejection and did not increase treatment-related GVHD in thalassemia patients with MC.
Collapse
Affiliation(s)
- Han Chen
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, China.,Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xin-Yu Li
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, China.,Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Ping Zhan
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, China.,Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jian-Pei Fang
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, China.,Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ke Huang
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, China.,Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yang Li
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, China.,Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wen-Jun Weng
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, China.,Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lv-Hong Xu
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, China.,Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong-Gui Xu
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, China.,Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dun-Hua Zhou
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, China.,Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
19
|
Villa A, Capo V, Castiello MC. Innovative Cell-Based Therapies and Conditioning to Cure RAG Deficiency. Front Immunol 2020; 11:607926. [PMID: 33329604 PMCID: PMC7711106 DOI: 10.3389/fimmu.2020.607926] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
Genetic defects in recombination activating genes (RAG) 1 and 2 cause a broad spectrum of severe immune defects ranging from early severe and repeated infections to inflammation and autoimmune manifestations. A correlation between in vitro recombination activity and immune phenotype has been described. Hematopoietic cell transplantation is the treatment of care; however, the availability of next generation sequencing and whole genome sequencing has allowed the identification of novel genetic RAG variants in immunodeficient patients at various ages, raising therapeutic questions. This review addresses the recent advances of novel therapeutic approaches for RAG deficiency. As conventional myeloablative conditioning regimens are associated with acute toxicities and transplanted-related mortality, innovative minimal conditioning regimens based on the use of monoclonal antibodies are now emerging and show promising results. To overcome shortage of compatible donors, gene therapy has been developed in various RAG preclinical models. Overall, the transplantation of autologous gene corrected hematopoietic precursors and the use of non-genotoxic conditioning will open a new era, offering a cure to an increasing number of RAG patients regardless of donor availability and severity of clinical conditions.
Collapse
Affiliation(s)
- Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy.,Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (IRGB-CNR), Milan, Italy
| | - Valentina Capo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy.,Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (IRGB-CNR), Milan, Italy
| | - Maria Carmina Castiello
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy.,Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (IRGB-CNR), Milan, Italy
| |
Collapse
|
20
|
Mountjoy L, Palmer J, Kunze KL, Khera N, Sproat LZ, Leis JF, Noel P, Slack JL, Jain T. Does early chimerism testing predict outcomes after allogeneic hematopoietic stem cell transplantation? Leuk Lymphoma 2020; 62:252-254. [PMID: 33012186 DOI: 10.1080/10428194.2020.1827249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Luke Mountjoy
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Jeanne Palmer
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Katie L Kunze
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Phoenix, AZ, USA
| | - Nandita Khera
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Lisa Z Sproat
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Jose F Leis
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Pierre Noel
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - James L Slack
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Tania Jain
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
21
|
Song Y, Wang J, Wang Y, Wang Z. HLA-mismatched GPBSC infusion therapy in refractory Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis: an observational study from a single center. Stem Cell Res Ther 2020; 11:265. [PMID: 32611452 PMCID: PMC7329501 DOI: 10.1186/s13287-020-01779-4] [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: 02/02/2020] [Revised: 05/24/2020] [Accepted: 06/18/2020] [Indexed: 11/22/2022] Open
Abstract
Background Hemophagocytic lymphohistiocytosis (HLH) is a severe or even fatal inflammatory state. Epstein–Barr virus (EBV) infection-associated HLH (EBV-HLH) is one of the most common secondary HLH and suffers a very poor prognosis. Allo-HSCT is often required for refractory EBV-HLH, but some patients still cannot proceed to the next allo-HSCT due to various factors. This study aimed to observe the efficacy of HLA-mismatched granulocyte colony-stimulating factor (G-CSF)-mobilized peripheral blood stem cells (GPBSCs) infusion for refractory EBV-HLH. Methods A retrospective case-control study of refractory EBV-HLH patients with GPBSC infusion from HLA-mismatched donors after chemotherapy (as GPBSC group) and sole chemotherapy (as control group) was performed. Efficacy was evaluated 2 and 4 weeks and all patients were followed-up until March 1, 2018. Results There were 18 cases who accepted infusion between March 2016 and Sep 2017 and 19 were randomly selected from refractory EBV-HLH patients who underwent salvage therapy during the same period for the control group. In GPBSC group, WBC (p = 0.017), Fbg (p = 0.040), and ferritin (p = 0.039) improved significantly after treatment. The overall response rate was 66.7% (CR 22.2%, PR 44.4%). However, there are no significant differences in changes of WBC, HGB, PLT, TG, Fbg, Ferritin, AST, ALT, and T-bil between two groups. Only the Fbg level was recovered better in the GPBSC infusion group (p = 0.003). In the GPBSC group, EBV-DNA decreased significantly after 2 weeks (p = 0.001) and 4 weeks (p = 0.012) after treatment, and the effect of the decrease was significantly better than that of the chemotherapy alone group in 2 weeks but not 4 weeks (p2w = 0.011, p4w = 0.145). The median survival time in the infusion group was 20.4 weeks [95% CI 10.9, 29.9], and the median survival time in the control group was 10.8 weeks [95% CI 0–24.34]. In the short-term, the infusion group’s survival rate was better (2-month 88.89% vs. 52.63%, p = 0.008; 3-month 83.33% vs. 47.09%, p = 0.012), but there was no difference in OS (p = 0.287). Conclusions Infusing GPBSCs combined with chemotherapy is effective, especially in decreasing EBV-DNA, performs better than chemotherapy alone, and improves short-term survival rate. GPBSC infusion is suggested as a bridging treatment method to allo-HSCT.
Collapse
Affiliation(s)
- Yue Song
- Department of Hematology, Beijing Friendship Hospital, Capital Medical University, YongAn Road 95th Xicheng District, Beijing, 100050, China
| | - Jingshi Wang
- Department of Hematology, Beijing Friendship Hospital, Capital Medical University, YongAn Road 95th Xicheng District, Beijing, 100050, China
| | - Yini Wang
- Department of Hematology, Beijing Friendship Hospital, Capital Medical University, YongAn Road 95th Xicheng District, Beijing, 100050, China
| | - Zhao Wang
- Department of Hematology, Beijing Friendship Hospital, Capital Medical University, YongAn Road 95th Xicheng District, Beijing, 100050, China.
| |
Collapse
|
22
|
Mitchell R. Hematopoietic Stem Cell Transplantation Beyond Severe Combined Immunodeficiency: Seeking a Cure for Primary Immunodeficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2020; 7:776-785. [PMID: 30832892 DOI: 10.1016/j.jaip.2018.12.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/27/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) can provide definitive therapy for patients with primary immunodeficiency disease (PIDD). Modern HSCT techniques and supportive care have significantly improved outcomes for patients with PIDD. This review examines current HSCT practice for PIDD other than severe combined immunodeficiency, and explores indications, risks, and long-term outcomes for this group of challenging diseases.
Collapse
Affiliation(s)
- Richard Mitchell
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, New South Wales, Australia; School of Women and Children's Health, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
23
|
Lejman M, Zawitkowska J, Zaucha-Prażmo A, Cienkusz M, Mroczkowska A, Kowalczyk J, Drabko K. Influence of Mixed Chimerism on Outcome in Children With Anaemia After Haematopoietic Stem Cell Transplantation. In Vivo 2020; 33:2051-2057. [PMID: 31662537 DOI: 10.21873/invivo.11703] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/13/2019] [Accepted: 09/04/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND/AIM In patients with non-malignant diseases, mixed chimerism is not a rare phenomenon. The clinical impacts of chimerism following allogeneic haematopoietic stem cell transplantation (allo-HSCT) in children with congenital anaemia (CA) and severe aplastic anaemia (SAA) were analysed. PATIENTS AND METHODS We studied twenty-seven consecutive children with congenital and acquired anaemia who had undergone allogeneic haematopoietic stem cell transplantations. In the observed group of patients, the median of the follow-up was 6.12 years (2.00-14.8 years). RESULTS Overall survival (OS) did not depend on the type of disease p=0.1. OS did not significantly differ in patients who received more than 5x106/kg stem cells (91%) and those who received less than 5x106/kg (85%) (p=0.61). Two patterns of stable mixed chimerism (SMC) were observed: SMC (95-97% cells of the donor), and SMC with a fluctuation between 50-90% of the cells of the donor. None of the surviving patients received immunosuppression treatments of chronic Graft-versus-Host Disease (cGvHD). CONCLUSION Our results showed that mixed chimerism did not influence the survival of children with congenital and aplastic anaemia following allo-HSCT.
Collapse
Affiliation(s)
- Monika Lejman
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Laboratory of Genetic Diagnostics, Lublin, Poland
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Agnieszka Zaucha-Prażmo
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Magdalena Cienkusz
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Aleksandra Mroczkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Jerzy Kowalczyk
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Katarzyna Drabko
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
24
|
Dholaria B, Savani BN, Labopin M, Luznik L, Ruggeri A, Mielke S, Al Malki MM, Kongtim P, Fuchs E, Huang XJ, Locatelli F, Aversa F, Castagna L, Bacigalupo A, Martelli M, Blaise D, Ben Soussan P, Arnault Y, Handgretinger R, Roy DC, O'Donnell P, Bashey A, Solomon S, Romee R, Lewalle P, Gayoso J, Maschan M, Lazarus HM, Ballen K, Giebel S, Baron F, Ciceri F, Esteve J, Gorin NC, Spyridonidis A, Schmid C, Ciurea SO, Nagler A, Mohty M. Clinical applications of donor lymphocyte infusion from an HLA-haploidentical donor: consensus recommendations from the Acute Leukemia Working Party of the EBMT. Haematologica 2020; 105:47-58. [PMID: 31537691 PMCID: PMC6939532 DOI: 10.3324/haematol.2019.219790] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 09/19/2019] [Indexed: 01/30/2023] Open
Abstract
Donor lymphocyte infusion has been used in the management of relapsed hematologic malignancies after allogeneic hematopoietic cell transplantation. It can eradicate minimal residual disease or be used to rescue a hematologic relapse, being able to induce durable remissions in a subset of patients. With the increased use of haploidentical hematopoietic cell transplantation, there is renewed interest in the use of donor lymphocytes to either treat or prevent disease relapse post transplant. Published retrospective and small prospective studies have shown encouraging results with therapeutic donor lymphocyte infusion in different haploidentical transplantation platforms. In this consensus paper, finalized on behalf of the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation, we summarize the available evidence on the use of donor lymphocyte infusion from haploidentical donor, and provide recommendations on its therapeutic, pre-emptive and prophylactic use in clinical practice.
Collapse
Affiliation(s)
- Bhagirathbhai Dholaria
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bipin N Savani
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Myriam Labopin
- Department of Haematology and EBMT Paris study office / CEREST-TC, Saint Antoine Hospital, Paris, France
| | - Leo Luznik
- Department of Oncology Hematologic Malignancies, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Annalisa Ruggeri
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Stephan Mielke
- Department of Laboratory Medicine, CAST, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Monzr M Al Malki
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Piyanuch Kongtim
- Stem Cell Transplant and Cellular Therapy, Thammasat University, Pathumthani, Thailand
| | - Ephraim Fuchs
- Johns Hopkins University, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing China
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Sapienza, University of Rome, Italy
| | - Franco Aversa
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Andrea Bacigalupo
- Fondazione Policlinico Universitario Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Didier Blaise
- Department of Hematology, Institut Paoli Calmettes, Marseille France
| | - Patrick Ben Soussan
- Department of Clinical Psychology, Paoli-Calmettes Institute, Marseille, France
| | - Yolande Arnault
- Institut Paoli-Calmette, département de psychologie clinique, Marseille, France
| | - Rupert Handgretinger
- Department of Hematology and Oncology, University Children's Hospital Tübingen, Tübingen Germany
| | - Denis-Claude Roy
- Division of Hematology and Medical Oncology, Hospital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Paul O'Donnell
- Hematology-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Asad Bashey
- Blood and Marrow Transplant Program at Northside Hospital, Atlanta, GA, USA
| | - Scott Solomon
- Blood and Marrow Transplant Program at Northside Hospital, Atlanta, GA, USA
| | - Rizwan Romee
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Philippe Lewalle
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Jorge Gayoso
- HGU Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Michael Maschan
- Oncology and immunology, Dmitriy Rogachev National Medical Center of pediatric hematology, Moscow, Russia
| | - Hillard M Lazarus
- Adult Hematologic Malignancies & Stem Cell Transplant Section, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | - Karen Ballen
- Division of hematology/oncology, University of Virginia Health System, Charlottesville, VA, USA
| | - Sebastian Giebel
- Dept. of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Frederic Baron
- Laboratory of Hematology, University of Liège, Liège, Belgium
| | - Fabio Ciceri
- Hematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milano Italy
| | - Jordi Esteve
- Hematology department, Hospital Clínic de Barcelona, Barcelona Spain
| | - Norbert-Claude Gorin
- Service d'hématologie et thérapie cellulaire Centre international greffes APHP-EBMT-INCa Hospital, Saint Antoine Hospital, Paris France
| | - Alexandros Spyridonidis
- Bone Marrow Transplantation Unit and CBMDP Donor Center, University Hospital of Patras, Patras, Greece
| | - Christoph Schmid
- Department of Hematology and Oncology, Klinikum Augsburg, Augsburg, Germany
| | - Stefan O Ciurea
- Stem Cell Transplant and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Arnon Nagler
- Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel and EBMT ALWP office, Saint Antoine Hospital, Paris, France
| | - Mohamad Mohty
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, AP-HP, Sorobonne University, and INSERM UMRs 938, Paris, France
| |
Collapse
|
25
|
Thekkudan SF, Lima M, Metheny L. Prevention of relapse after allogeneic stem cell transplantation in acute myeloid leukemia: Updates and challenges. ACTA ACUST UNITED AC 2019. [DOI: 10.1002/acg2.77] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Shinto F. Thekkudan
- Stem Cell Transplant Program University Hospitals Cleveland Medical Center Case Western Reserve University Cleveland OH USA
- Rajiv Gandhi Cancer Institute and Research Centre New Delhi India
| | - Marcos Lima
- Stem Cell Transplant Program University Hospitals Cleveland Medical Center Case Western Reserve University Cleveland OH USA
| | - Leland Metheny
- Stem Cell Transplant Program University Hospitals Cleveland Medical Center Case Western Reserve University Cleveland OH USA
| |
Collapse
|
26
|
Swaminathan VV, Uppuluri R, Patel S, Sivashankaran M, Ravichandran N, Ramanan KM, Ramakrishnan B, Vaidhyanathan L, Raj R. Safety and efficacy of fresh whole blood donor lymphocyte infusion in children. Bone Marrow Transplant 2019; 54:1892-1897. [DOI: 10.1038/s41409-019-0580-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 04/18/2019] [Accepted: 05/12/2019] [Indexed: 11/09/2022]
|
27
|
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. [PMID: 31131266 PMCID: PMC6509944 DOI: 10.3389/fped.2019.00171] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/12/2019] [Indexed: 11/13/2022] Open
Abstract
Background: Autoimmune cytopenias (AICs) are potentially life-threatening complications following hematopoietic cell transplantation (HCT), yet little is understood about the mechanism by which they develop. We hypothesized that discordant B cell and T cell recovery is associated with AICs in transplant patients, and that this might differ based on transplant indication. Methods: In this case control study of children who underwent HCT at our institution, we evaluated the clinical and transplant characteristics of subjects who developed AICs compared to a control group matched by transplant indication and donor type. In cases, we analyzed the state of immune reconstitution, including B cell recovery, T cell recovery, and chimerism, immediately prior to AIC onset. Subjects were stratified by primary indication for transplant as malignancy (n = 7), primary immune deficiency (PID, n = 9) or other non-malignant disease (n = 4). We then described the treatment and outcomes for 20 subjects who developed AICs. Results: In our cohort, cases were older than controls, were more likely to receive a myeloablative conditioning regimen and had a significantly lower prevalence of chronic GVHD. There were distinct differences in the state of immune recovery based on transplant indication. None of the patients (0/7) transplanted for primary malignancy had T cell recovery at AIC onset compared to 71% (5/7) of patients with PID and 33% (1/3) of patients with non-malignant disease. The subset of patients with PID and non-malignant disease who achieved T cell reconstitution (6/6) prior to AIC onset, all demonstrated mixed or split chimerism. Subjects with AIHA or multi-lineage cytopenias had particularly refractory courses with poor treatment response to IVIG, steroids, and rituximab. Conclusions: These results highlight the heterogeneity of AICs in this population and suggest that multiple mechanisms may contribute to the development of post-transplant AICs. Patients with full donor chimerism may have early B cell recovery without proper T cell regulation, while patients with mixed or split donor chimerism may have residual host B or plasma cells making antibodies against donor blood cells. A prospective, multi-center trial is needed to develop personalized treatment approaches that target the immune dysregulation present and improve outcomes in patients with post-transplant AICs.
Collapse
Affiliation(s)
- Jessica A Neely
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Christopher C Dvorak
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Matthew S Pantell
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Alexis Melton
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - James N Huang
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Kristin Ammon Shimano
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
28
|
Jia C, Wang B, Zhu G, Zhang R, Wang K, Yan Y, Qin M. Haploidentical hematopoietic stem cell transplantation using reduced-intensity conditioning for pediatric patients with familial hemophagocytic lymphohistiocytosis. Pediatr Investig 2018; 2:216-221. [PMID: 32851268 PMCID: PMC7331449 DOI: 10.1002/ped4.12096] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/12/2018] [Indexed: 11/12/2022] Open
Abstract
IMPORTANCE Allogeneic hematopoietic stem cell transplantation (HSCT) is considered to be the only curative treatment for familial hemophagocytic lymphohistiocytosis (FHLH). Treatment of pediatric FHLH with reduced-intensity conditioning (RIC)-based haploidentical donor (HID) HSCT has been rarely reported. OBJECTIVE To investigate outcomes and adverse events in patients with FHLH who received HID-HSCT. METHODS We conducted a retrospective study of five patients, including three with mutations in PRF1 and two with XIAP deficiency. Four of the five donors were heterozygous for these mutations. The conditioning regimen included fludarabine, cyclophosphamide, and antithymocyte globulin, with or without low-dose irradiation. Unmanipulated mobilized bone marrow and peripheral blood stem cells were used as the grafts. RESULTS All five patients were successfully engrafted. Four patients survived, and one patient died. All exhibited complete response (CR) after HSCT. All of the patients who survived exhibited CR to FHLH without severe regimen-related complications at a median of 29.5 months (range: 23-34 months) after HSCT. Four of the five patients had mixed donor chimerism. Three patients had 17% to 87% mixed donor chimerism but remained free of disease. Four patients received donor lymphocyte infusion (DLI), which improved the level of mixed donor chimerism. One patient experienced a decrease in donor chimerism to 1% and relapsed; Four patients developed acute graft-versus-host disease (GvHD) (grade I or II), and one patient developed grade IV GvHD. INTERPRETATION HID-HSCT with RIC can be considered for treatment for patients with FHLH, but the conditions and DLI regimens need to be optimized for long-term use, and more prospective studies should be conducted.
Collapse
Affiliation(s)
- Chenguang Jia
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics, Ministry of Education; Key Laboratory of Major Diseases in Children, Ministry of EducationHematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's HealthBeijingChina
| | - Bin Wang
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics, Ministry of Education; Key Laboratory of Major Diseases in Children, Ministry of EducationHematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's HealthBeijingChina
| | - Guanghua Zhu
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics, Ministry of Education; Key Laboratory of Major Diseases in Children, Ministry of EducationHematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's HealthBeijingChina
| | - Rui Zhang
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics, Ministry of Education; Key Laboratory of Major Diseases in Children, Ministry of EducationHematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's HealthBeijingChina
| | - Kai Wang
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics, Ministry of Education; Key Laboratory of Major Diseases in Children, Ministry of EducationHematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's HealthBeijingChina
| | - Yan Yan
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics, Ministry of Education; Key Laboratory of Major Diseases in Children, Ministry of EducationHematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's HealthBeijingChina
| | - Maoquan Qin
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics, Ministry of Education; Key Laboratory of Major Diseases in Children, Ministry of EducationHematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's HealthBeijingChina
| |
Collapse
|
29
|
Chandra S, Bleesing JJ, Jordan MB, Grimley MS, Khandelwal P, Davies SM, Edwards S, Leemhuis T, Marsh RA. Post-Transplant CD34+ Selected Stem Cell “Boost” for Mixed Chimerism after Reduced-Intensity Conditioning Hematopoietic Stem Cell Transplantation in Children and Young Adults with Primary Immune Deficiencies. Biol Blood Marrow Transplant 2018; 24:1527-1529. [DOI: 10.1016/j.bbmt.2018.03.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/12/2018] [Indexed: 11/17/2022]
|
30
|
Faraci M, Bagnasco F, Leoni M, Giardino S, Terranova P, Subissi L, Di Duca M, Di Martino D, Lanino E. Evaluation of Chimerism Dynamics after Allogeneic Hematopoietic Stem Cell Transplantation in Children with Nonmalignant Diseases. Biol Blood Marrow Transplant 2017; 24:1088-1093. [PMID: 29292059 DOI: 10.1016/j.bbmt.2017.12.801] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/20/2017] [Indexed: 11/16/2022]
Abstract
It is recognized that chimerism following hematopoietic stem cell transplantation (HSCT) is a dynamic process. The aims of this study were to describe the evolution of chimerism in children with nonmalignant diseases who underwent allogeneic HSCT, and to analyze the risk factors influencing chimerism status. A total of 101 HSCTs were performed in 85 patients with nonmalignant diseases. The donor was unrelated in 62.4% of HSCTs. Reduced-intensity conditioning (RIC) regimen was administered in 48.5% of patients. Acute graft-versus-host disease (aGVHD) occurred in 51.7% and chronic GVHD (cGVHD) in 39.7% of patients. Analysis of chimerism was performed through amplification of 9 specific short tandem repeats by polymerase chain reaction at engraftment and 1, 6, and 12 months after HSCT. Upon first evaluation, complete chimerism (CC) was detected in 34.7% and mixed chimerism (MC) in 55.4%, whereas graft failure occurred in 9.9% of patients. Severe aGVHD was associated with CC (P = .031). The last chimerism evaluation showed CC in 72.1%, stable MC in 12.8%, and progressive MC in 3.5%. CC was associated with a higher incidence of aGVHD (P = .016) and cGVHD (P = .022), whereas the RIC regimen was associated with graft failure (P = .026). One- and 3-year overall survival (OS) was 87.4% and 80.5%, respectively, with a lower OS at 3 years in patients with CC compared with those with MC (P = .008). aGVHD and cGVHD represent factors favoring CC, thus close, careful follow-up of chimerism is recommended in patients affected by nonmalignant disease.
Collapse
Affiliation(s)
- Maura Faraci
- Hematopoetic Stem Cell Transplant Unit, Hematology-Oncology, G. Gaslini Institute, Genoa, Italy.
| | | | - Massimiliano Leoni
- Hematopoetic Stem Cell Transplant Unit, Hematology-Oncology, G. Gaslini Institute, Genoa, Italy
| | - Stefano Giardino
- Hematopoetic Stem Cell Transplant Unit, Hematology-Oncology, G. Gaslini Institute, Genoa, Italy
| | - Paola Terranova
- Clinical and Experimental Hematology Unit, G. Gaslini Institute, Genoa, Italy
| | - Lorenzo Subissi
- Department of Epidemiology, Scientific Institute of Public Health (WIV-ISP), Brussels, Belgium
| | - Marco Di Duca
- Laboratory of Pathophysiology of Uremia, G. Gaslini Institute, Genoa, Italy
| | - Daniela Di Martino
- Hematopoetic Stem Cell Transplant Unit, Hematology-Oncology, G. Gaslini Institute, Genoa, Italy
| | - Edoardo Lanino
- Hematopoetic Stem Cell Transplant Unit, Hematology-Oncology, G. Gaslini Institute, Genoa, Italy
| |
Collapse
|
31
|
Kim VHD, Reid B, Atkinson A, Upton J, Grunebaum E, Roifman CM. Long-term immune reconstitution after matched unrelated hematopoietic stem cell transplantation for immunodeficiency. J Allergy Clin Immunol 2017; 141:1154-1157.e3. [PMID: 29128672 DOI: 10.1016/j.jaci.2017.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 08/30/2017] [Accepted: 10/04/2017] [Indexed: 11/20/2022]
Affiliation(s)
- Vy Hong-Diep Kim
- Division of Immunology & Allergy, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Brenda Reid
- Division of Immunology & Allergy, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada; Canadian Centre for Primary Immunodeficiency, Toronto, Ontario, Canada
| | - Adelle Atkinson
- Division of Immunology & Allergy, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Julia Upton
- Division of Immunology & Allergy, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Eyal Grunebaum
- Division of Immunology & Allergy, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Chaim M Roifman
- Division of Immunology & Allergy, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada; Canadian Centre for Primary Immunodeficiency, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
32
|
HLA-mismatched unrelated donor transplantation using TLI-ATG conditioning has a low risk of GVHD and potent antitumor activity. Blood Adv 2017; 1:1347-1357. [PMID: 29296777 DOI: 10.1182/bloodadvances.2017007716] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 06/15/2017] [Indexed: 12/21/2022] Open
Abstract
Many patients lack a fully HLA-matched donor for hematopoietic cell transplantation (HCT), and HLA mismatch is typically associated with inferior outcomes. Total lymphoid irradiation and antithymocyte globulin (TLI-ATG) is a nonmyeloablative conditioning regimen that is protective against graft-versus-host disease (GVHD), and we hypothesized that the protective effect would extend beyond HLA-matched donors. We report outcomes for all consecutively transplanted patients at Stanford University from December 2001 through May 2015 who received TLI-ATG conditioning and HCTs from 8 to 9 out of 10 HLA-mismatched unrelated donors (MMUDs, N = 72) compared with 10 out of 10 HLA-matched unrelated donors (MUDs, N = 193). The median age of the patients was 60 years with a median follow-up of 2 years, and there was a similar distribution of lymphoid and myeloid malignancies in both cohorts. There were no significant differences between MMUD and MUD cohorts in overall survival (46% vs 46% at 5 years, P = .86), disease-free survival (38% vs 28% at 5 years, P = .25), nonrelapse mortality (17% vs 12% at 2 years, P = .34), acute GVHD grades III-IV (6% vs 3% at day +100, P = .61), or chronic GVHD (39% vs 35% at 5 years, P = .49). There was a trend toward less relapse in the MMUD cohort (45% vs 60% at 5 years, hazard ratio: 0.71, P = .094), which was significant for patients with lymphoid malignancies (29% vs 57% at 5 years, hazard ratio: 0.55, P = .044). Achieving full donor chimerism was strongly associated with lower relapse rates. TLI-ATG conditioning may overcome the traditionally poorer outcome associated with HLA-mismatched donors and may be particularly well suited for patients with lymphoid malignancies who lack HLA-matched donors.
Collapse
|
33
|
Umeda K, Adachi S, Tanaka S, Miki M, Okada K, Hashii Y, Inoue M, Cho Y, Koh K, Goto H, Kajiwara R, Hyakuna N, Kato K, Morio T, Yabe H. Comparison of second transplantation and donor lymphocyte infusion for donor mixed chimerism after allogeneic stem cell transplantation for nonmalignant diseases. Pediatr Blood Cancer 2016; 63:2221-2229. [PMID: 27554591 DOI: 10.1002/pbc.26141] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 06/11/2016] [Accepted: 06/14/2016] [Indexed: 11/12/2022]
Abstract
BACKGROUND Donor mixed chimerism (MC) is an increasing problem after hematopoietic stem cell transplantation (HSCT) for nonmalignant diseases. PROCEDURE In this study, a self-administered questionnaire was used to retrospectively compare efficacy and safety in 49 patients undergoing second HSCT (n = 13) or donor lymphocyte infusion (DLI; n = 36) as treatment for MC. RESULTS The response rate to DLI of patients with secondary graft failure (GF) (25.0%) was significantly lower than that of patients without secondary GF (81.3%; P = 0.041). Among patients undergoing DLI, the rates of successful response were significantly higher in patients having at least 30% donor chimerism (94.1%) than in patients having less than 30% donor chimerism (61.1%; P = 0.041). Furthermore, the rates of successful response were significantly higher in patients receiving larger first or maximum doses of DLI. Sixteen (50.0%) of 32 patients without secondary GF attained complete chimerism after DLI. The cumulative incidence of grade II-IV acute graft-versus-host disease and cytopenia was 37.6 and 26.1%, respectively. CONCLUSIONS DLI yields promising response rates in most patients with higher donor chimerism levels, whereas second HSCT is more likely to benefit patients with lower donor chimerism levels.
Collapse
Affiliation(s)
- Katsutsugu Umeda
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Souichi Adachi
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shiro Tanaka
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| | - Mizuka Miki
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan
| | - Keiko Okada
- Department of Pediatric Hematology/Oncology, Osaka City General Hospital, Osaka, Japan
| | - Yoshiko Hashii
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masami Inoue
- Department of Hematology/Oncology, Osaka Medical Center and Research Institute for Maternal and Child Health, Izumi, Japan
| | - Yuko Cho
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Hiroaki Goto
- Division of Hemato-oncology/Regenerative Medicine, Kanagawa Children's Medical Center, Kanagawa, Japan
| | - Ryosuke Kajiwara
- Department of Pediatrics, Yokohama City University School of Medicine, Yokohama, Japan
| | - Nobuyuki Hyakuna
- Center of Bone Marrow Transplantation, Ryukyu University Hospital, Okinawa, Japan
| | - Koji Kato
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiromasa Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| | | |
Collapse
|
34
|
Del Fante C, Scudeller L, Martinasso A, Viarengo G, Perotti C. Comparison of two automated mononuclear cell collection systems in patients undergoing extracorporeal photopheresis: a prospective crossover equivalence study. Transfusion 2016; 56:2078-84. [DOI: 10.1111/trf.13672] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 04/11/2016] [Accepted: 04/18/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Claudia Del Fante
- Immunohaematology and Transfusion Service, Apheresis and Cell Therapy Unit; Fondazione IRCCS Policlinico San Matteo; Pavia Italy
| | - Luigia Scudeller
- Scientific Direction, Clinical Epidemiology and Biostatistics Unit; Fondazione IRCCS Policlinico San Matteo; Pavia Italy
| | - Alberto Martinasso
- Immunohaematology and Transfusion Service, Apheresis and Cell Therapy Unit; Fondazione IRCCS Policlinico San Matteo; Pavia Italy
| | - Gianluca Viarengo
- Immunohaematology and Transfusion Service, Apheresis and Cell Therapy Unit; Fondazione IRCCS Policlinico San Matteo; Pavia Italy
| | - Cesare Perotti
- Immunohaematology and Transfusion Service, Apheresis and Cell Therapy Unit; Fondazione IRCCS Policlinico San Matteo; Pavia Italy
| |
Collapse
|
35
|
Long-Term Stable Mixed Chimerism after Hematopoietic Stem Cell Transplantation in Patients with Non-Malignant Disease, Shall We Be Tolerant? PLoS One 2016; 11:e0154737. [PMID: 27152621 PMCID: PMC4859543 DOI: 10.1371/journal.pone.0154737] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/18/2016] [Indexed: 11/19/2022] Open
Abstract
Long-term stable mixed chimerism is a rare and poorly understood phenomenon post hematopoietic stem cell transplantation. This study aims to shed light on whether the two hematopoietic systems in patients with mixed chimerism remain functional. Additionally, we investigate possible immunologic differences in these individuals compared to patients with only donor derived immune cells. Patients with donor and mixed chimerism, at median 10 (5–16) years post-HSCT for non-malignant diseases, were assessed regarding clinical situation and immune system (phenotypical and functional). No difference in long-term outcome was seen in terms of general wellbeing, central phenotypic immune system features (e.g., differentiation status, CD4/CD8 ratio, B and NK-cell frequency) and antibody responses to immunizations. At a median of 10 years post transplantation, patients with mixed chimerism had significantly higher IgG3 and platelet levels. Additionally, these patients had higher NKT-cell levels (CD94+CD8+ and CD56+CD8+) than patients with donor chimerism. In depth phenotypic analysis of patients with mixed chimerism demonstrated recipient-derived fractions in most immune cell lineages (e.g., T-cell, B-cell and NK-cell subsets). Recipient cells were also capable of responding to mitogenic stimulation with production of several cytokines. In conclusion, long-term mixed chimerism did not negatively affect patient wellbeing and long-term outcome. Moreover, recipient-derived immunity may still be functional in these patients, suggesting an active state of tolerance and immunologic dependence on both hematopoietic systems.
Collapse
|
36
|
Pichler H, Fritsch G, König M, Daxberger H, Glogova E, Pötschger U, Breuer S, Lawitschka A, Güclü ED, Karlhuber S, Holter W, Haas OA, Lion T, Matthes-Martin S. Peripheral blood late mixed chimerism in leucocyte subpopulations following allogeneic stem cell transplantation for childhood malignancies: does it matter? Br J Haematol 2016; 173:905-17. [PMID: 26996395 DOI: 10.1111/bjh.14008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/03/2016] [Indexed: 01/26/2023]
Abstract
The impact of persistent mixed chimerism (MC) after haematopoietic stem cell transplantation (HSCT) remains unclarified. We investigated the incidence of MC in peripheral blood beyond day +50 after HSCT and its impact on rejection, chronic graft-versus-host disease (c-GvHD) and relapse in 161 children receiving allogeneic HSCT for haematological malignancies. The 1-year incidence of late MC was 26%. Spontaneous conversion to complete donor chimerism (CC) occurred in 43% of patients as compared to 62% after donor lymphocyte infusions. No graft rejection occurred. The 1-year incidence of c-GvHD was 20 ± 7% for MC, and 18 ± 4% for CC patients (P = 0·734). The 3-year cumulative incidence of relapse (CIR) according to chimerism status at days +50 and +100 was 22 ± 4% for CC patients vs. 22 ± 8% for MC patients (day +50; P = 0·935) and 21 ± 4% vs. 20 ± 7% (day +100; P = 0·907). Three-year CIRs in patients with persistent MC and patients with CC/limited MC were comparable (8 ± 7% vs. 19 ± 4%; P = 0·960). HSCT for acute leukaemia or myelodysplastic syndrome as secondary malignancies (hazard ratio (HR) 4·7; P = 0·008), for AML (HR 3·0; P = 0·02) and from mismatched donors (HR 3·1; P = 0·03) were independent factors associated with relapse. Our data suggest that late MC neither protects from c-GvHD nor does it reliably predict impending disease relapse.
Collapse
Affiliation(s)
- Herbert Pichler
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Gerhard Fritsch
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Margit König
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Helga Daxberger
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Evgenia Glogova
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Ulrike Pötschger
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Sabine Breuer
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Anita Lawitschka
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Ece D Güclü
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Susanne Karlhuber
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Holter
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Oskar A Haas
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria.,Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Thomas Lion
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Susanne Matthes-Martin
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
37
|
Rettinger E, Huenecke S, Bonig H, Merker M, Jarisch A, Soerensen J, Willasch A, Bug G, Schulz A, Klingebiel T, Bader P. Interleukin-15-activated cytokine-induced killer cells may sustain remission in leukemia patients after allogeneic stem cell transplantation: feasibility, safety and first insights on efficacy. Haematologica 2016; 101:e153-6. [PMID: 26768688 DOI: 10.3324/haematol.2015.138016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Eva Rettinger
- JW Goethe University Frankfurt, Hospital for Children and Adolescents, Department of Stem Cell Transplantation and Immunology, Germany
| | - Sabine Huenecke
- JW Goethe University Frankfurt, Hospital for Children and Adolescents, Department of Stem Cell Transplantation and Immunology, Germany
| | - Halvard Bonig
- JW Goethe University Frankfurt, Institute for Transfusion Medicine and Immunohematology, and German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Department of Cellular Therapeutics / Cell Processing (GMP), Frankfurt/Main, Germany University of Washington, Department of Medicine/Hematology, Seattle, WA, USA
| | - Michael Merker
- JW Goethe University Frankfurt, Hospital for Children and Adolescents, Department of Stem Cell Transplantation and Immunology, Germany
| | - Andrea Jarisch
- JW Goethe University Frankfurt, Hospital for Children and Adolescents, Department of Stem Cell Transplantation and Immunology, Germany
| | - Jan Soerensen
- JW Goethe University Frankfurt, Hospital for Children and Adolescents, Department of Stem Cell Transplantation and Immunology, Germany
| | - Andre Willasch
- JW Goethe University Frankfurt, Hospital for Children and Adolescents, Department of Stem Cell Transplantation and Immunology, Germany
| | - Gesine Bug
- JW Goethe University Frankfurt, Department of Medicine II, Hematology, Oncology, Rheumatology and Infectious Diseases, Germany
| | - Ansgar Schulz
- University Medical Center Ulm, Department of Pediatrics, Germany
| | - Thomas Klingebiel
- JW Goethe University Frankfurt, Hospital for Children and Adolescents, Department of Stem Cell Transplantation and Immunology, Germany
| | - Peter Bader
- JW Goethe University Frankfurt, Hospital for Children and Adolescents, Department of Stem Cell Transplantation and Immunology, Germany
| |
Collapse
|
38
|
Successful outcome with second hematopoietic stem cell transplantation in a patient with IL-10R deficiency. Bone Marrow Transplant 2015; 51:615-6. [PMID: 26691419 DOI: 10.1038/bmt.2015.310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
39
|
Alemtuzumab levels impact acute GVHD, mixed chimerism, and lymphocyte recovery following alemtuzumab, fludarabine, and melphalan RIC HCT. Blood 2015; 127:503-12. [PMID: 26644451 DOI: 10.1182/blood-2015-07-659672] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 11/21/2015] [Indexed: 01/02/2023] Open
Abstract
Reduced intensity conditioning (RIC) allogeneic hematopoietic cell transplantation (HCT) with alemtuzumab, fludarabine, and melphalan is an effective approach for patients with nonmalignant disorders. Mixed chimerism and graft-versus-host-disease (GVHD) remain limitations on success. We hypothesized that higher levels of alemtuzumab at day 0 would result in a low risk of acute GVHD, a higher risk of mixed chimerism, and delayed early lymphocyte recovery and that alemtuzumab level thresholds for increased risks of these outcomes would be definable. We collected data from 105 patients to examine the influence of peritransplant alemtuzumab levels on acute GVHD, mixed chimerism, and lymphocyte recovery. The cumulative incidences of initial grades I-IV, II-IV, and III-IV acute GVHD in patients with alemtuzumab levels ≤0.15 vs ≥0.16 μg/mL were 68% vs 18% (P < .0001), 47% vs 13% (P = .0002), and 32% vs 8%, respectively (P = .005). The cumulative incidence of mixed chimerism in patients with an alemtuzumab level ≤0.15 μg/mL was 21%, vs 42% with levels of 0.16 to 4.35 μg/mL, and 100% with levels >4.35 μg/mL (P = .003). Patients with alemtuzumab levels ≤0.15 or 0.16 to 0.56 μg/mL had higher lymphocyte counts at day +30 and higher T-cell counts at day +100 compared with patients with levels ≥0.57 μg/mL (all P < .05). We conclude that peritransplant alemtuzumab levels impact acute GVHD, mixed chimerism, and lymphocyte recovery following RIC HCT with alemtuzumab, fludarabine, and melphalan. Precision dosing trials are warranted. We recommend a day 0 therapeutic range of 0.2 to 0.4 μg/mL.
Collapse
|
40
|
New strategies of DLI in the management of relapse of hematological malignancies after allogeneic hematopoietic SCT. Bone Marrow Transplant 2015; 51:324-32. [PMID: 26595077 DOI: 10.1038/bmt.2015.288] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/24/2015] [Accepted: 10/19/2015] [Indexed: 12/17/2022]
Abstract
DLI is an effective strategy for patients with recurrent hematological malignancies after allogeneic hematopoietic SCT (allo-HSCT). DLI has been widely applied to boost the graft vs tumor (GVT) or GVL effects. However, given the potentially severe complications associated with conventional DLI and transient GVL effect, new strategies for DLI are emerging. In this review, we have discussed the recent important studies on DLI as a prophylactic or therapeutic modality for relapsed hematological disorders after allo-HSCT. The strategies to separate GVL from GVHD have also been discussed. Leukemia-targeting therapy and lymphodepletion combined with DLI, and prophylactic DLI after allo-HSCT are often employed for patients with high risk of relapse, which has been reviewed as well. In addition, we have also discussed the issues on DLI to be further addressed, such as the doses, timing and frequency of DLI in different clinical settings, leukemic antigen-specific DLI as well as how to augment GVL effect while attenuating GVHD.
Collapse
|
41
|
Worth AJJ, Thrasher AJ. Current and emerging treatment options for Wiskott–Aldrich syndrome. Expert Rev Clin Immunol 2015; 11:1015-32. [DOI: 10.1586/1744666x.2015.1062366] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|