Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59.268
Filter
1.
Front Immunol ; 15: 1454614, 2024.
Article in English | MEDLINE | ID: mdl-39355240

ABSTRACT

The incidence of Acute myeloid leukemia (AML) increases with advancing age, and the prognosis for elderly patients is significantly poorer compared to younger patients. Although the combination therapy of venetoclax and hypomethylating agents has demonstrated improved prognosis in patients unable to tolerate intensive chemotherapy, there remains a therapeutic blank for those who fail to achieve remission with current treatment regimens. Here, we report the successful clinical utilization of autogenous CLL1 CAR-T therapy combined with hematopoietic stem cell transplantation in a 73-year-old patient diagnosed with refractory AML. The patient achieved morphological complete remission (CR) with incomplete marrow recovery and a slight presence of minimal residual disease (MRD) after receiving CLL1 CAR-T therapy. To further enhance the treatment and promote the recovery of hemopoiesis, we performed bridged allogenic hematopoietic stem cell transplantation (allo-HSCT) 20 days after the infusion of CLL1 CAR-T cells. The patient achieved MRD-negative CR following HSCT treatment. His primary disease maintained a complete remission status during the 11-month follow-up period. The patient encountered grade 2 cytokine release syndrome and grade 4 granulocytopenia subsequent to the infusion of CAR-T cells, while several rounds of infection and graft-versus-host disease were observed following allo-HSCT. Nevertheless, all these concerns were successfully addressed through comprehensive provision of supportive treatments. We have successfully demonstrated a highly effective and safe combination strategy involving CLL1 CAR-T therapy and allo-HSCT, which has exhibited remarkable tolerability and holds great promise even for elderly patients with AML.


Subject(s)
Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Leukemia, Myeloid, Acute , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Aged , Male , Leukemia, Myeloid, Acute/therapy , Immunotherapy, Adoptive/methods , Combined Modality Therapy , Treatment Outcome , Remission Induction
2.
Yakugaku Zasshi ; 144(10): 957-962, 2024.
Article in Japanese | MEDLINE | ID: mdl-39358252

ABSTRACT

This survey aimed to reveal the actual preventing exposure for handling of clothing and sweat of patients treated with anticancer drugs, following the publication of "Guideline for Preventing Occupational Exposure in Cancer Chemotherapy Drugs, 2019 Edition" (Guideline 2019). A survey was conducted among nurses working at 95 hematopoietic stem cell transplantation promotion base hospitals from September 1, 2023 to October 31, 2023. The response rate was 84.2% (80 facilities). Of the respondents, 45% wore gloves when touching patients' skin to administer anticancer drugs. Almost the nurses identified "urine" and "feces" as fluids on contaminated linen, while 14.1% also identified "sweat." For new staff, the results for preventing exposure education on "if touching the patients' skin" and "if handling clothing and linen" were 23.8% and 34.9%, respectively. This survey shows that nurses may not be following the Guideline 2019 for use of personal protective equipment and handling of clothes. Medical institutions handling anticancer drugs need to educate their staff for preventing occupational exposure.


Subject(s)
Antineoplastic Agents , Hematopoietic Stem Cell Transplantation , Occupational Exposure , Sweat , Humans , Antineoplastic Agents/adverse effects , Occupational Exposure/prevention & control , Surveys and Questionnaires , Sweat/chemistry , Personal Protective Equipment , Gloves, Protective , Guideline Adherence , Clothing , Practice Guidelines as Topic
3.
Rinsho Ketsueki ; 65(9): 1148-1154, 2024.
Article in Japanese | MEDLINE | ID: mdl-39358272

ABSTRACT

Treatment for chronic graft-versus-host disease, the most important long-term complication of allogeneic hematopoietic cell transplantation, has changed significantly over the past decade. Development of novel targeted therapies has advanced as objective criteria for the diagnosis and evaluation of chronic GVHD have been established and understanding of the biological pathways to pathogenesis has increased. This paradigm shift is driving chronic GVHD practice significantly toward individualized therapy while minimizing exposure to steroids. Treatment using a variety of novel agents, tailored to each individual patient's condition, is expected to improve quality of life and overall survival by preventing chronic GVHD, controlling disease progression, and maintaining motor and occupational functions. This article reviews the pathogenesis of chronic GVHD and discusses prospects for the treatment of chronic GVHD, along with recently approved drugs and promising drugs in development.


Subject(s)
Graft vs Host Disease , Graft vs Host Disease/therapy , Graft vs Host Disease/diagnosis , Graft vs Host Disease/etiology , Humans , Chronic Disease , Hematopoietic Stem Cell Transplantation/adverse effects
4.
Rinsho Ketsueki ; 65(9): 1140-1147, 2024.
Article in Japanese | MEDLINE | ID: mdl-39358271

ABSTRACT

The intestinal microbiota is an important prognostic factor for allogeneic hematopoietic stem cell transplantation (allo-HCT), and decreased diversity of the intestinal microbiota is linked to shorter overall survival, high transplant-related mortality, and acute graft-versus-host disease (GVHD). Major factors that alter the intestinal microbiota during allo-HCT are broad-spectrum antibiotics and intestinal GVHD. Broad-spectrum antibiotics dysregulate the immune system and impair intestinal epithelial regeneration by reducing beneficial commensal bacteria and activating mucus-degrading bacteria, which disrupts the colonic barrier function. Intestinal GVHD leads to decreased secretion of antimicrobial peptides into the intestinal lumen, as well as mitochondrial dysfunction in the intestinal epithelium, altering the intestinal microbiota. Various therapeutic approaches targeting the intestinal microbiota have been investigated in clinical trials. Protecting the intestinal microbiota may further enhance the safety and efficacy of allo-HCT by regulating intestinal immune responses, promoting intestinal epithelial regeneration, and facilitating the production of beneficial metabolites derived from commensal bacteria.


Subject(s)
Gastrointestinal Microbiome , Graft vs Host Disease , Graft vs Host Disease/immunology , Humans , Hematopoietic Stem Cell Transplantation , Animals , Transplantation, Homologous
5.
Rinsho Ketsueki ; 65(9): 1174-1178, 2024.
Article in Japanese | MEDLINE | ID: mdl-39358275

ABSTRACT

Hematopoietic stem cell (HSC)-targeted gene therapy is curative for various genetic blood diseases, and its efficacy has been demonstrated in recent clinical trials. HSCs have self-renewal and hematopoietic multipotency; therefore, repairing pathological mutations or defects in HSCs allows for a lifelong cure with a single treatment. Autologous HSC gene therapy has been developed by lentiviral gene addition or gene editing, and is an option for most patients because it does not require a compatible donor. Current HSC gene therapy is based on ex vivo methods, in which patient HSCs are harvested, genetically modified ex vivo, and autologously transplanted into patients. However, the complexity of this process and the high cost of treatment are hindering the spread of gene therapy. Therefore, in vivo HSC gene therapy is being developed to deliver gene therapy tools directly into bone marrow HSCs by administration without ex vivo culture.


Subject(s)
Genetic Therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Humans , Hematopoietic Stem Cells/cytology , Animals
6.
Rinsho Ketsueki ; 65(9): 967-975, 2024.
Article in Japanese | MEDLINE | ID: mdl-39358297

ABSTRACT

Atypical chronic myeloid leukemia (aCML) is a rare disease classified as a myelodysplastic/myeloproliferative neoplasm (MDS/MPN). Recent advances in gene mutational profiling have clarified the characteristics of aCML as a disease entity relative to other MDS/MPNs. Although some studies suggest the efficacy of DNA demethylating agents and tyrosine kinase inhibitors, data about these agents are limited due to the small number of patients. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is only therapeutic option that can provide durable remission for aCML and other MDS/MPNs. Retrospective studies from Europe and Japan revealed the clinical results of allo-HSCT for aCML. This review summarizes the pathogenesis of aCML and the development of allo-HSCT and other therapeutic options.


Subject(s)
Hematopoietic Stem Cell Transplantation , Transplantation, Homologous , Humans , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/therapy
7.
Rinsho Ketsueki ; 65(9): 928-936, 2024.
Article in Japanese | MEDLINE | ID: mdl-39358292

ABSTRACT

Acute myeloid leukemia (AML) accounts for approximately 25% of pediatric leukemia cases in Japan, with approximately 150 patients being newly diagnosed with AML annually. Pediatric acute myeloid leukemia is classified into three groups: myeloid leukemia associated with Down syndrome (ML-DS), acute promyelocytic leukemia (APL), and de novo AML. Patients with ML-DS have an event-free survival rate over 80%; however, relapsed patients have dismal outcomes, even if they receive hematopoietic cell transplantation. APL is curable with all-trans retinoic acid and arsenic trioxide. In de novo AML, 10% of patients fail to achieve remission, and approximately 30% of patients who successfully achieve remission subsequently experience AML relapse. This review highlights the therapeutic approach for these three diseases with context from past clinical studies in Japan, and shares promising new therapeutic options for relapsed/refractory de novo AML.


Subject(s)
Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/diagnosis , Child , Recurrence , Hematopoietic Stem Cell Transplantation
8.
Virol J ; 21(1): 236, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39350155

ABSTRACT

BACKGROUND: Cytomegalovirus (CMV) reactivation is a serious problem in recipients of allogeneic hematopoietic stem cell transplantation. Long-term latency depends on specific T cell immune reconstitution, which identifies various pathogens by T cell receptors (TCRs). However, the mechanisms underlying the selection of CMV-specific TCRs in recipients after transplantation remain unclear. METHODS: Using high-throughput sequencing and bioinformatics analysis, the T cell immune repertoire of seven CMV reactivated recipients (CRRs) were analyzed and compared to those of seven CMV non-activated recipients (CNRs) at an early stage after transplant. RESULTS: The counts of unique complementarity-determining region 3 (CDR3) were significantly higher in CNRs than in CRRs. The CDR3 clones in the CNRs exhibit higher homogeneity compared to the CRRs. With regard to T cell receptor ß-chain variable region (TRBV) and joint region (TRBJ) genotypes, significant differences were observed in the frequencies of TRBV6, BV23, and BV7-8 between the two groups. In addition to TRBV29-1/BJ1-2, TRBV2/BJ2-2, and TRBV12-4/BJ1-5, 11 V-J combinations had significantly different expression levels between CRRs and CNRs. CONCLUSIONS: The differences in TCR diversity, TRBV segments, and TRBV-BJ combinations observed between CNRs and CRRs might be associated with post-transplant CMV reactivation and could serve as a foundation for further research.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Hematopoietic Stem Cell Transplantation , Receptors, Antigen, T-Cell , Transplantation, Homologous , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Cytomegalovirus/immunology , Cytomegalovirus/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/immunology , Male , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Adult , Female , Middle Aged , Transplantation, Homologous/adverse effects , Complementarity Determining Regions/genetics , Transplant Recipients , High-Throughput Nucleotide Sequencing , Young Adult , Virus Activation , Genotype , T-Lymphocytes/immunology , Adolescent , Computational Biology/methods
9.
Front Immunol ; 15: 1411392, 2024.
Article in English | MEDLINE | ID: mdl-39351218

ABSTRACT

Haploidentical hematopoietic cell transplantation (haplo-HCT) is associated with an increased risk of allograft rejection. Here, we employed a major histocompatibility complex (MHC)-mismatched allogeneic HCT (allo-HCT) murine model to better understand the role of Gal-1 in immune tolerance. Transplanted mice were classified into either rejected or engrafted based on donor chimerism levels. We noted significantly higher frequencies of CD4+ T cells, CD8+ T cells, natural killer cells, IFN-γ and TNF-α producing CD4+ T cells, and IFN-γ producing dendritic cells and macrophages in rejected mice. Conversely, we found significantly increased frequencies of regulatory T cells (Tregs), predominantly Helios+, IL-10-producing CD4+ T cells, type 1 regulatory (Tr1) cells, and the proportion of Tr1+Gal-1+ cells in engrafted mice. Further, Gal-1 specific blockade in Tregs reduced suppression of effector T cells in engrafted mice. Lastly, effector T cells from engrafted mice were more prone to undergo apoptosis. Collectively, we have shown that Gal-1 may favor HSC engraftment in an MHC-mismatched murine model. Our results demonstrate that Gal-1-expressing Tregs, especially at earlier time points post-transplant, are associated with inducing immune tolerance and stable mixed chimerism after HCT.


Subject(s)
Galectin 1 , Hematopoietic Stem Cell Transplantation , T-Lymphocytes, Regulatory , Animals , Mice , Galectin 1/immunology , Galectin 1/metabolism , T-Lymphocytes, Regulatory/immunology , Mice, Inbred C57BL , Graft Rejection/immunology , Transplantation, Homologous , Major Histocompatibility Complex/immunology , Graft Survival/immunology , Mice, Inbred BALB C , Immune Tolerance
12.
Recenti Prog Med ; 115(9): 404-414, 2024 Sep.
Article in Italian | MEDLINE | ID: mdl-39269355

ABSTRACT

INTRODUCTION: The diffuse large B-cell lymphoma (Dlbcl) is the most common non-Hodgkin lymphoma and at highest incidence among the elderly. Despite the improved outcomes of patients treated with the first-line (1L) standard of care until the end of 2022, composed by rituximab and polychemotherapy (R-Chop), during the last 20 years, the rate of relapsed and refractory Dlbcl (rrDlbcl) remains elevated. This study has identified and analyzed patients newly diagnosed with Dlbcl and treated with 1L, from the perspective of the Italian National Health Service (Ssn). METHODS: From the administrative database of Fondazione Ricerca e Salute (ReS) including ~5.5 million inhabitants/year in Italy, adults with a new in-hospital Dlbcl diagnosis (index date) and treated with 1L in 2018, 2019, 2020 and 2021 were identified and characterized in terms of demographics and comorbidities during a period (from 4 to 8 years) preceding index date. From 1 to 4 years following index date (follow-up), overall survival (Kaplan-Meier curves), percentage distribution of patients by line of therapy including dispensation/administration of chemo-immunotherapy, hemopoietic stem cell transplantation (Hsct), and direct healthcare costs charge to the Ssn, were evaluated. RESULTS: Overall, from the ReS database, 206 patients newly diagnosed with Dlbcl and treated with 1L from 2018 to 2021 in Italy (incidence from 0.9 to 1.7 x100,000 adult inhabitants) were identified. They were mainly older (median age 68 [56; 75] years), males (56%) and affected by ≥2 comorbidities (52%), mostly cardiometabolic. During 4 years of follow-up, 56% of cases in 2018 survived. During the first follow-up year: 73%, 80%, 100% and 35% of cases in 2018, 2019, 2020 and 2021, respectively, received a 2L; 42% and 64% of cases in 2018 and 2020, respectively, received a 3L. At least one Hsct was found as a 2L among cases in 2018, 2020 and 2021. On average, each patient newly diagnosed with Dlbcl and treated with 1L from 2018 to 2021 caused a total expenditure directly charged to the Ssn ranging from € 20,000 to € 30,000 during the first follow-up year (chemo-immunotherapy accounted for 40-53%), which reduced with time in favor of other drugs and Hsct. CONCLUSIONS: This analysis confirms the high rate of rrDlbcl and the high economic impact charged to the SSN to support first the chemo-immunotherapy, then the chronic care and the absence of standardized further lines of therapy for patients with rrDlbcl.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Databases, Factual , Lymphoma, Large B-Cell, Diffuse , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/therapy , Lymphoma, Large B-Cell, Diffuse/epidemiology , Italy , Male , Aged , Female , Middle Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/economics , Aged, 80 and over , Adult , Rituximab/administration & dosage , Kaplan-Meier Estimate , Hematopoietic Stem Cell Transplantation , Health Care Costs/statistics & numerical data , Vincristine/administration & dosage , Follow-Up Studies , Cyclophosphamide/administration & dosage , Cyclophosphamide/therapeutic use , Doxorubicin/administration & dosage , Prednisone/administration & dosage , Prednisone/therapeutic use
13.
Front Immunol ; 15: 1426640, 2024.
Article in English | MEDLINE | ID: mdl-39253078

ABSTRACT

Introduction: Pre-HSCT disease control, suboptimal long-term prognosis, and a high recurrence incidence (RI) continue to pose significant challenges for hematopoietic stem cell transplantation (HSCT) in juvenile myelomonocytic leukemia (JMML) patients. Methods: This retrospective cohort study assessed the effectiveness of a decitabine (DAC)-based protocol in JMML patients undergoing HSCT. The pre-HSCT treatment includes initial and bridging treatment. The efficacy of DAC monotherapy versus DAC combined with cytotoxic chemotherapy(C-DAC) as initial treatment was compared, followed by DAC plus FLAG (fludarabine, cytarabine, and GCSF) as bridging treatment. The HSCT regimens were based on DAC, fludarabine, and busulfan. Post-HSCT, low-dose DAC was used as maintenance therapy. The study endpoints focused on pretransplantation simplified clinical response and post-HSCT survival. Results: There were 109 patients, including 45 receiving DAC monotherapy and 64 undergoing C-DAC treatment. 106 patients completed bridging treatment. All patients were administered planned HSCT regimens and post-HSCT treatment. The initial treatment resulted in 88.1% of patients achieving clinical remission without a significant difference between the DAC and C-DAC groups (p=0.769). Clinical remission rates significantly improved following bridging treatment (p=0.019). The 5-year overall survival, leukemia-free survival, and RI were 92.2%, 88.4%, and 8.0%, respectively. A poor clinical response to pre-HSCT treatment emerged as a risk factor for OS (hazard ratio: 9.8, 95% CI: 2.3-41.1, p=0.002). Conclusion: Implementing a DAC-based administration strategy throughout the pre-HSCT period, during HSCT regimens, and in post-HSCT maintenance significantly reduced relapse and improved survival in JMML patients. Both DAC monotherapy and the DAC plus FLAG protocol proved effective as pre-HSCT treatments.


Subject(s)
Decitabine , Hematopoietic Stem Cell Transplantation , Leukemia, Myelomonocytic, Juvenile , Humans , Decitabine/therapeutic use , Decitabine/administration & dosage , Hematopoietic Stem Cell Transplantation/adverse effects , Retrospective Studies , Female , Male , Child, Preschool , Leukemia, Myelomonocytic, Juvenile/therapy , Leukemia, Myelomonocytic, Juvenile/mortality , Treatment Outcome , Infant , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use , Vidarabine/administration & dosage , Cytarabine/therapeutic use , Cytarabine/administration & dosage
14.
J Clin Invest ; 134(17)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39225097

ABSTRACT

The ubiquitously expressed small GTPase Ras-related protein 1B (RAP1B) acts as a molecular switch that regulates cell signaling, cytoskeletal remodeling, and cell trafficking and activates integrins in platelets and lymphocytes. The residue G12 in the P-loop is required for the RAP1B-GTPase conformational switch. Heterozygous germline RAP1B variants have been described in patients with syndromic thrombocytopenia. However, the causality and pathophysiological impact remained unexplored. We report a boy with neonatal thrombocytopenia, combined immunodeficiency, neutropenia, and monocytopenia caused by a heterozygous de novo single nucleotide substitution, c.35G>A (p.G12E) in RAP1B. We demonstrate that G12E and the previously described G12V and G60R were gain-of-function variants that increased RAP1B activation, talin recruitment, and integrin activation, thereby modifying late responses such as platelet activation, T cell proliferation, and migration. We show that in our patient, G12E was a somatic variant whose allele frequency decreased over time in the peripheral immune compartment, but remained stable in bone marrow cells, suggesting a differential effect in distinct cell populations. Allogeneic hematopoietic stem cell transplantation fully restored the patient's hemato-immunological phenotype. Our findings define monoallelic RAP1B gain-of-function variants as a cause for constitutive immunodeficiency and thrombocytopenia. The phenotypic spectrum ranged from isolated hematological manifestations in our patient with somatic mosaicism to complex syndromic features in patients with reported germline RAP1B variants.


Subject(s)
Gain of Function Mutation , Thrombocytopenia , rap GTP-Binding Proteins , Humans , Male , Amino Acid Substitution , Hematopoietic Stem Cell Transplantation , Immunologic Deficiency Syndromes/genetics , Mutation, Missense , rap GTP-Binding Proteins/genetics , rap GTP-Binding Proteins/metabolism , Thrombocytopenia/genetics , Thrombocytopenia/pathology , Infant, Newborn , Infant , Child, Preschool , Child
15.
Zhonghua Yi Xue Za Zhi ; 104(35): 3347-3350, 2024 Sep 10.
Article in Chinese | MEDLINE | ID: mdl-39266500

ABSTRACT

Clinical data of 1 494 patients with hematological diseases who were scheduled to receive allogeneic hematopoietic stem cell transplantation and received the anti-human-leukocyte-antigen (HLA) antibody test for the first time at the First Affiliated Hospital of Soochow University from 2016 to 2018 was collected to analyze the positive rates and distribution characteristics of different types of pre-existing anti-HLA antibodies in patients with different hematological diseases. Among 1 494 patients with hematological diseases, there were 849 males and 645 females, aged [31 (17, 45)] years, and included 577 cases of acute myeloid leukemia (AML), 373 cases of acute lymphocytic leukemia (ALL), 234 cases of aplastic anemia (AA), 175 cases of myelodysplastic syndrome (MDS), and 135 cases of other diseases. The total positive rate of pre-existing anti-HLA antibodies was 25.1% (375/1 494), among which the positive rates of anti-HLA class Ⅰ, anti-HLA class Ⅱ, and anti-HLA class Ⅰ+Ⅱ antibodies were 11.2% (168/1 494), 4.9% (73/1 494), and 9.0% (134/1 494), respectively.The total positive rates of pre-existing anti-HLA antibodies in patients with MDS、AA、AML、ALL and other diseases were 40.6% (71/175), 30.8% (72/234), 26.2% (151/577), 12.3% (46/373), and 25.9% (35/135), respectively, with statistically significant difference (P<0.001). The positive rates of anti-HLA class Ⅰ, anti-HLA class Ⅱ, and anti-HLA class Ⅰ+Ⅱ antibodies in patients with different hematological diseases showed statistically significant differences (all P<0.001). Given the varying positive rates and distribution characteristics of pre-existing anti-HLA antibodies among patients with different hematological diseases, anti-HLA antibody test should be performed before receiving hematopoietic stem cell transplantation.


Subject(s)
Anemia, Aplastic , HLA Antigens , Hematologic Diseases , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Humans , Adult , Male , Female , Middle Aged , Myelodysplastic Syndromes/immunology , Adolescent , HLA Antigens/immunology , Hematologic Diseases/immunology , Young Adult , Anemia, Aplastic/immunology , Leukemia, Myeloid, Acute/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Transplantation, Homologous
16.
J Clin Immunol ; 45(1): 3, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39264459

ABSTRACT

LRBA deficiency is an inborn error of immunity defined by autoimmunity, lymphoproliferation, recurrent infections, cytopenia, and inflammatory bowel disease. Despite recent advances in managing this disease with targeted biologic therapy, haematopoietic stem cell transplant (HSCT) remains the only cure. However, great variability exists between protocols used to transplant patients with LRBA deficiency. We describe a cohort of seven patients with LRBA deficiency who underwent HSCT using a myeloablative, reduced toxicity regime of fludarabine, treosulfan, and thiotepa at two transplantation centres from 2016 to 2019. Data were collected both retrospectively and prospectively, measuring time to engraftment, infectious complications, incidence of graft versus host disease, and post-transplantation chimerism. Six of seven patients survived transplantation, and four of six surviving patients achieving treatment-free survival. We thus recommend that HSCT with fludarabine, treosulfan, and thiotepa-based conditioning be considered in patients with LRBA deficiency.


Subject(s)
Busulfan , Hematopoietic Stem Cell Transplantation , Thiotepa , Transplantation Conditioning , Vidarabine , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use , Humans , Hematopoietic Stem Cell Transplantation/methods , Busulfan/analogs & derivatives , Busulfan/therapeutic use , Transplantation Conditioning/methods , Thiotepa/therapeutic use , Male , Female , Infant , Child, Preschool , Graft vs Host Disease/etiology , Child , Retrospective Studies , Treatment Outcome , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/therapy
19.
Stem Cell Res Ther ; 15(1): 317, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39304924

ABSTRACT

BACKGROUND: Ex vivo haematopoietic stem/progenitor cell (HSPCs) expansion constitutes an important area of research, and has the potential to improve access to umbilical cord blood (UCB) as a source of stem cells for haematopoietic stem cell transplantation (HSCT). The ability to improve stem cell dose and thereby reduce delayed engraftment times, which has plagued the use of UCB as a stem cell source since inception, is a recognised advantage. The extent to which cluster of differentiation (CD)34 sub-populations are affected by expansion with StemRegenin1 (SR1), and whether a particular subtype may account for better engraftment than others, is currently unknown. The purpose of this study was to determine the impact of SR1-induced HSPC expansion on CD34+ immunophenotypic subsets and gene expression profiles. METHODS: UCB-derived CD34+ HSPCs were characterised before (D0) and after expansion (D7) with SR1 using an extensive immunophenotypic panel. In addition, gene expression was assessed and differentially expressed genes were categorised into biological processes. RESULTS: A dose-dependent increase in the number of CD34+ HSPCs was observed with SR1 treatment, and unbiased and extensive HSPC immunophenotyping proved to be a powerful tool in identifying unique sub-populations within the HSPC repertoire. In this regard, we found that SR1 promotes the emergence of HSPC subsets which may aid engraftment post expansion. In addition, we observed that SR1 has a minimal effect on the transcriptome of 7-day expanded CD34+ HSPCs when compared to cells expanded without SR1, with only two genes being downregulated in the former. CONCLUSION: This study revealed that SR1 selects for potentially novel immunophenotypic HSPC subsets post expansion and has a minimal effect on the transcriptome of 7-day expanded HSPCs when compared to vehicle controls. Whether these distinct immunophenotypic sub-populations possess greater engraftment capacity remains to be tested in animal models.


Subject(s)
Antigens, CD34 , Fetal Blood , Hematopoietic Stem Cells , Immunophenotyping , Humans , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Antigens, CD34/metabolism , Fetal Blood/cytology , Fetal Blood/metabolism , Cell Differentiation , Hematopoietic Stem Cell Transplantation/methods , Purines
20.
Zhonghua Xue Ye Xue Za Zhi ; 45(8): 713-726, 2024 Aug 14.
Article in Chinese | MEDLINE | ID: mdl-39307718

ABSTRACT

Chronic graft-versus-host disease (cGVHD) is a common and severe complication following allogeneic hematopoietic stem cell transplantation, which significantly impacts patients' survival and quality of life. In recent years, notable progress has been made in the diagnosis, prevention, and treatment of cGVHD, driven by the emergence of novel therapies such as targeted drugs and the advancement of clinical research. This consensus, based on the latest developments in cGVHD research and growing data from evidence-based medicine, has been revised and updated from the "Chinese consensus on the diagnosis and management of chronic graft-versus-host disease (2021)" to better guide clinical practice.


Subject(s)
Consensus , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Humans , China , Chronic Disease , Evidence-Based Medicine , Graft vs Host Disease/diagnosis , Graft vs Host Disease/etiology , Graft vs Host Disease/therapy , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Quality of Life , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL