Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58.373
Filter
1.
J Clin Immunol ; 44(6): 139, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38822857

ABSTRACT

We evaluated the impact of early recovery of mucosal-associated invariant T cells (MAIT) and gamma-delta (γδ) T cells, especially Vδ2+ T cells, on the clinical outcomes of 76 patients who underwent allogeneic hematopoietic cell transplantation (allo-HCT). MAIT cells were identified at day 20-30 post-transplant using flow cytometry and defined as CD3+ TCRVα7.2+CD161+. Two subsets of Vδ2+ T cells were analyzed according to the expression of CD26. The cytotoxicity profile of MAIT and Vδ2+ T cells was analyzed according to the intracellular expression of perforin and granzyme B, and intracellular IFN-γ was evaluated after in vitro activation. CD26+Vδ2+ T cells displayed higher intracellular levels of IFN-γ, whereas CD26- Vδ2+ T were found to be more cytotoxic. Moreover, MAIT cell frequency was correlated with the frequency of Vδ2+ T cells with a better correlation observed with Vδ2+CD26+ than with the Vδ2+CD26- T cell subset. By using the composite endpoint graft-versus-host disease (GvHD)-free, relapse-free survival (GRFS) as the primary endpoint, we found that patients with a higher MAIT cell frequency at day 20-30 after allo-HCT had a significantly increased GRFS and a better overall survival (OS) and disease-free survival (DFS). Moreover, patients with a low CD69 expression by MAIT cells had an increased cumulative incidence of grade 2-4 acute GvHD (aGvHD). These results suggest that MAIT cell reconstitution may provide mitigating effects early after allo-HCT depending on their activation markers and functional status. Patients with a high frequency of Vδ2+CD26+ T cells had a significantly higher GRFS, OS and DFS, but there was no impact on cumulative incidence of grade 2-4 aGVHD, non-relapse mortality and relapse. These results revealed that the impact of Vδ2+ T cells on the success of allo-HCT may vary according to the frequency of the CD26+ subset.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Mucosal-Associated Invariant T Cells , Transplantation, Homologous , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Male , Female , Adult , Middle Aged , Graft vs Host Disease/immunology , Graft vs Host Disease/etiology , Mucosal-Associated Invariant T Cells/immunology , Young Adult , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adolescent , Aged , Treatment Outcome , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Dipeptidyl Peptidase 4/metabolism , Cytotoxicity, Immunologic
2.
HLA ; 103(6): e15560, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38839559

ABSTRACT

De novo anti-HLA donor-specific antibodies (DSAs) were rarely reported in stem cell transplantation patients. We present a case of 39-year-old acute myelogenous leukaemia patient who developed de novo DSAs only 16 days after transplantation with the highest mean fluorescence intensity (MFI) of 7406.23, which were associated with poor graft function (PGF). We used plasma exchange (PE) and intravenous immunoglobulin (IVIg) to reduce DSA level. A series of treatment including mesenchymal stem cells and donor cell transfusion were used to help recover graft function. On day 130, the patient achieved a successful engraftment.


Subject(s)
HLA Antigens , Hematopoietic Stem Cell Transplantation , Isoantibodies , Leukemia, Myeloid, Acute , Humans , Hematopoietic Stem Cell Transplantation/methods , Adult , HLA Antigens/immunology , Isoantibodies/immunology , Isoantibodies/blood , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/immunology , Male , Tissue Donors , Transplantation, Haploidentical/methods , Immunoglobulins, Intravenous/therapeutic use , Plasma Exchange/methods , Female , Histocompatibility Testing
3.
Rinsho Ketsueki ; 65(5): 330-334, 2024.
Article in Japanese | MEDLINE | ID: mdl-38825509

ABSTRACT

A 53-year-old woman presented with shortness of breath and hyperleukocytosis and was admitted to our hospital. Shortly after, she went into cardiopulmonary arrest and was resuscitated. Her white blood cell count was 566,000/µl, with 94.5% cup-like blasts positive for MPO staining and FLT3-ITD positive, so she was diagnosed with acute myeloid leukemia (AML) M1. She also had disseminated intravascular coagulation and tumor lysis syndrome. Extracorporeal membrane oxygenation (ECMO) was started to manage bilateral pulmonary thromboembolism that had developed due to deep vein thrombosis, and induction therapy was performed under ECMO. On the third day of illness, the patient developed cerebral hemorrhage. Hematological remission was confirmed on the 39th day of illness. After consolidation therapy with chemotherapy and an FLT3 inhibitor, she underwent allogeneic hematopoietic stem cell transplantation, and remains alive. Case reports suggest strong evidence of mortality benefit from ECMO in patients with hematologic malignancies, particularly when ECMO served as a bridge through chemotherapy. Our patient suffered from cardiopulmonary arrest due to hyperleukocytosis and pulmonary thromboembolism, but was saved by induction of remission under ECMO. Improvements in supportive care should lead to reduction in early deaths during induction therapy.


Subject(s)
Extracorporeal Membrane Oxygenation , Leukemia, Myeloid, Acute , Humans , Female , Middle Aged , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/complications , Remission Induction , Treatment Outcome , Induction Chemotherapy , Hematopoietic Stem Cell Transplantation , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
4.
Rinsho Ketsueki ; 65(5): 391-400, 2024.
Article in Japanese | MEDLINE | ID: mdl-38825519

ABSTRACT

There is growing recognition of post-transplant cyclophosphamide (PTCy) as the new standard prophylaxis for graft-versus-host disease (GVHD) in HLA-matched peripheral blood stem cell transplants with reduced intensity conditioning, based on recent results of randomized phase III trials of PTCy. Allogeneic hematopoietic cell transplantation (HCT) with PTCy is thought to have GVHD-dependent and -independent graft-versus-tumor (GVT) effects. Its GVHD-dependent effects may be attenuated by PTCy-induced alloreactive T cell dysfunction and preferential recovery of regulatory T cells after HCT, but its GVT effects do not appear to be significantly impaired in patients in remission or with indolent disease. As patients not in remission are often also candidates for transplantation in Japan, it will be necessary to use PTCy as a platform to establish a strategy that could also be effective in patients not in remission and to revise the donor selection algorithm.


Subject(s)
Cyclophosphamide , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Transplantation, Homologous , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Graft vs Host Disease/prevention & control , Cyclophosphamide/administration & dosage , Transplantation Conditioning
5.
Rinsho Ketsueki ; 65(5): 412-419, 2024.
Article in Japanese | MEDLINE | ID: mdl-38825521

ABSTRACT

The advancement of information and communication technology (ICT) is bringing significant changes to hematopoietic stem cell transplantation. Digital transformation (DX) simplifies the collection and analysis of medical data, enabling provision of medical services beyond geographical constraints through telemedicine. Convenient access to electronic medical records and vital data from wearable devices could facilitate personalized medicine, for example, by predicting of disease onset. Online consultations are effective in improving the efficiency of posttransplant follow-ups, donor recruitment, and donor screening in rural areas. Moreover, patient-reported outcomes are effective in improving treatment outcomes and patient management. The effective utilization of ICT necessitates the enhancement of information technology (IT) literacy among healthcare professionals and patients, as well as development of IT proficiency among medical personnel. DX in hematopoietic stem cell transplantation contributes to the improvement of treatment outcomes, quality of medical care, and patient convenience while introducing new possibilities for the future of healthcare.


Subject(s)
Hematopoietic Stem Cell Transplantation , Humans , Telemedicine , Electronic Health Records
6.
Rinsho Ketsueki ; 65(5): 401-411, 2024.
Article in Japanese | MEDLINE | ID: mdl-38825520

ABSTRACT

Chronic graft-versus-host disease (cGVHD) negatively impacts long-term survival and quality of life (QOL) after allogeneic hematopoietic cell transplantation. Corticosteroids are the first-line treatment for cGVHD, but approximately 30% to 70% of patients develop steroid-refractory cGVHD (SR-cGVHD), which has an extremely poor prognosis. The pathophysiology of cGVHD is more complicated than acute GVHD, but recent advances using murine models in conjunction with human studies indicate three major phases: 1) acute inflammation, 2) chronic inflammation with loss of immune tolerance, and 3) disrupted target organ homeostasis and fibrosis. Strategies that help prevent cGVHD include optimal donor selection and choice of conditioning regimen as well as pharmacologic and graft manipulation strategies. The key cellular mediators of SR-cGVHD are T cells, B cells, antigen-presenting cells, and fibroblasts. T cells and B cells are now targetable with the inhibitors ibrutinib and ruxolitinib, respectively. Recently, promising results have been obtained by modulating pathologic T cell responses with Rock2 inhibitors and targeting fibrosis with CSF-1R inhibitors. To optimize the use of these medications, a better understanding of the biological and target organ-specific mechanisms of cGVHD is needed. Here we review recent advances in cGVHD pathogenesis and discuss how best to implement recently approved biology-driven treatments for cGVHD.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Transplantation, Homologous , Humans , Graft vs Host Disease/prevention & control , Graft vs Host Disease/etiology , Graft vs Host Disease/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Chronic Disease , Animals , Bronchiolitis Obliterans Syndrome
7.
Zhonghua Nei Ke Za Zhi ; 63(6): 587-592, 2024 Jun 01.
Article in Chinese | MEDLINE | ID: mdl-38825927

ABSTRACT

Objective: To evaluate the effect of autologous hematopoietic stem cell transplantation (ASCT) on the treatment of relapsed/refractory multiple myeloma (RRMM) with chimeric antigen receptor T cell (CAR-T) therapy. Methods: A retrospective cohort study. The clinical data of 168 patients with RRMM who underwent CAR-T therapy at the Department of Hematology, Xuzhou Medical University Hospital from 3 January 2020 to 13 September 2022 were analyzed. Patients were classified into a transplantation group (TG; n=47) and non-transplantation group (NTG; n=121) based on whether or not they had undergone ASCT previously. The objective response rate (ORR), progression-free survival (PFS), overall survival (OS) and the levels of CD3, CD4, CD8, CD19, CD56 and natural killer (NK) cells before CAR-T infusion were analyzed by χ2 test, Kaplan-Meier method and independent sample t-test. Results: Among 168 patients with RRMM, 98 (58.3%) were male. The median age of onset was 57 (range 30-70) years. After CAR-T therapy, the ORR of patients was 89.3% (92/103) in the NTG and 72.9% (27/73) in the TG. The ORR of the NTG was better than that of the TG (χ2=5.71, P=0.017). After 1 year of CAR-T therapy, the ORR of the NTG was 78.1% (75/96), and that of the TG was 59.4% (19/32). The ORR of the NTG was better than that of the TG (χ2=4.32, P=0.038). The median OS and PFS in the NTG were significantly longer than those in the TG (OS, 30 vs. 20 months; PFS, 26 vs. 12 months; both P<0.05). The CD4 level before CAR-T infusion in the TG was significantly lower than that in the NTG (25.65±13.56 vs. 32.64±17.21; t=-2.15, P=0.034), and there were no significant differences in the counts of CD3, CD8, CD19, CD56, and NK cells between the TG and NTG (all P>0.05). Conclusion: Among patients suffering from RRMM who received CAR-T therapy, patients who did not receive ASCT had significantly better outcomes than those who had received ASCT previously, which may have been related to the CD4 level before receiving CAR-T therapy.


Subject(s)
Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Multiple Myeloma , Transplantation, Autologous , Humans , Multiple Myeloma/therapy , Hematopoietic Stem Cell Transplantation/methods , Male , Middle Aged , Retrospective Studies , Female , Immunotherapy, Adoptive/methods , Aged , Adult , Treatment Outcome , Receptors, Chimeric Antigen
8.
Hematology ; 29(1): 2360843, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38828928

ABSTRACT

The outcomes of relapsed Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ALL) resistant to new drugs such as tyrosine kinase inhibitors, inotuzumab ozogamicin (InO) and blinatumomab are dismal. We treated two cases of Ph+ALL resistant to these drugs that achieved long-term survival after treatment with chimeric antigen receptor (CAR)-T cell therapy or a second allogeneic hematopoietic stem cell transplantation (HCT) with a sequential conditioning regimen. Case 1: A 15-year-old boy was diagnosed with Ph+ALL. Despite the second HCT after the treatment of ponatinib and blinatumomab, hematological relapse occurred. InO was ineffective and he was transferred to a CAR-T center. After the CAR-T cell therapy, negative measurable residual disease (MRD) was achieved and maintained for 38 months without maintenance therapy. Case 2: A 21-year-old man was diagnosed with Ph+ALL. Hematological relapse occurred after the first HCT. Despite of the treatment with InO, ponatinib, and blinatumomab, hematological remission was not achieved. The second HCT was performed using a sequential conditioning regimen with clofarabine. Negative MRD was subsequently achieved and maintained for 42 months without maintenance therapy. These strategies are suggestive and helpful to treat Ph+ALL resistant to multiple immunotherapies.


Subject(s)
Hematopoietic Stem Cell Transplantation , Imidazoles , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Pyridazines , Humans , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Pyridazines/therapeutic use , Adolescent , Imidazoles/therapeutic use , Young Adult , Immunotherapy , Recurrence , Transplantation, Homologous
9.
Cell Transplant ; 33: 9636897241257568, 2024.
Article in English | MEDLINE | ID: mdl-38832653

ABSTRACT

Basiliximab is an important treatment for steroid-refractory acute graft-versus-host disease (SR-aGVHD). We performed this retrospective study to evaluate the efficacy and safety of basiliximab treatment in SR-aGVHD patients following matched sibling donor hematopoietic stem cell transplantation (MSD-HSCT) (n = 63). Overall response rate (ORR) was 63.5% and 54% at any time and at day 28 after basiliximab treatment. Grade III-IV aGVHD before basiliximab treatment predicted a poor ORR after basiliximab treatment. The rates of virus, bacteria, and fungi infections were 54%, 23.8%, and 3.1%, respectively. With a median follow-up of 730 (range, 67-3,042) days, the 1-year probability of overall survival and disease-free survival after basiliximab treatment were 58.6% (95% confidence interval [CI] = 47.6%-72.2%) and 55.4% (95% CI = 44.3%-69.2%), respectively. The 3-year cumulative incidence of relapse and non-relapse mortality after basiliximab treatment were 18.9% (95% CI = 8.3%-29.5%) and 33.8% (95% CI = 21.8%-45.7%), respectively. Comorbidities burden before allo-HSCT, severity of aGVHD and liver aGVHD before basiliximab treatment showed negative influences on survival. Thus, basiliximab was safe and effective treatment for SR-aGVHD following MSD-HSCT.


Subject(s)
Antibodies, Monoclonal , Basiliximab , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Recombinant Fusion Proteins , Humans , Graft vs Host Disease/drug therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Basiliximab/therapeutic use , Male , Female , Adult , Middle Aged , Recombinant Fusion Proteins/therapeutic use , Antibodies, Monoclonal/therapeutic use , Retrospective Studies , Adolescent , Siblings , Young Adult , Immunosuppressive Agents/therapeutic use , Steroids/therapeutic use , Acute Disease , Child , Treatment Outcome , Tissue Donors
10.
Article in English | MEDLINE | ID: mdl-38847554

ABSTRACT

The aim of the present study was to determine the associations between the MICB genetic variability and the expression and the risk of development of post-transplant complications after allogeneic hematopoietic stem cell transplantation (HSCT). HSCT recipients and their donors were genotyped for two MICB polymorphisms (rs1065075, rs3828903). Moreover, the expression of a soluble form of MICB was determined in the recipients' serum samples after transplantation using the Luminex assay. Our results revealed a favorable role of the MICB rs1065075 G allele. Recipients with donors carrying this genetic variant were less prone to developing chronic graft-versus-host disease (cGvHD) when compared to recipients without any symptoms of this disease (41.41% vs. 65.38%, p = 0.046). Moreover, the MICB rs1065075 G allele was associated with a lower incidence of cytomegalovirus (CMV) reactivation, both as a donor (p = 0.015) and as a recipient allele (p = 0.039). The MICB rs1065075 G variant was also found to be associated with decreased serum soluble MICB (sMICB) levels, whereas serum sMICB levels were significantly higher in recipients diagnosed with CMV infection (p = 0.0386) and cGvHD (p = 0.0008) compared to recipients without those complications. A protective role of the G allele was also observed for the rs3828903 polymorphism, as it was more frequently detected among donors of recipients without cGvHD (89.90% vs. 69.23%; p = 0.013). MICB genetic variants, as well as serum levels of sMICB, may serve as prognostic factors for the risk of developing cGvHD and CMV infection after allogeneic HSCT.


Subject(s)
Cytomegalovirus Infections , Genetic Predisposition to Disease , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Minor Histocompatibility Antigens , Transplantation, Homologous , Humans , Graft vs Host Disease/genetics , Graft vs Host Disease/etiology , Cytomegalovirus Infections/genetics , Hematopoietic Stem Cell Transplantation/adverse effects , Male , Female , Transplantation, Homologous/adverse effects , Adult , Middle Aged , Chronic Disease , Minor Histocompatibility Antigens/genetics , Histocompatibility Antigens Class I/genetics , Polymorphism, Single Nucleotide , Alleles , Genotype , Young Adult , Cytomegalovirus/physiology , Adolescent , Risk , Risk Factors
11.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 199-205, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836658

ABSTRACT

The present research aimed to conduct a comprehensive critical analysis of existing literature, focusing on the differentiation of myeloid cells from hematopoietic stem cells within the context of immunological tolerance during pregnancy. A comprehensive systematic review was conducted by searching databases including PubMed, Scopus Biomedicine, EBSCOhost, ScienceDirect, Embase, Cochrane Library, and Web of Science. The focus was on the role of myeloid differentiation from hematopoietic stem cells in modulating immune tolerance, particularly during pregnancy and in certain disease states where they act to suppress the immune response. The quality of the evidence gathered was assessed using the GRADE rating system. Our analysis maintains objectivity and independence from the outcomes presented. The current systematic review offers a synthesis of existing research on the transformation of hematopoietic stem cells into fibroblasts across different tissue types. A thorough search of databases such as PubMed, EBSCOhost, Embase, ScienceDirect, Cochrane Library, and Web of Science was performed in conjunction with a specialist in medical information to identify original research on the derivation of fibroblasts following hematopoietic stem cell transplantation. This search yielded a total of 159 studies, of which 10 met the criteria for inclusion in this review. Reflecting on the constraints of this preliminary review, further in-depth and scientific investigations are warranted to comprehensively assess the impact of varied treatments, with a recommendation for clinicians to proceed with increased circumspection. The myeloid differentiation pathway of hematopoietic stem cells is pivotal in modulating the immune environment during pregnancy, supporting the sustenance of a healthy gestational period. Future research in this domain is expected to advance our understanding of the immunological processes occurring at the maternal-fetal boundary.


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells , Immune Tolerance , Female , Humans , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/cytology , Pregnancy , Cell Differentiation/immunology , Myeloid Cells/immunology , Myeloid Cells/cytology , Hematopoietic Stem Cell Transplantation , Fibroblasts/immunology , Fibroblasts/cytology
12.
J Cancer Res Clin Oncol ; 150(6): 301, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38850365

ABSTRACT

PURPOSE: Systemic sclerosis (SSc) is a rare autoimmune disease associated with high morbidity and mortality. SSc treatment is still challenging, and evidence is scarce. In the last decades high-dose chemotherapy and autologous stem cell transplantation (HD-ASCT) has proven to be effective. However, treatment related morbidity and mortality (TRM) are high. We conducted a retrospective, single-center analysis of SSc patients following HD-ASCT focusing on TRM and risk factors. METHODS: 32 patients who underwent HD-ASCT at our hospital between June 2000 and September 2020 were included. Clinical characteristics were evaluated based on chart review before and after HD-ASCT. Analyses focused on overall survival (OS), TRM, and response to HD-ASCT. RESULTS: Median OS was 81 months (range 0-243). Within one year, 20 of 32 (76.9%) patients responded to HD-ASCT. Overall, 6 patients (18.8%) died in the context of HD-ASCT. Patients with subjective response to HD-ASCT (p = 0.024) and those with shorter time to platelet engraftment (p = 0.047) had significantly longer OS. Impaired renal function, age at HD-ASCT ≥ 55, disease duration < 12 months, high Hematopoietic cell transplantation-specific comorbidity index (HCT-CI) and Charlton Comorbidity Index (CCI) scores were associated with higher TRM. Patients receiving conditioning chemotherapy with thiotepa needed longer time for neutrophil (p = 0.035) and platelet engraftment (p = 0.021). CONCLUSION: This study confirms the efficacy of HD-ASCT for patients with SSc in a single center real-world setting. High TRM is still a challenge. However, TRM could be reduced by exclusion of high-risk patients and attention to prognostic parameters and scores as suggested in this study.


Subject(s)
Hematopoietic Stem Cell Transplantation , Transplantation, Autologous , Humans , Hematopoietic Stem Cell Transplantation/methods , Retrospective Studies , Female , Male , Middle Aged , Adult , Prognosis , Aged , Scleroderma, Diffuse/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Treatment Outcome , Young Adult , Combined Modality Therapy
14.
Stem Cell Res Ther ; 15(1): 164, 2024 Jun 09.
Article in English | MEDLINE | ID: mdl-38853275

ABSTRACT

BACKGROUND: Transplantation of CD34+ hematopoietic stem and progenitor cells (HSPC) into immunodeficient mice is an established method to generate humanized mice harbouring a human immune system. Different sources and methods for CD34+ isolation have been employed by various research groups, resulting in customized models that are difficult to compare. A more detailed characterization of CD34+ isolates is needed for a better understanding of engraftable hematopoietic and potentially non-hematopoietic cells. Here we have performed a direct comparison of CD34+ isolated from cord blood (CB-CD34+) or fetal liver (FL-CD34+ and FL-CD34+CD14-) and their engraftment into immunocompromised NOD/Shi-scid Il2rgnull (NOG) mice. METHODS: NOG mice were transplanted with either CB-CD34+, FL-CD34+ or FL-CD34+CD14- to generate CB-NOG, FL-NOG and FL-CD14--NOG, respectively. After 15-20 weeks, the mice were sacrificed and human immune cell reconstitution was assessed in blood and several organs. Liver sections were pathologically assessed upon Haematoxylin and Eosin staining. To assess the capability of allogenic tumor rejection in CB- vs. FL-reconstituted mice, animals were subcutaneously engrafted with an HLA-mismatched melanoma cell line. Tumor growth was assessed by calliper measurements and a Luminex-based assay was used to compare the cytokine/chemokine profiles. RESULTS: We show that CB-CD34+ are a uniform population of HSPC that reconstitute NOG mice more rapidly than FL-CD34+ due to faster B cell development. However, upon long-term engraftment, FL-NOG display increased numbers of neutrophils, dendritic cells and macrophages in multiple tissues. In addition to HSPC, FL-CD34+ isolates contain non-hematopoietic CD14+ endothelial cells that enhance the engraftment of the human immune system in FL-NOG mice. We demonstrate that these CD14+CD34+ cells are capable of reconstituting Factor VIII-producing liver sinusoidal endothelial cells (LSEC) in FL-NOG. However, CD14+CD34+ also contribute to hepatic sinusoidal dilatation and immune cell infiltration, which may culminate in a graft-versus-host disease (GVHD) pathology upon long-term engraftment. Finally, using an HLA-A mismatched CDX melanoma model, we show that FL-NOG, but not CB-NOG, can mount a graft-versus-tumor (GVT) response resulting in tumor rejection. CONCLUSION: Our results highlight important phenotypical and functional differences between CB- and FL-NOG and reveal FL-NOG as a potential model to study hepatic sinusoidal dilatation and mechanisms of GVT.


Subject(s)
Antigens, CD34 , Liver , Animals , Humans , Antigens, CD34/metabolism , Mice , Liver/metabolism , Liver/pathology , Mice, Inbred NOD , Hematopoietic Stem Cell Transplantation , Mice, SCID , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/transplantation , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Fetal Blood/cytology , Melanoma/pathology , Melanoma/immunology
15.
Cell Transplant ; 33: 9636897241259722, 2024.
Article in English | MEDLINE | ID: mdl-38856035

ABSTRACT

Posttransplant lymphoproliferative disorder (PTLD) is a rare lymphoid and/or plasmocytic proliferation that occurs after allogeneic hematopoietic stem cell transplantation (allo-HSCT). We aimed to identify the pathologic features and clinical outcomes of T-cell PTLD, an extremely rare subtype of PTLD, after allo-HSCT. In this study, six allo-HSCT recipients with T-cell PTLD from five transplant centers in China were enrolled. All the T-cell PTLD were donor-derived, and three patients were with monomorphic and three with polymorphic types, respectively. All patients received cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP)-based chemotherapy. Five patients achieved complete response (CR), and one experienced progressive disease (PD). The median time from HSCT to onset was 4 (range: 0.6-72) months, analyzed in combination with the other 16 patients with T-cell PTLD identified from previous reports. About 56.3% of the T-cell samples (9/16) were positive for in situ hybridization with an Epstein-Barr virus (EBV)-encoded small nuclear early region (EBER ISH). CHOP-based chemotherapy might be the optimal strategy for patients who showed no response to empiric therapy with a CR rate of 87.5%. In conclusion, our study observed that T-cell PTLD has distinct clinical manifestations and morphological features, which characterized by less relation to EBV, later occurrence, and poorer prognosis when compared with B-cell PTLD.


Subject(s)
Hematopoietic Stem Cell Transplantation , Lymphoproliferative Disorders , T-Lymphocytes , Transplantation, Homologous , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Lymphoproliferative Disorders/etiology , Male , Female , Adult , T-Lymphocytes/immunology , Transplantation, Homologous/methods , Transplantation, Homologous/adverse effects , Adolescent , Child , Middle Aged , Young Adult , Cyclophosphamide/therapeutic use
16.
Trials ; 25(1): 361, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840199

ABSTRACT

BACKGROUND: Although hematopoietic stem cell transplantation provides the chances of survival for aplastic anemia patients, it is also related to many treatment-related physical and psychological side effects that severely influence the quality of life. Exercise interventions have shown positive results in mixed hematology populations. The study aims to determine the effectiveness of exercise rehabilitation in improving the quality of life, fatigue, and physical function in these patients. METHODS: The study will enroll a total of 82 aplastic anemia patients receiving hematopoietic stem cell transplantation. They will be randomly divided into two groups in a 1:1 ratio. The intervention group will participate in structured exercise rehabilitation (plus usual care), while control group participants will receive usual care. The exercise rehabilitation program will be performed from neutrophil and platelet engraftment until 100 days after transplantation. All outcomes will be measured at the following time points: the neutrophil and platelet engraftment (± 1day, T0), discharge from the transplantation module (± 1 day, T1), hospital discharge (± 1 day, T2), and 100 days post-transplantation (± 5 days, T3). DISCUSSION: This study aims to assess the effectiveness of exercise rehabilitation for aplastic anemia patients receiving hematopoietic stem cell transplantation in a Chinese single center. It is particularly vital to conduct the studies in this population. Moreover, the evidence obtained from the study will provide evidence for future research and clinical practice to exercise in aplastic anemia patients. TRIAL REGISTRATION: ChiCTR2200060762. Registered on May 2022, www.trialregister.nl/trial/7702.


Subject(s)
Anemia, Aplastic , Exercise Therapy , Hematopoietic Stem Cell Transplantation , Quality of Life , Randomized Controlled Trials as Topic , Humans , Anemia, Aplastic/rehabilitation , Anemia, Aplastic/therapy , Exercise Therapy/methods , Treatment Outcome , Adult , Male , Adolescent , Female , Young Adult , Fatigue/etiology , Middle Aged , Time Factors , China , Recovery of Function
17.
Front Immunol ; 15: 1381308, 2024.
Article in English | MEDLINE | ID: mdl-38745670

ABSTRACT

Chimeric antigen receptor T cells (CAR T) targeting CD7 for T-cell acute lymphoblastic leukemia/lymphoma (T-ALL/LBL) showed promising efficacy and safety in some clinical trials. However, most of them were bridged with allogeneic hematopoietic stem cell transplantation (allo-HSCT). We described successful treatment with preventive donor-derived anti-CD7 CAR-T therapy in a case of refractory T lymphoblastic lymphoma following allo-HSCT, who could not receive autologous anti-CD7 CAR-T products due to the low-quality of T lymphocytes. To date, the patient's complete remission has persisted for 20 months after HSCT.


Subject(s)
Antigens, CD7 , Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Transplantation, Homologous , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/adverse effects , Antigens, CD7/immunology , Receptors, Chimeric Antigen/immunology , Male , Tissue Donors , T-Lymphocytes/immunology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Treatment Outcome , Adult
18.
BMC Pediatr ; 24(1): 333, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745164

ABSTRACT

BACKGROUND: The risk factors for hemorrhagic cystitis (HC) in children undergoing hematopoietic stem cell transplantation (HSCT) are unclear. Therefore, we conducted this systematic review and meta-analysis to investigate the risk factors for HC in children undergoing HSCT. METHODS: We performed this meta-analysis by retrieving studies from PubMed, EMBASE, and the Cochrane Library up to October 10, 2023, and analyzing those that met the inclusion criteria. I2 statistics were used to evaluate heterogeneity. RESULTS: Twelve studies, including 2,764 patients, were analyzed. Male sex (odds ratio [OR] = 1.52; 95% confidence interval [CI], 1.16-2.00; p = 0.003, I2 = 0%), allogeneic donor (OR = 5.28; 95% CI, 2.60-10.74; p < 0.00001, I2 = 0%), human leukocyte antigen (HLA) mismatched donor (OR = 1.86; 95% CI, 1.00-3.44; p = 0.05, I2 = 31%), unrelated donor (OR = 1.58; 95% CI, 1.10-2.28; p = 0.01, I2 = 1%), myeloablative conditioning (MAC) (OR = 3.17; 95% CI, 1.26-7.97; p = 0.01, I2 = 0%), busulfan (OR = 2.18; 95% CI, 1.33-3.58; p = 0.002, I2 = 0%) or anti-thymoglobulin (OR = 1.65; 95% CI, 1.07-2.54; p = 0.02, I2 = 16%) use, and cytomegalovirus (CMV) reactivation (OR = 2.64; 95% CI, 1.44-4.82; p = 0.002, I2 = 0%) were risk factors for HC in children undergoing HSCT. CONCLUSIONS: Male sex, allogeneic donor, HLA-mismatched, unrelated donor, MAC, use of busulfan or anti-thymoglobulin, and CMV reactivation are risk factors for HC in children undergoing HSCT.


Subject(s)
Cystitis , Hematopoietic Stem Cell Transplantation , Hemorrhage , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Cystitis/etiology , Risk Factors , Child , Hemorrhage/etiology , Transplantation Conditioning/adverse effects , Sex Factors , Male , Female , Cystitis, Hemorrhagic
19.
Exp Clin Transplant ; 22(3): 229-238, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38695592

ABSTRACT

OBJECTIVES: The eradication of leukemia cells while sparing hematopoietic stem cells in the graft before autologous hematopoietic stem cell transplant is critical to prevention of leukemia relapse. Proliferating cells have been shown to be more prone to apoptosis than differentiated cells in response to ultraviolet radiation; however, whether leukemia cells are more sensitive to ultraviolet LED radiation than hematopoietic stem cells remains unclear. MATERIALS AND METHODS: We compared the in vitro responses between murine leukemia L1210 cells and murine hematopoietic stem cells to 280-nm ultraviolet LED radiation. We also investigated the effects of ultraviolet LED radiation on the tumorigenic and metastatic capacity of L1210 cells and hematopoietic stem cell hematopoiesis in a mouse model of hematopoietic stem cell transplant. RESULTS: L1210 cells were more sensitive to ultraviolet LED radiation than hematopoietic stem cells in vitro, as evidenced by significantly reduced colony formation rates and cell proliferation rates, along with remarkably increased apoptosis rates in L1210 cells. Compared with corresponding unirradiated cells, ultraviolet LED-irradiated L1210 cells failed to generate palpable tumors in mice, whereas ultraviolet LED-irradiated bone marrow cells restored hematopoiesis in vivo. Furthermore, transplant with an irradiated mixture of L1210 cells and bone marrow cells showed later onset of leukemia, milder leukemic infiltration, and prolonged survival in mice, compared with unirradiated cell transplant. CONCLUSIONS: Our results suggest that ultraviolet LED radiation can suppress the proliferative and tumorigenic abilities of leukemia cells without reducing the hematopoietic reconstitution capacity of hematopoietic stem cells, serving as a promising approach to kill leukemia cells in autograft before autologous hematopoietic stem cell transplant.


Subject(s)
Apoptosis , Cell Proliferation , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Animals , Hematopoietic Stem Cells/radiation effects , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/metabolism , Apoptosis/radiation effects , Hematopoiesis/radiation effects , Cell Proliferation/radiation effects , Cell Line, Tumor , Ultraviolet Rays/adverse effects , Mice , Mice, Inbred C57BL , Time Factors , Ultraviolet Therapy
20.
Front Immunol ; 15: 1366962, 2024.
Article in English | MEDLINE | ID: mdl-38736880

ABSTRACT

Hematopoietic stem cell transplantation and cell therapies like CAR-T are costly, complex therapeutic procedures. Outpatient models, including at-home transplantation, have been developed, resulting in similar survival results, reduced costs, and increased patient satisfaction. The complexity and safety of the process can be addressed with various emerging technologies (artificial intelligence, wearable sensors, point-of-care analytical devices, drones, virtual assistants) that allow continuous patient monitoring and improved decision-making processes. Patients, caregivers, and staff can also benefit from improved training with simulation or virtual reality. However, many technical, operational, and above all, ethical concerns need to be addressed. Finally, outpatient or at-home hematopoietic transplantation or CAR-T therapy creates a different, integrated operative system that must be planned, designed, and carefully adapted to the patient's characteristics and distance from the hospital. Patients, clinicians, and their clinical environments can benefit from technically improved at-home transplantation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Home Care Services , Humans , Hematopoietic Stem Cell Transplantation/methods , Immunotherapy, Adoptive/methods , Artificial Intelligence
SELECTION OF CITATIONS
SEARCH DETAIL
...