Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.548
Filter
1.
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
2.
J Immunother Cancer ; 12(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39379097

ABSTRACT

Humanized immunodeficient mice serve as critical models for investigating the functional interplay between transplanted human cells and a pre-reconstituted human immune system. These models facilitate the study of molecular and cellular pathogenic mechanisms and enable the evaluation of the efficacy and toxicity of immunotherapies, thereby accelerating their preclinical and clinical development. Current strategies rely on inefficient, long-term/delayed hematopoietic reconstitution by CD34+ hematopoietic progenitors or short-term reconstitution with peripheral blood mononuclear cells (PB-MNCs) associated with high rates of graft-versus-host disease (GvHD) and an inefficient representation of immune cell populations. Here, we hypothesized that immunologically naïve cord blood mononuclear cells (CB-MNCs) could serve as a superior alternative, providing long-lasting and functionally effective immune reconstitution. We conducted a comprehensive comparison between the non-obese diabetic (NOD).Cg-Prkdc∧ˆscid-IL2rg∧ˆtm1Wjl/SzJ (NSG) and NSG-Tg(CMV-IL3,CSF2,KITLG)∧ˆ1Eav/MloySzJ (NSGS) immunodeficient mouse models following humanization with either PB-MNCs or CB-MNCs. We assessed the engraftment dynamics of various human immune cells over time and monitored the development of GvHD in both models. For the most promising model, we extensively evaluated immune cell functionality in vitro and in vivo using sarcoma and leukemia xenografts. Humanizing NSGS mice with CB-MNCs results in a rapid, robust, and sustained representation of a diverse range of functional human lymphoid and myeloid cell populations while minimizing GvHD incidence. In this model, human immune cell populations significantly impair the growth and engraftment of sarcoma and B-cell acute lymphoblastic leukemia cells, with a significant inverse correlation between immune cell levels and tumor growth. This study establishes a fast, efficient, and reliable in vivo platform for various applications in cancer immunotherapy, particularly for exploring the complex interactions between cancer cells, immune cells, and the tumor microenvironment in vivo, prior to clinical development.


Subject(s)
Fetal Blood , Graft vs Host Disease , Leukocytes, Mononuclear , Mice, Inbred NOD , Animals , Humans , Graft vs Host Disease/immunology , Mice , Fetal Blood/cytology , Fetal Blood/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/transplantation , Leukocytes, Mononuclear/metabolism , Disease Models, Animal , Myeloid Cells/immunology , Myeloid Cells/metabolism , Lymphocytes/immunology , Mice, SCID
3.
Front Immunol ; 15: 1440911, 2024.
Article in English | MEDLINE | ID: mdl-39229273

ABSTRACT

Introduction: The human leukocyte antigen (HLA) evolutionary divergence (HED) reflects immunopeptidome diversity and has been shown to predict the response of tumors to immunotherapy. Its impact on allogeneic hematopoietic stem cell transplantation (HSCT) is controversial in different studies. Methods: In this study, we retrospectively analyzed the clinical impact of class I and II HED in 225 acute lymphoblastic leukemia patients undergoing HSCT from related haploidentical donors. The HED for recipient, donor, and donor-recipient pair was calculated based on Grantham distance, which accounts for variations in the composition, polarity, and volume of each amino acid within the peptide-binding groove of two HLA alleles. The median value of HED scores was used as a cut-off to stratify patients with high or low HED. Results: The class I HED for recipient (R_HEDclass I) showed the strongest association with cumulative incidence of relapse (12.2 vs. 25.0%, P = 0.00814) but not with acute graft-versus-host disease. The patients with high class II HED for donor-recipient (D/R_HEDclass II) showed a significantly higher cumulative incidence of severe aGVHD than those with low D/R_HEDclass II (24.0% vs. 6.1%, P = 0.0027). Multivariate analysis indicated that a high D/R_HEDclass II was an independent risk factor for the development of severe aGVHD (P = 0.007), and a high R_HEDclass I had a more than two-fold reduced risk of relapse (P = 0.028). However, there was no discernible difference in overall survival (OS) or disease-free survival (DFS) for patients with high or low HED, which was inconsistent with the previous investigation. Discussion: While the observation are limited by the presented single center retrospective cohort, the results show that HED has poor prognostic value in OS or DFS, as well as the associations with relapse and aGVHD. In haploidentical setting, class II HED for donor-recipient pair (D/R_HEDclass II) is an independent and novel risk factor for finding the best haploidentical donor, which could potentially influence clinical practice if verified in larger cohorts.


Subject(s)
Donor Selection , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Male , Female , Adult , Adolescent , Middle Aged , Child , Retrospective Studies , Risk Factors , Graft vs Host Disease/immunology , Graft vs Host Disease/etiology , Graft vs Host Disease/genetics , Young Adult , HLA Antigens/genetics , HLA Antigens/immunology , Child, Preschool , Transplantation, Haploidentical , Tissue Donors , Evolution, Molecular
4.
Cell Rep Med ; 5(9): 101717, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39243754

ABSTRACT

Indoleamine-2,3-dioxygenase (IDO)1 degrades tryptophan, obtained through dietary intake, into immunoregulatory metabolites of the kynurenine pathway. Deficiency or blockade of IDO1 results in the enhancement of autoimmune severity in rodent models and increased susceptibility to developing autoimmunity in humans. Despite this, therapeutic modalities that leverage IDO1 for the treatment of autoimmunity remain limited. Here, we use messenger (m)RNA formulated in lipid nanoparticles (LNPs) to deliver a human IDO1 variant containing the myristoylation site of Src to anchor the protein to the inner face of the plasma membrane. This membrane-anchored IDO1 has increased protein production, leading to increased metabolite changes, and ultimately ameliorates disease in three models of T cell-mediated autoimmunity: experimental autoimmune encephalomyelitis (EAE), rat collagen-induced arthritis (CIA), and acute graft-versus-host disease (aGVHD). The efficacy of IDO1 is correlated with hepatic expression and systemic tryptophan depletion. Thus, the delivery of membrane-anchored IDO1 by mRNA suppresses the immune response in several well-characterized models of autoimmunity.


Subject(s)
Autoimmunity , Encephalomyelitis, Autoimmune, Experimental , Indoleamine-Pyrrole 2,3,-Dioxygenase , RNA, Messenger , T-Lymphocytes , Tryptophan , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Animals , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Rats , Tryptophan/metabolism , Graft vs Host Disease/immunology , Arthritis, Experimental/immunology , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Mice , Nanoparticles/chemistry , Female
5.
N Engl J Med ; 391(11): 1002-1014, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39292927

ABSTRACT

BACKGROUND: Colony-stimulating factor 1 receptor (CSF1R)-dependent monocytes and macrophages are key mediators of chronic graft-versus-host disease (GVHD), a major long-term complication of allogeneic hematopoietic stem-cell transplantation. The CSF1R-blocking antibody axatilimab has shown promising clinical activity in chronic GVHD. METHODS: In this phase 2, multinational, pivotal, randomized study, we evaluated axatilimab at three different doses in patients with recurrent or refractory chronic GVHD. Patients were randomly assigned to receive axatilimab, administered intravenously, at a dose of 0.3 mg per kilogram of body weight every 2 weeks (0.3-mg dose group), at a dose of 1 mg per kilogram every 2 weeks (1-mg dose group), or at a dose of 3 mg per kilogram every 4 weeks (3-mg dose group). The primary end point was overall response (complete or partial response) in the first six cycles; the key secondary end point was a patient-reported decrease in chronic GVHD symptom burden, as assessed by a reduction of more than 5 points on the modified Lee Symptom Scale (range, 0 to 100, with higher scores indicating worse symptoms). The primary end point would be met if the lower bound of the 95% confidence interval exceeded 30%. RESULTS: A total of 241 patients were enrolled (80 patients in the 0.3-mg dose group, 81 in the 1-mg dose group, and 80 in the 3-mg dose group). The primary end point was met in all the groups; an overall response was observed in 74% (95% confidence interval [CI], 63 to 83) of the patients in the 0.3-mg dose group, 67% (95% CI, 55 to 77) of the patients in the 1-mg dose group, and 50% (95% CI, 39 to 61) of the patients in the 3-mg dose group. A reduction of more than 5 points on the modified Lee Symptom Scale was reported in 60%, 69%, and 41% of the patients in the three dose groups, respectively. The most common adverse events were dose-dependent transient laboratory abnormalities related to CSF1R blockade. Adverse events leading to discontinuation of axatilimab occurred in 6% of the patients in the 0.3-mg dose group, 22% in the 1-mg dose group, and 18% in the 3-mg dose group. CONCLUSIONS: Targeting CSF1R-dependent monocytes and macrophages with axatilimab resulted in a high incidence of response among patients with recurrent or refractory chronic GVHD. (Funded by Syndax Pharmaceuticals and Incyte; AGAVE-201 ClinicalTrials.gov number, NCT04710576.).


Subject(s)
Antibodies, Monoclonal, Humanized , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Receptor, Macrophage Colony-Stimulating Factor , Adolescent , Adult , Aged , Aged, 80 and over , Child , Female , Humans , Male , Middle Aged , Young Adult , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Chronic Disease/drug therapy , Dose-Response Relationship, Drug , Graft vs Host Disease/diagnosis , Graft vs Host Disease/drug therapy , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects , Infusions, Intravenous , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Recurrence , Severity of Illness Index , Treatment Outcome
8.
Virulence ; 15(1): 2404225, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39267271

ABSTRACT

The THαß host immunological pathway contributes to the response to infectious particles (viruses and prions). Furthermore, there is increasing evidence for associations between autoimmune diseases, and particularly type 2 hypersensitivity disorders, and the THαß immune response. For example, patients with systemic lupus erythematosus often produce anti-double stranded DNA antibodies and anti-nuclear antibodies and show elevated levels of type 1 interferons, type 3 interferons, interleukin-10, IgG1, and IgA1 throughout the disease course. These cytokines and antibody isotypes are associated with the THαß host immunological pathway. Similarly, the type 2 hypersensitivity disorders myasthenia gravis, Graves' disease, graft-versus-host disease, autoimmune hemolytic anemia, immune thrombocytopenia, dermatomyositis, and Sjögren's syndrome have also been linked to the THαß pathway. Considering the potential associations between these diseases and dysregulated THαß immune responses, therapeutic strategies such as anti-interleukin-10 or anti-interferon α/ß could be explored for effective management.


Subject(s)
Lupus Erythematosus, Systemic , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/complications , Sjogren's Syndrome/immunology , Graft vs Host Disease/immunology , Autoimmune Diseases/immunology , Cytokines/immunology , Myasthenia Gravis/immunology , Anemia, Hemolytic, Autoimmune/immunology , Graves Disease/immunology , Graves Disease/complications , Dermatomyositis/immunology
9.
J Immunother Cancer ; 12(9)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39242117

ABSTRACT

BACKGROUND: In allogeneic-hematopoietic stem cell transplantation for acute myeloid leukemia (AML), donor T cells combat leukemia through the graft-versus-leukemia (GVL) effect, while they also pose a risk of triggering life-threatening graft-versus-host disease (GVHD) by interacting with recipient cells. The onset of GVHD hinges on the interplay between donor T cells and recipient antigen-presenting cells (APCs), sparking T-cell activation. However, effective methods to balance GVHD and GVL are lacking. METHODS: In our study, we crafted nanocapsules by layering polycationic aminated gelatin and polyanionic alginate onto the surface of T cells, examining potential alterations in their fundamental physiological functions. Subsequently, we established an AML mouse model and treated it with transplantation of bone marrow cells (BMCs) combined with encapsulated T cells to investigate the GVL and anti-GVHD effects of encapsulated T cells. In vitro co-culture was employed to probe the effects of encapsulation on immune synapses, co-stimulatory molecules, and tumor-killing pathways. RESULTS: Transplantation of BMCs combined with donor T cells selectively encapsulated onto AML mice significantly alleviates GVHD symptoms while preserving essential GVL effects. Encapsulated T cells exerted their immunomodulatory effects by impeding the formation of immune synapses with recipient APCs, thereby downregulating co-stimulatory signals such as CD28-CD80, ICOS-ICOSL, and CD40L-CD40. Recipient mice receiving encapsulated T-cell transplantation exhibited a marked increase in donor Ly-5.1-BMC cell numbers, accompanied by unaltered in vivo expression levels of perforin and granzyme B. While transient inhibition of donor T-cell cytotoxicity in the tumor microenvironment was observed in vitro following single-cell nanoencapsulation, subsequent restoration to normal antitumor activity ensued, attributed to selective permeability of encapsulated vesicle shells and material degradation. Moreover, the expression of apoptotic proteins and FAS-FAS ligand pathway at normal levels was still observed in leukemia tumor cells. CONCLUSIONS: Encapsulated donor T cells effectively mitigate GVHD while preserving the GVL effect by minimizing co-stimulatory signaling with APCs through early immune isolation. Subsequent degradation of nanocapsules restores T-cell cytotoxic efficacy against AML cells, mediated by cytotoxic pathways. Using transplant-encapsulated T cells offers a promising strategy to suppress GVHD while preserving the GVL effect.


Subject(s)
Disease Models, Animal , Graft vs Host Disease , Leukemia, Myeloid, Acute , T-Lymphocytes , Animals , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/immunology , Mice , Graft vs Host Disease/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Humans , Graft vs Leukemia Effect , Nanocapsules/chemistry
10.
J Immunother Cancer ; 12(9)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39244214

ABSTRACT

BACKGROUND: Immunodeficient mice engrafted with peripheral blood mononuclear cells (PBMCs) are models to study new cancer immunotherapy agents. However, this approach is associated with xenograft-versus-host disease (xGVHD), which starts early after PBMC transfer and limits the duration and interpretation of experiments. Here, we explore different approaches to overcome xGVHD and better support the development of cancer immunotherapies. METHODS: Immunodeficient NOD-scid IL2Rgnull (NSG) mice were intravenously transferred with human PBMCs and subcutaneously co-engrafted with HT29 human colon carcinoma cells. Diverse strategies to reduce xGVHD while preserving the antitumor activity of human immune cells were evaluated: (1) ex vivo immune graft modification by depleting CD4+ T cells pre-transfer using magnetic beads, (2) post-transplantation cyclophosphamide administration to eliminate proliferating xenoreactive T-cell clones and (3) using major histocompatibility complex (MHC) class I and II-deficient NSG mice: (Kb Db)null (IA)null (MHC-dKO NSG). Body weight and plasma murine alanine aminotransferase levels were measured as indicators of xGVHD and tumor size was measured every 2-3 days to monitor antitumor activity. The antitumor effects and pharmacodynamics of nivolumab plus ipilimumab and an anti-epithelial cell adhesion molecule (EpCAM)/CD3 T-cell engager (αEpCAM/CD3 bispecific antibody (BsAb)) were evaluated in the model. RESULTS: CD4+ T-cell depletion attenuates xGVHD but also abrogates the antitumor activity. Cyclophosphamide limits the antitumor response and does not substantially prevent xGVHD. In contrast, xGVHD was significantly attenuated in MHC-dKO NSG recipients, while the antitumor effect of human PBMCs was preserved. Furthermore, the administration of nivolumab plus ipilimumab caused exacerbated xGVHD in conventional NSG mice, thereby precluding the observation of their antitumor effects. Severe xGVHD did not occur in MHC-dKO NSG mice thus enabling the study of complete and durable tumor rejections. Similarly, NSG mice treated with an αEpCAM/CD3 BsAb showed complete tumor regressions, but died due to xGVHD. In contrast, MHC-dKO NSG mice on treatment with the αEpCAM/CD3 BsAb achieved complete tumor responses without severe xGVHD. A significant proportion of mice rendered tumor-free showed tumor rejection on rechallenge with HT29 cells without further treatment. Finally, tumor-infiltrating CD8+ T-cell number increase, activation and CD137 upregulation were observed on αEpCAM/CD3 BsAb treatment. CONCLUSION: Humanized MHC-dKO immunodeficient mice allow and refine the preclinical testing of immunotherapy agents for which experimentation is precluded in conventional immunodeficient mice due to severe xGVHD.


Subject(s)
Immune Checkpoint Inhibitors , Animals , Humans , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice, SCID , Mice, Inbred NOD , Histocompatibility Antigens Class I/immunology , Xenograft Model Antitumor Assays , Histocompatibility Antigens Class II/immunology , Graft vs Host Disease/drug therapy , Graft vs Host Disease/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
11.
Transplant Proc ; 56(8): 1841-1846, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39232921

ABSTRACT

BACKGROUND: Haploidentical hematopoietic stem cell transplantation (haplo-HSCT) with TCR αß+/CD19+ cell depletion is a promising therapeutic alternative for children with nonmalignant hematologic disorders, especially in low-income countries where finding a compatible donor is challenging. The use of this transplantation approach for nonmalignant hematologic disorders has not been previously described in the Peruvian pediatric population. METHODS: We present the outcomes of children under 19 with nonmalignant hematologic disorders who underwent haplo-HSCT with TCR αß+/CD19+ cell depletion between 2018-2022 at a referral center in Lima, Peru. Survival probabilities and cumulative incidence functions were calculated using the Kaplan-Meier method. RESULTS: A total of 17 children aged between 1 to 18.6 years (median = 9.7 years) were included. The follow-up period ranged from 10 days to 66.20 months, with a median of 4.34 months. The probability of overall survival, event-free survival, and failure-free survival was 33.70%, 31.40%, and 68.8%, respectively. The incidence rate of graft failure was 49.80%, while the mortality rate not associated with graft failure was 18.8%. The incidence rate of acute graft-versus-host disease (GvHD) was 25.60%, and the incidence rate of viral infections was 59.40%. CONCLUSIONS: The high incidence rates of graft failure and viral infections suggest that these factors may negatively impact the survival of children with nonmalignant hematologic disorders who undergo haplo-HSCT with TCR αß+/CD19+ cell depletion. Therefore, optimizing the current conditioning regimens and ensuring timely access to first, second, and third-line antivirals is crucial to improve the survival of these patients.


Subject(s)
Antigens, CD19 , Graft vs Host Disease , Hematologic Diseases , Hematopoietic Stem Cell Transplantation , Transplantation, Haploidentical , Humans , Child , Adolescent , Child, Preschool , Hematopoietic Stem Cell Transplantation/adverse effects , Peru , Male , Female , Infant , Hematologic Diseases/therapy , Hematologic Diseases/surgery , Graft vs Host Disease/immunology , Receptors, Antigen, T-Cell, alpha-beta , Treatment Outcome , Lymphocyte Depletion , Retrospective Studies
12.
Int Immunopharmacol ; 142(Pt A): 112973, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39217881

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by persistent immune cell activation and the overproduction of autoantibodies, affecting various organs such as joints, kidneys, and skin. Interleukin-15 (IL-15) is a pleiotropic cytokine that modulates immune cells of the innate and adaptive immune systems, playing a crucial role in the development of inflammatory and protective immune responses. However, the role of IL-15 in SLE pathogenesis and the therapeutic effects of IL-15 blockade on SLE remain unknown. In this study, we conducted flow cytometry analysis and identified a significant increase in the frequencies of IL-15+ and IL-15R+ cells in peripheral blood CD4+ T cells, CD8+ T cells, dendritic cells (DCs), monocytes, and natural killer (NK) cells of patients with SLE compared to healthy controls (HCs). Besides, we found elevated levels of serum IL-15 in SLE patients compared to HCs. Furthermore, we evaluted the effectiveness of IL-15 mAb treatment in a chronic graft-versus-host disease (cGVHD) mouse model of SLE. We observed that the IL-15 mAb treatment effectively reduced the frequencies of CD4+CD44hiCD62LloPD-1+CD153+ senescent CD4+ T cells, B220+CD11c+T-bet+ age-associated B cells (ABCs), Tfh cells, and germinal center (GC) B cells, alleviated lupus-associated manifestations such as serum anti-double-stranded DNA antibody (anti-dsDNA) and kidney injury in the SLE mouse model of cGVHD. These findings provide compelling preclinical evidence suggesting the pathogenic role of IL-15 in SLE and the therapeutic potential of IL-15 blockade in the treatment of SLE.


Subject(s)
Interleukin-15 , Lupus Erythematosus, Systemic , Interleukin-15/blood , Interleukin-15/metabolism , Interleukin-15/immunology , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/blood , Animals , Female , Adult , Mice , Male , Graft vs Host Disease/immunology , Graft vs Host Disease/drug therapy , Graft vs Host Disease/blood , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Killer Cells, Natural/immunology , Middle Aged , Dendritic Cells/immunology , Receptors, Interleukin-15/metabolism , CD4-Positive T-Lymphocytes/immunology
13.
Curr Res Transl Med ; 72(3): 103464, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39232416

ABSTRACT

BACKGROUND: While the detrimental role of donor-specific anti-HLA antibodies (DSAs) is well-described in the setting of hematopoietic stem cell transplantation (HSCT), few studies focus on non donor-specific ones and with controversial results. METHODS: We here report our monocenter experience on 64 adult patients receiving allogeneic HSCT from a HLA-mismatched donor between 2014 and 2022 who were tested for the presence of anti-HLA antibodies before transplant, focusing on fifteen patients with non donor-specific anti-HLA antibodies. RESULTS: The survival of patients with non donor-specific anti-HLA antibodies was inferior with respect to patients without anti-HLA antibodies and similar to patients with DSAs. Median survival of patients with non donor-specific anti-HLA antibodies was 21 months (95 % CI: 9-42) vs. 61 months (95 % CI: 17-77) among the anti-HLA antibody-negative patients, with a significantly higher mortality incidence rate ratio (3.3 times-fold greater, p = 0.01). No pattern of death causes was found CONCLUSIONS: In this monocenter series of HLA-mismatched HSCTs, impaired survival was observed in adult patients having non donor-specific anti-HLA antibodies before transplant, similar to those with DSAs. Our findings support those antibodies as a negative predictive factor even if they are not directed against the donor, thus warranting further investigation on larger cohorts.


Subject(s)
HLA Antigens , Hematopoietic Stem Cell Transplantation , Transplantation, Homologous , Humans , Male , Female , Adult , HLA Antigens/immunology , Hematopoietic Stem Cell Transplantation/mortality , Hematopoietic Stem Cell Transplantation/adverse effects , Middle Aged , Young Adult , Isoantibodies/blood , Isoantibodies/immunology , Tissue Donors , Histocompatibility Testing , Aged , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Graft vs Host Disease/etiology , Adolescent , Retrospective Studies
14.
Int Rev Cell Mol Biol ; 388: 95-123, 2024.
Article in English | MEDLINE | ID: mdl-39260939

ABSTRACT

Organ transplantation increases life expectancy and improves the quality of life of patients experiencing specific conditions such as terminal organ failure. Despite matching efforts between donor and recipient, immune activation can interfere with allograft survival after transplantation if immunosuppression is not used. With both innate and adaptive responses, this is a complicated immunological process. This can lead to organ rejection, or graft-versus-host disease (GVHD), depending on the origin of the immune response. Inflammatory factors, such as chemokine receptors and their ligands, are involved in a wide variety of immunological processes, including modulating transplant rejection or GVHD, therefore, chemokine biology has been a major focus of transplantation studies. These molecules attract circulating peripheral leukocytes to infiltrate into the allograft and facilitate dendritic and T cell trafficking between lymph nodes and the graft during the allogeneic response. In this chapter, we will review the most relevant chemokine receptors such as CXCR3 and CCR5, among others, and their ligands involved in the process of allograft rejection for solid organ transplantation and graft-versus-host disease in the context of hematopoietic cell transplantation.


Subject(s)
Graft Rejection , Graft vs Host Disease , Receptors, Chemokine , Humans , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Graft Rejection/immunology , Animals , Receptors, Chemokine/metabolism
15.
Stem Cell Res Ther ; 15(1): 320, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39334441

ABSTRACT

Although liver transplantation (LT) is an effective strategy for end-stage liver diseases, the shortage of donor organs and the immune rejection hinder its widespread implementation in clinical practice. Mesenchymal stem cells (MSCs) transplantation offers a promising approach for patients undergoing liver transplantation due to their immune regulatory capabilities, hepatic protection properties, and multidirectional differentiation potential. In this review, we summarize the potential applications of MSCs transplantation in various LT scenarios. MSCs transplantation has demonstrated effectiveness in alleviating hepatic ischemia-reperfusion injury, enhancing the viability of liver grafts, preventing acute graft-versus-host disease, and promoting liver regeneration in split LT therapy. We also discuss the clinical progress, and explore the immunomodulatory functions of MSCs in response to both adaptive and innate immune responses. Furthermore, we emphasize the interactions between MSCs and different immune cells, including T cells, B cells, plasma cells, natural killer cells, dendritic cells, Kupffer cells, and neutrophils, to provide new insights into the immunomodulatory properties of MSCs in adoptive cell therapy.


Subject(s)
Immunomodulation , Liver Transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/cytology , Animals , Reperfusion Injury/therapy , Reperfusion Injury/immunology , Graft vs Host Disease/immunology , Graft vs Host Disease/therapy , Liver Regeneration
16.
Front Immunol ; 15: 1433091, 2024.
Article in English | MEDLINE | ID: mdl-39328417

ABSTRACT

Acute graft-versus-host disease (aGVHD) is a major complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT) and contributes to high morbidity and mortality. However, our current understanding of the development and progression of aGVHD after allo-HSCT remains limited. To identify the potential biomarkers for the prevention and treatment of aGVHD during the early hematopoietic reconstruction after transplantation, we meticulously performed a comparative analysis of single-cell RNA sequencing data from post-transplant patients with or without aGVHD. Prior to the onset of aGVHD, monocytes in the peripheral blood of patients with aGVHD experienced a dramatic rise and activation on day 21 post-transplantation. This phenomenon is closely aligned with clinical cohort results obtained from blood routine examinations. Furthermore, in vitro co-culture experiments showed that peripheral blood monocytes extracted from patients with aGVHD approximately 21 days post-transplantation induced a significantly higher proliferation rate of allogeneic T cells compared to those from patients without aGVHD. Our study indicates that monocytes could be a crucial early clinical risk factor for the development of aGVHD, and this insight could potentially guide the timing of monitoring efforts, recommending assessments at the pivotal juncture of approximately day 21 post-transplantation, shedding fresh light on the significance of early hematopoietic regeneration in relation to the onset of aGVHD.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Monocytes , Transplantation, Homologous , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Monocytes/immunology , Male , Female , Adult , Middle Aged , Transplantation, Homologous/adverse effects , Risk Factors , Acute Disease , Young Adult , Adolescent , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
Clin Transl Sci ; 17(8): e70014, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39162578

ABSTRACT

Dose optimization of sirolimus may further improve outcomes in allogeneic hematopoietic cell transplant (HCT) patients receiving post-transplantation cyclophosphamide (PTCy) to prevent graft-versus-host disease (GVHD). Sirolimus exposure-response association studies in HCT patients (i.e., the association of trough concentration with clinical outcomes) have been conflicting. Sirolimus has important effects on T-cells, including conventional (Tcons) and regulatory T-cells (Tregs), both of which have been implicated in the mechanisms by which PTCy prevents GVHD, but there is an absence of validated biomarkers of sirolimus effects on these cell subsets. Considering the paucity of existing biomarkers and the complexities of the immune system, we conducted a literature review to inform a quantitative systems pharmacology (QSP) model of GVHD. The published literature presented multiple challenges. The sirolimus pharmacokinetic models insufficiently describe sirolimus distribution to relevant physiological compartments. Despite multiple publications describing sirolimus effects on Tcons and Tregs in preclinical and human ex vivo models, consistent parameters relating sirolimus concentrations to circulating Tcons and Tregs could not be found. Each aspect presents a challenge in building a QSP model of sirolimus and its temporal effects on T-cell subsets and GVHD prevention. To optimize GVHD prevention regimens, phase I studies and systematic studies of immunosuppressant concentration-effect association are needed for QSP modeling.


Subject(s)
Cyclophosphamide , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Immunosuppressive Agents , Sirolimus , Humans , Sirolimus/administration & dosage , Graft vs Host Disease/prevention & control , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Cyclophosphamide/adverse effects , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacokinetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Animals , Models, Biological
18.
Pathol Res Pract ; 262: 155518, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39146830

ABSTRACT

Currently, CAR-T cell therapy relies on an individualized manufacturing process in which patient's own T cells are infused back into patients after being engineered and expanded ex vivo. Despite the astonishing outcomes of autologous CAR-T cell therapy, this approach is endowed with several limitations and drawbacks, such as high cost and time-consuming manufacturing process. Switching the armature of CAR-T cell therapy from autologous settings to allogeneic can overcome several bottlenecks of the current approach. Nevertheless, the use of allogeneic CAR-T cells is limited by the risk of life-threatening GvHD. Thus, in recent years, developing a method to move CAR-T cell therapy to allogeneic settings without the risk of GvHD has become a hot research topic in this field. Since the alloreactivity of αß T-cell receptor (TCR) accounts for developing GvHD, several efforts have been made to disrupt endogenous TCR of allogeneic CAR-T cells using gene editing tools to prevent GvHD. Nonetheless, the off-target activity of gene editing tools and their associated genotoxicities, as well as the negative consequences of endogenous TCR disruption, are the main concerns of using this approach. As an alternative, CAR αß-T cells can be replaced with other types of CAR-engineered cells that are capable of recognizing and killing malignant cells through CAR while avoiding the induction of GvHD. These alternatives include T cell subsets with restricted TCR repertoire (γδ-T, iNKT, virus-specific T, double negative T cells, and MAIT cells), killer cells (NK and CIK cells), non-lymphocytic cells (neutrophils and macrophages), stem/progenitor cells, and cell-free extracellular vesicles. In this review, we discuss how these alternatives can move CAR-based immunotherapy to allogeneic settings to overcome the bottlenecks of autologous manner without the risk of GvHD. We comprehensively discuss the pros and cons of these alternatives over the traditional CAR αß-T cells in light of their preclinical studies and clinical trials.


Subject(s)
Graft vs Host Disease , Immunotherapy, Adoptive , Neoplasms , Receptors, Chimeric Antigen , Humans , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Graft vs Host Disease/therapy , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Neoplasms/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/immunology , Animals , Gene Editing/methods , Transplantation, Homologous/methods
19.
Front Immunol ; 15: 1365946, 2024.
Article in English | MEDLINE | ID: mdl-39131155

ABSTRACT

Introduction: Humanized mouse models to recapitulate human biological systems still have limitations, such as the onset of lethal graft-versus-host disease (GvHD), a variable success rate, and the low accessibility of total body irradiation (TBI). Recently, mice modified with the CD47-SIRPA axis have been studied to improve humanized mouse models. However, such trials have been rarely applied in NOD mice. In this study, we created a novel mouse strain, NOD-CD47nullRag2nullIL-2rγnull (RTKO) mice, and applied it to generate humanized mice. Methods: Four-week-old female NOD-Rag2nullIL-2rγnull (RID) and RTKO mice pre-conditioned with TBI or busulfan (BSF) injection were used for generating human CD34+ hematopoietic stem cell (HSC) engrafted humanized mice. Clinical signs were observed twice a week, and body weight was measured once a week. Flow cytometry for human leukocyte antigens was performed at intervals of four weeks or two weeks, and mice were sacrificed at 48 weeks after HSC injection. Results: For a long period from 16 to 40 weeks post transplantation, the percentage of hCD45 was mostly maintained above 25% in all groups, and it was sustained the longest and highest in the RTKO BSF group. Reconstruction of human leukocytes, including hCD3, was also most prominent in the RTKO BSF group. Only two mice died before 40 weeks post transplantation in all groups, and there were no life-threatening GvHD lesions except in the dead mice. The occurrence of GvHD has been identified as mainly due to human T cells infiltrating tissues and their related cytokines. Discussion: Humanized mouse models under all conditions applied in this study are considered suitable models for long-term experiments based on the improvement of human leukocytes reconstruction and the stable animal health. Especially, RTKO mice pretreated with BSF are expected to be a valuable platform not only for generating humanized mice but also for various immune research fields.


Subject(s)
Busulfan , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Mice, Inbred NOD , Mice, Knockout , Transplantation Conditioning , Animals , Busulfan/pharmacology , Humans , Mice , Hematopoietic Stem Cell Transplantation/methods , Transplantation Conditioning/methods , Hematopoietic Stem Cells/metabolism , Female , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/deficiency , Graft vs Host Disease/prevention & control , Graft vs Host Disease/immunology , Disease Models, Animal , Whole-Body Irradiation
20.
Front Immunol ; 15: 1429335, 2024.
Article in English | MEDLINE | ID: mdl-39131162

ABSTRACT

We have previously reported that nanoparticles (NPs) loaded with IL-2 and TGF-ß and targeted to T cells induced polyclonal T regulatory cells (Tregs) that protected mice from graft-versus-host disease (GvHD). Here, we evaluated whether administration of these NPs during alloantigen immunization could prevent allograft rejection by converting immunogenic responses to tolerogenic ones. Using C57BL/6 mice and BALB/c mice as either donors or recipients of allogeneic splenocytes, we found that treatment with the tolerogenic NPs in both strains of mice resulted in a marked inhibition of mixed lymphocyte reaction (MLR) to donor cell alloantigen but not to third-party control mouse cells after transfer of the allogeneic cells. The decreased alloreactivity associated with a four- to fivefold increase in the number of CD4+ and CD8+ T regulatory cells (Tregs) and the acquisition of a tolerogenic phenotype by recipient dendritic cells (DCs) in NP-treated mice. As allogeneic cells persisted in NP-treated mice, these findings suggest that tolerogenic NPs can induce alloantigen-specific Tregs and tolerogenic DCs promoting tolerogenic responses to alloantigen. By inhibiting reactivity to allotransplant, this approach could help reduce the need for immune suppression for the maintenance of allografts.


Subject(s)
Interleukin-2 , Isoantigens , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles , T-Lymphocytes, Regulatory , Transforming Growth Factor beta , Transplantation Tolerance , Animals , Isoantigens/immunology , Transplantation Tolerance/immunology , Mice , Transforming Growth Factor beta/immunology , T-Lymphocytes, Regulatory/immunology , Interleukin-2/immunology , Dendritic Cells/immunology , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Female
SELECTION OF CITATIONS
SEARCH DETAIL