Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 142(15): 1281-1296, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37478401

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is a rare X-linked disorder characterized by combined immunodeficiency, eczema, microthrombocytopenia, autoimmunity, and lymphoid malignancies. Gene therapy (GT) to modify autologous CD34+ cells is an emerging alternative treatment with advantages over standard allogeneic hematopoietic stem cell transplantation for patients who lack well-matched donors, avoiding graft-versus-host-disease. We report the outcomes of a phase 1/2 clinical trial in which 5 patients with severe WAS underwent GT using a self-inactivating lentiviral vector expressing the human WAS complementary DNA under the control of a 1.6-kB fragment of the autologous promoter after busulfan and fludarabine conditioning. All patients were alive and well with sustained multilineage vector gene marking (median follow-up: 7.6 years). Clinical improvement of eczema, infections, and bleeding diathesis was universal. Immune function was consistently improved despite subphysiologic levels of transgenic WAS protein expression. Improvements in platelet count and cytoskeletal function in myeloid cells were most prominent in patients with high vector copy number in the transduced product. Two patients with a history of autoimmunity had flares of autoimmunity after GT, despite similar percentages of WAS protein-expressing cells and gene marking to those without autoimmunity. Patients with flares of autoimmunity demonstrated poor numerical recovery of T cells and regulatory T cells (Tregs), interleukin-10-producing regulatory B cells (Bregs), and transitional B cells. Thus, recovery of the Breg compartment, along with Tregs appears to be protective against development of autoimmunity after GT. These results indicate that clinical and laboratory manifestations of WAS are improved with GT with an acceptable safety profile. This trial is registered at clinicaltrials.gov as #NCT01410825.


Subject(s)
Eczema , Hematopoietic Stem Cell Transplantation , Wiskott-Aldrich Syndrome , Humans , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/therapy , Wiskott-Aldrich Syndrome Protein/genetics , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects , Genetic Therapy/methods , Eczema/etiology , Eczema/metabolism , Eczema/therapy
2.
Clin Immunol ; 238: 109014, 2022 05.
Article in English | MEDLINE | ID: mdl-35447312

ABSTRACT

Potential etiologies of T-B+NK+ SCID include both hematopoietic defects and thymic aplasia. The management of patients with this phenotype, identified by newborn screen, may be unclear in the absence of a genetic diagnosis. We report an infant with lymphocyte flow cytometry consistent with T-B+NK+ SCID and reduced proliferative response to phytohemagglutinin. The patient had no genetic diagnosis after targeted panel and exome sequencing. The decision to trend laboratory values rather than move immediately to hematopoietic cell transplant was made given the absence of a genetic defect and the finding of a normal thymus on ultrasound. During the course of evaluation for transplant, the patient unexpectedly had normalization of T cell number and function. This case demonstrates a role for mediastinal ultrasound and the utility of trending laboratory values in patients with severe T cell lymphopenia but no genetic diagnosis, given the small but important possibility of spontaneous resolution.


Subject(s)
Hematopoietic Stem Cell Transplantation , Lymphopenia , Severe Combined Immunodeficiency , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Infant, Newborn , Lymphopenia/complications , Neonatal Screening , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/genetics , T-Lymphocytes
3.
Blood ; 131(26): 2967-2977, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29728406

ABSTRACT

Allogeneic hematopoietic stem cell transplant (HSCT) typically results in donor T-cell engraftment and function in patients with severe combined immunodeficiency (SCID), but humoral immunity, particularly when using donors other than matched siblings, is variable. B-cell function after HSCT for SCID depends on the genetic cause, the use of pre-HSCT conditioning, and whether donor B-cell chimerism is achieved. Patients with defects in IL2RG or JAK3 undergoing HSCT without conditioning often have poor B-cell function post-HSCT, perhaps as a result of impairment of IL-21 signaling in host-derived B cells. To investigate the effect of pre-HSCT conditioning on B-cell function, and the relationship of in vitro B-cell function to clinical humoral immune status, we analyzed 48 patients with IL2RG/JAK3 SCID who were older than 2 years after HSCT with donors other than matched siblings. T follicular helper cells (TFH) developed in these patients with kinetics similar to healthy young children; thus, poor B-cell function could not be attributed to a failure of TFH development. In vitro differentiation of B cells into plasmablasts and immunoglobulin secretion in response to IL-21 strongly correlated with the use of conditioning, donor B-cell engraftment, freedom from immunoglobulin replacement, and response to tetanus vaccine. Patients receiving immunoglobulin replacement who had normal serum immunoglobulin M showed poor response to IL-21 in vitro, similar to those with low serum IgM. In vitro response of B cells to IL-21 may predict clinically relevant humoral immune function in patients with IL2RG/JAK3 SCID after HSCT.


Subject(s)
B-Lymphocytes/immunology , Hematopoietic Stem Cell Transplantation/methods , Interleukin Receptor Common gamma Subunit/immunology , Interleukins/immunology , Janus Kinase 3/immunology , Severe Combined Immunodeficiency/therapy , Transplantation Conditioning/methods , Adolescent , B-Lymphocytes/cytology , Cell Differentiation , Child , Child, Preschool , Female , Humans , Immunity, Humoral , Interleukin Receptor Common gamma Subunit/genetics , Janus Kinase 3/genetics , Lymphocyte Activation , Male , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/immunology , Transplantation, Homologous , Young Adult
4.
Cancer Cell ; 9(5): 405-16, 2006 May.
Article in English | MEDLINE | ID: mdl-16697960

ABSTRACT

Our understanding of Ewing's sarcoma development mediated by the EWS/FLI fusion protein has been limited by a lack of knowledge regarding the tumor cell of origin. To circumvent this, we analyzed the function of EWS/FLI in Ewing's sarcoma itself. By combining retroviral-mediated RNA interference with reexpression studies, we show that ongoing EWS/FLI expression is required for the tumorigenic phenotype of Ewing's sarcoma. We used this system to define the full complement of EWS/FLI-regulated genes in Ewing's sarcoma. Functional analysis revealed that NKX2.2 is an EWS/FLI-regulated gene that is necessary for oncogenic transformation in this tumor. Thus, we developed a highly validated transcriptional profile for the EWS/FLI fusion protein and identified a critical target gene in Ewing's sarcoma development.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Genes, Neoplasm/genetics , Homeodomain Proteins/genetics , Oncogene Proteins, Fusion/genetics , Sarcoma, Ewing/genetics , Transcription Factors/genetics , Animals , Cell Transformation, Neoplastic , Homeobox Protein Nkx-2.2 , Humans , Mice , Mice, Nude , Microarray Analysis , Nuclear Proteins , Proto-Oncogene Protein c-fli-1 , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Protein EWS , Retroviridae/genetics , Transcription, Genetic , Zebrafish Proteins
5.
Blood Adv ; 8(12): 3001-3012, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38625984

ABSTRACT

ABSTRACT: Patients with Epstein-Barr virus (EBV)-positive posttransplant lymphoproliferative disease (EBV+ PTLD) in whom initial treatment fails have few options and historically low median overall survival (OS) of 0.7 months after allogeneic hematopoietic cell transplant (HCT) and 4.1 months after solid organ transplant (SOT). Tabelecleucel is an off-the-shelf, allogeneic EBV-specific cytotoxic T-lymphocyte immunotherapy for EBV+ PTLD. Previous single-center experience showed responses in patients with EBV+ PTLD after HCT or SOT. We now report outcomes from a multicenter expanded access protocol in HCT (n = 14) and SOT (n = 12) recipients treated with tabelecleucel for EBV+ PTLD that was relapsed/refractory (R/R) to rituximab with/without chemotherapy. The investigator-assessed objective response rate was 65.4% overall (including 38.5% with a complete and 26.9% with a partial response), 50.0% in HCT, and 83.3% in SOT. The estimated 1- and 2-year OS rates were both 70.0% (95% confidence interval [CI], 46.5-84.7) overall, both 61.5% (95% CI, 30.8-81.8) in HCT, and both 81.5% (95% CI, 43.5-95.1) in SOT (median follow-up: 8.2, 2.8, and 22.5 months, respectively). Patients responding to tabelecleucel had higher 1- and 2-year OS rates (94.1%) than nonresponders (0%). Treatment was well tolerated, with no reports of tumor flare, cytokine release syndrome, or rejection of marrow and SOT. Results demonstrate clinically meaningful outcomes across a broad population treated with tabelecleucel, indicating a potentially transformative and accessible treatment advance for R/R EBV+ PTLD after HCT or SOT. This trial was registered at www.ClinicalTrials.gov as #NCT02822495.


Subject(s)
Epstein-Barr Virus Infections , Hematopoietic Stem Cell Transplantation , Lymphoproliferative Disorders , Transplantation, Homologous , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/therapy , Female , Male , Adult , Middle Aged , Herpesvirus 4, Human , Organ Transplantation/adverse effects , Aged , Treatment Outcome , Young Adult
6.
Blood Adv ; 7(16): 4647-4657, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37603347

ABSTRACT

The majority of patients with chronic graft-versus-host disease (cGVHD) are steroid refractory (SR), creating a need for safe and effective therapies. Subcutaneous low-dose interleukin-2 (LD IL-2), which preferentially expands CD4+ regulatory T cells (Tregs), has been evaluated in 5 clinical trials at our center with partial responses (PR) in ∼50% of adults and 82% of children by week 8. We now report additional real-world experience with LD IL-2 in 15 children and young adults. We conducted a retrospective chart review of patients with SR-cGVHD at our center who received LD IL-2 from August 2016 to July 2022 not on a research trial. The median age at start of LD IL-2 was 10.4 years (range, 1.2-23.2 years) at a median of 234 days from cGVHD diagnosis (range, 11-542 days). Patients had a median of 2.5 (range, 1-3) active organs at LD IL-2 start and received a median of 3 (range, 1-5) prior therapies. The median duration of LD IL-2 therapy was 462 days (range, 8-1489 days). Most patients received 1 × 106 IU/m2 per day. There were no serious adverse effects. The overall response rate in 13 patients who received >4 weeks of therapy was 85% (complete response, n = 5; PR, n = 6) with responses in diverse organs. Most patients significantly weaned corticosteroids. Tregs preferentially expanded with a median peak fold increase of 2.8 in the ratio of Tregs to CD4+ conventional T cells (range, 2.0-19.8) by 8 weeks on therapy. LD IL-2 is a well-tolerated, steroid-sparing agent with a high response rate in children and young adults with SR-cGVHD.


Subject(s)
Graft vs Host Disease , Interleukin-2 , Child , Humans , Young Adult , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Immunotherapy , Interleukin-2/administration & dosage , Retrospective Studies , Infant , Child, Preschool , Adolescent
7.
Front Immunol ; 13: 954966, 2022.
Article in English | MEDLINE | ID: mdl-36189229

ABSTRACT

Chronic graft-versus-host disease (cGVHD) remains a frequent cause of non-relapse morbidity and mortality after allogeneic hematopoietic stem cell transplantation. In our single center trials of low-dose interleukin-2 (LD IL-2), the immunomodulatory properties of regulatory T cells (Tregs) have been harnessed to treat steroid-refractory cGVHD (SR-cGVHD) safely and effectively in adults and children. In these trials, 50-60% of patients showed clinical improvement of their cGVHD manifestations with partial responses at the primary response endpoint of 8-12 weeks. Many patients continued extended duration LD IL-2 therapy and achieved deeper clinical responses, including some complete responses. However, the durability of the clinical and immunologic improvement following IL-2 discontinuation has not been reported previously. We examined 20 adult and 2 pediatric patients who received extended duration LD IL-2 for a median of 103 weeks (range, 21-258) and had stable improvement or resolution of their cGVHD symptoms before discontinuing LD IL-2 therapy. The median follow-up after stopping IL-2 was 203 weeks (range 92-599). During this time, 16 patients (73%) were able to wean off all systemic immunosuppression without disease flare or progression. Among 13 patients with available immune cell data, the median fold change in absolute Treg count was 0.58 between 1 to 10 weeks after stopping IL-2 whereas CD4+ conventional T-cell (Tcon) and CD8+ T-cell numbers remained stable. Despite a decline in Treg numbers after IL-2 discontinuation, Treg numbers remained above the pre-treatment baseline. In addition, many patients had sustained clinical improvement after stopping IL-2, suggesting that extended IL-2 therapy can lead to immune tolerance.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Adult , Child , Chronic Disease , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Humans , Interleukin-2/therapeutic use , Steroids/therapeutic use
8.
Transplant Cell Ther ; 28(8): 502.e1-502.e12, 2022 08.
Article in English | MEDLINE | ID: mdl-35623615

ABSTRACT

Pulmonary toxicity after allogeneic hematopoietic stem cell transplantation (allo-HSCT) for childhood leukemia and myelodysplastic syndrome (MDS), along with the impact of different myeloablative conditioning regimens, remain incompletely described. Here we compared the acute and long-term incidence of pulmonary toxicity (PT) after total body irradiation (TBI)- and busulfan-based myeloablative conditioning. We conducted this retrospective cohort study of 311 consecutive pediatric patients with leukemia or MDS who underwent allo-HSCT at Dana-Farber Cancer Institute/Boston Children's Hospital between 2008 and 2018. PT was graded using Common Terminology Criteria for Adverse Events version 5.0. The primary objective was to compare the cumulative incidence of grade ≥3 and grade 5 PT after TBI-based and busulfan-based myeloablative conditioning using Gray's test. Secondary objectives were to determine factors associated with PT and overall survival (OS) using competing risk analysis and Cox regression analyses, respectively. There was no significant difference between the TBI-conditioned group (n = 227) and the busulfan-conditioned group (n = 84) in the incidence of grade ≥3 PT (29.2% versus 34.7% at 2 years; P = .26) or grade 5 pulmonary toxicity (6.2% versus 6.1% at 2 years; P = .47). Age (hazard ratio [HR], 1.70, 95% confidence interval [CI], 1.11 to 2.59; P = .01), grade ≥2 PT prior to allo-HSCT or preexisting pulmonary conditions (HR, 1.84, 95% CI, 1.24 to 2.72; P < .01), acute graft-versus-host disease (GVHD) (HR, 2.50; 95% CI, 1.51 to 4.14; P < .01), and chronic GVHD (HR, 2.61; 95% CI, 1.26 to 5.42; P = .01) were associated with grade ≥3 PT on multivariable analysis. Grade ≥3 PT was associated with worse OS (81.1% versus 61.5% at 2 years; P < .01). In pediatric allo-HSCT recipients, rates of PT were similar in recipients of TBI-based and recipients of busulfan-based myeloablative conditioning regimens. Age, the presence of PT or preexisting pulmonary conditions prior to transplantation, and the development of either acute or chronic GVHD were associated with grade ≥3 PT post-transplantation. Furthermore, the occurrence of grade 3-4 PT post-transplantation was associated with inferior OS.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Leukemia , Myelodysplastic Syndromes , Busulfan/adverse effects , Child , Graft vs Host Disease/epidemiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Leukemia/etiology , Myelodysplastic Syndromes/therapy , Retrospective Studies , Transplantation, Homologous , Treatment Outcome , Whole-Body Irradiation/adverse effects
9.
Blood Adv ; 6(21): 5786-5796, 2022 11 08.
Article in English | MEDLINE | ID: mdl-35475885

ABSTRACT

Chronic graft-versus-host disease (cGVHD) remains a frequent cause of nonrelapse morbidity and mortality after allogeneic hematopoietic stem cell transplantation. Despite recent advances, options for steroid-refractory (SR) cGVHD are limited. In previous trials of low-dose interleukin-2 (LD IL-2), the immunomodulatory properties of regulatory T cells (Tregs) have been harnessed to treat SR-cGVHD safely and effectively. In the present study, we combined a single infusion of Treg-enriched lymphocytes (Treg DLI) from the original stem cell donor with in vivo Treg expansion using LD IL-2 (1 × 106 IU/m2 per day for 8 weeks) in 25 adult patients with SR-cGVHD. Treg were not expanded ex vivo. Treg DLI was initiated at 0.1 × 106 cells per kg patient and escalated to a maximum dose of 1 × 106 cells per kg. Treg DLI plus LD IL-2 was well tolerated and led to partial responses (PR) in 5 of 25 patients (20%) after 8 weeks of therapy. Ten additional patients (40%) had stable disease with minor responses not meeting PR criteria. Patients at all dose levels had similar Treg expansion without significant changes in CD4+ conventional T cells or CD8+ T cells. High-throughput sequencing of the T-cell receptor ß locus showed selective improvement of Treg diversity. A subset of DLI-derived Treg clones showed preferential expansion at week 8 and long-term persistence 1-year postinfusion. We demonstrate for the first time that infusion of polyclonal healthy donor Tregs followed by expansion with LD IL-2 is safe in patients with SR-cGVHD, thus establishing a foundation for future adoptive Treg therapies in the posttransplant setting. This trial was registered at www.clinicaltrials.gov as #NCT01937468.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Adult , Humans , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Interleukin-2/therapeutic use , T-Lymphocytes, Regulatory , Hematopoietic Stem Cell Transplantation/adverse effects , Steroids/therapeutic use
10.
JCI Insight ; 7(7)2022 04 08.
Article in English | MEDLINE | ID: mdl-35239511

ABSTRACT

BACKGROUNDGut decontamination (GD) can decrease the incidence and severity of acute graft-versus-host disease (aGVHD) in murine models of allogeneic hematopoietic cell transplantation (HCT). In this pilot study, we examined the impact of GD on gut microbiome composition and the incidence of aGVHD in HCT patients.METHODSWe randomized 20 patients undergoing allogeneic HCT to receive (GD) or not receive (no-GD) oral vancomycin-polymyxin B from day -5 through neutrophil engraftment. We evaluated shotgun metagenomic sequencing of serial stool samples to compare the composition and diversity of the gut microbiome between study arms. We assessed clinical outcomes in the 2 arms and performed strain-specific analyses of pathogens that caused bloodstream infections (BSI).RESULTSThe 2 arms did not differ in the predefined primary outcome of Shannon diversity of the gut microbiome at 2 weeks post-HCT (genus, P = 0.8; species, P = 0.44) or aGVHD incidence (P = 0.58). Immune reconstitution of T cell and B cell subsets was similar between groups. Five patients in the no-GD arm had 8 BSI episodes versus 1 episode in the GD arm (P = 0.09). The BSI-causing pathogens were traceable to the gut in 7 of 8 BSI episodes in the no-GD arm, including Staphylococcus species.CONCLUSIONWhile GD did not differentially affect Shannon diversity or clinical outcomes, our findings suggest that GD may protect against gut-derived BSI in HCT patients by decreasing the prevalence or abundance of gut pathogens.TRIAL REGISTRATIONClinicalTrials.gov NCT02641236.FUNDINGNIH, Damon Runyon Cancer Research Foundation, V Foundation, Sloan Foundation, Emerson Collective, and Stanford Maternal & Child Health Research Institute.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Microbiota , Animals , Child , Decontamination , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Mice , Pilot Projects
11.
Trends Genet ; 24(6): 297-305, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18450316

ABSTRACT

The discovery of RNA interference (RNAi), the process of sequence-specific gene silencing initiated by double-stranded RNA (dsRNA), has broadened our understanding of gene regulation and has revolutionized methods for genetic analysis. A remarkable property of RNAi in the nematode Caenorhabditis elegans and in some other multicellular organisms is its systemic nature: silencing signals can cross cellular boundaries and spread between cells and tissues. Furthermore, C. elegans and some other organisms can also perform environmental RNAi: sequence-specific gene silencing in response to environmentally encountered dsRNA. This phenomenon has facilitated significant technological advances in diverse fields including functional genomics and agricultural pest control. Here, we describe the characterization and current understanding of environmental RNAi and discuss its potential applications.


Subject(s)
Caenorhabditis elegans/genetics , Environment , RNA Interference/physiology , Animals , Gene Silencing , Models, Biological , RNA Transport/physiology
12.
J Allergy Clin Immunol Pract ; 9(2): 753-759.e2, 2021 02.
Article in English | MEDLINE | ID: mdl-33096268

ABSTRACT

BACKGROUND: Iron uptake mediated by transferrin receptor 1 (TfR1), encoded by the TFRC gene, is essential for lymphocyte development and proliferation. Autosomal-recessive mutations in the human TFRC gene cause a combined immunodeficiency characterized by defective T- and B-cell proliferation as well as impaired class-switching. Clinical presentations have been severe in all reported cases, with symptoms including recurrent sinopulmonary infections, hypogammaglobulinemia, chronic diarrhea, and intermittent cytopenias. OBJECTIVE: To describe outcomes of allogeneic hematopoietic stem cell transplantation (HSCT) in patients with TFRC deficiency. METHODS: Retrospective chart review study of 5 patients with TFRC deficiency who underwent allogeneic HSCT between July 2011 and May 2018 at Boston Children's Hospital. RESULTS: Intermittent thrombocytopenia and neutropenia were a predominant feature of the clinical presentation in our cohort, and 3 patients who underwent bone marrow evaluation before HSCT were found to have signs of dysmyelopoiesis and dysplasia. One patient, who had a transplant at age 11 years, developed a clonal cytogenetic abnormality concerning for myelodysplastic syndrome. All 5 patients tolerated myeloablative conditioning regimens and had robust donor cell engraftment with resolution of cytopenias and independence from intravenous immunoglobulin substitution. All 5 patients were alive at a median follow-up of 47.1 months posttransplant (range, 15.7-85.4) and none had developed acute or chronic graft-versus-host disease. CONCLUSIONS: Allogeneic HSCT is curative for TFRC deficiency and rescues all known disease manifestations. Patients with TFRC deficiency may have a predisposition to malignant transformation of hematopoietic cells and may benefit from HSCT earlier in their disease course.


Subject(s)
Hematopoietic Stem Cell Transplantation , Receptors, Transferrin/deficiency , Transplantation Conditioning , Boston , Child , Humans , Receptors, Transferrin/genetics , Retrospective Studies , Transplantation, Homologous
13.
Expert Rev Hematol ; 13(2): 141-154, 2020 02.
Article in English | MEDLINE | ID: mdl-31874061

ABSTRACT

Introduction: Despite improvements in human leukocyte antigen (HLA) matching algorithms and supportive care, graft-versus-host disease (GVHD) remains the leading cause of non-relapse morbidity and mortality following allogeneic hematopoietic stem cell transplantation (HSCT). Acute GVHD, typically occurring in the first 100 days post-HSCT, is mediated by mature effector T cells from the donor (graft) that become activated after encountering alloantigens in the recipient (host). Chronic GVHD, characterized by aberrant immune responses to both autoantigens and alloantigens, occurs later and arises from a failure to develop tolerance after HSCT. CD4+ CD25+ CD127- FOXP3+ regulatory T cells (Tregs) function to suppress auto- and alloreactive immune responses and are key mediators of immune tolerance.Areas covered: In this review, authors discuss the biologic and therapeutic roles of Tregs in acute and chronic GVHD, including in vivo and ex vivo strategies for Treg expansion and adoptive Treg cellular therapy.Expert opinion: Although they comprise only a small subset of circulating CD4 + T cells, Tregs play an important role in establishing and maintaining immune tolerance following allogeneic HSCT. The development of GVHD has been associated with reduced Treg frequency or numbers. Consequently, the immunosuppressive properties of Tregs are being harnessed in clinical trials for GVHD prevention and treatment.


Subject(s)
Graft vs Host Disease , HLA Antigens/immunology , Hematopoietic Stem Cells , T-Lymphocytes, Regulatory , Allografts , Chronic Disease , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Humans , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
14.
Blood Adv ; 3(7): 984-994, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30936059

ABSTRACT

Patients with chronic graft-versus-host disease (cGVHD) have a paucity of regulatory CD4 T cells (CD4Tregs) that mediate peripheral tolerance. In clinical trials, daily low-dose interleukin-2 (IL-2) has been administered safely for prolonged periods in patients with steroid-refractory cGVHD. Peripheral CD4Tregs expand dramatically in all patients during IL-2 therapy but clinical improvement was observed in ∼50% of patients. Here, we examined the impact of low-dose IL-2 therapy on functional T-cell markers and the T-cell repertoire within CD4Tregs, conventional CD4 T cells (CD4Tcons), and CD8+ T cells. IL-2 had profound effects on CD4Tregs homeostasis in both response groups including selective expansion of the naive subset, improved thymic output, and increased expression of Ki67, FOXP3, and B-cell lymphoma 2 within CD4Tregs. Similar changes were not seen in CD4Tcons or CD8 T cells. Functionally, low-dose IL-2 enhanced, in vitro, CD4Treg-suppressive activity in both response groups, and all patient CD4Tcons were similarly suppressed by healthy donor CD4Tregs. High-throughput sequencing of the T-cell receptor ß (TCRß) locus demonstrated that low-dose IL-2 therapy increased TCR repertoire diversity and decreased evenness within CD4Tregs without affecting CD4Tcons or CD8 T cells. Using clone-tracking analysis, we observed rapid turnover of highly prevalent clones in CD4Tregs as well as the conversion of CD4Tcons to CD4Tregs. After 12 weeks of daily IL-2, clinical responders had a greater influx of novel clones within the CD4Treg compartment compared with nonresponders. Further studies to define the function and specificity of these novel CD4Treg clones may help establish the mechanisms whereby low-dose IL-2 therapy promotes immune tolerance.


Subject(s)
Drug Resistance , Graft vs Host Disease/drug therapy , Interleukin-2/administration & dosage , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Proliferation , Chronic Disease , Female , Genetic Variation , Graft vs Host Disease/immunology , Humans , Immune Tolerance/drug effects , Interleukin-2/pharmacology , Lymphocyte Count , Male , Middle Aged , Receptors, Antigen, T-Cell/genetics , Steroids/pharmacology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects
15.
Blood Adv ; 3(17): 2550-2561, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31471324

ABSTRACT

Low-dose interleukin-2 (IL-2) therapy for chronic graft-versus-host disease (cGVHD) generates a rapid rise in plasma IL-2 levels and CD4+CD25+CD127-Foxp3+ regulatory T-cell (CD4Treg) proliferation, but both decrease over time despite continued daily administration. To test whether IL-2 dose escalation at the time of anticipated falls in plasma levels could circumvent tachyphylaxis and enhance CD4Treg expansion, we conducted a phase 1 trial in 10 adult and 11 pediatric patients with steroid-refractory cGVHD (www.clinicaltrials.gov: NCT02318082). Daily IL-2 was initiated in children and adults (0.33 × 106 and 0.67 × 106 IU/m2 per day, respectively). Dose escalations were scheduled at weeks 2 and 4 to a maximum dose of 1 × 106 IU/m2 per day in children and 2 × 106 IU/m2 per day in adults. Patients continued at their maximum tolerated dose (MTD) until week 8. Children tolerated IL-2 dose escalation with partial responses (PRs) in 9 of 11 patients (82%) at multiple cGVHD sites, including lung. Patient-reported outcome scores for skin and lung improved significantly in pediatric patients. In contrast, 5 of 10 adults required dose reduction, and only 2 of 7 evaluable patients (29%) had PRs at week 8. CD4Tregs and natural killer cells expanded in both cohorts without significant changes in conventional CD4+ T cells (Tcons) or CD8+ T cells. Children achieved a higher median CD4Treg/Tcon ratio at week 8 (0.4 vs 0.18, P = .02) despite lower IL-2 doses. We show for the first time that low-dose IL-2 is safe and effective in children with advanced cGVHD. In adults, escalation above the previously defined MTD did not improve CD4Treg expansion or clinical response.


Subject(s)
Graft vs Host Disease/drug therapy , Interleukin-2/administration & dosage , Adult , Cell Proliferation/drug effects , Child , Chronic Disease , Female , Graft vs Host Disease/pathology , Humans , Immunotherapy/methods , Interleukin-2/pharmacology , Killer Cells, Natural/cytology , Lung Diseases/drug therapy , Male , Maximum Tolerated Dose , Skin Diseases/drug therapy , T-Lymphocytes, Regulatory/cytology
16.
G3 (Bethesda) ; 7(12): 3887-3899, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29025917

ABSTRACT

In the nematode Caenorhabditis elegans, RNA interference (RNAi) triggered by double-stranded RNA (dsRNA) spreads systemically to cause gene silencing throughout the organism and its progeny. We confirm that Caenorhabditis nematode SID-1 orthologs have dsRNA transport activity and demonstrate that the SID-1 paralog CHUP-1 does not transport dsRNA. Sequence comparison of these similar proteins, in conjunction with analysis of loss-of-function missense alleles, identifies several conserved 2-7 amino acid microdomains within the extracellular domain (ECD) that are important for dsRNA transport. Among these missense alleles, we identify and characterize a sid-1 allele, qt95, which causes tissue-specific silencing defects most easily explained as a systemic RNAi export defect. However, we conclude from genetic and biochemical analyses that sid-1(qt95) disrupts only import, and speculate that the apparent export defect is caused by the cumulative effect of sequentially impaired dsRNA import steps. Thus, consistent with previous studies, we fail to detect a requirement for sid-1 in dsRNA export, but demonstrate for the first time that SID-1 functions in the intestine to support environmental RNAi (eRNAi).


Subject(s)
Caenorhabditis elegans Proteins/genetics , Membrane Proteins/genetics , RNA Transport/genetics , RNA, Double-Stranded/genetics , Animals , Animals, Genetically Modified/genetics , Caenorhabditis elegans/genetics , Gene Silencing , Intestinal Mucosa/metabolism , RNA Interference
SELECTION OF CITATIONS
SEARCH DETAIL