Bruce R. Blazar’s research while affiliated with University of Minnesota and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (832)


Single-Chain Nanobody Inhibition of Notch and Avidity Enhancement Utilizing the β-Pore-Forming Toxin Aerolysin
  • Article

March 2025

·

1 Read

ACS Chemical Biology

Andrew C D Lemmex

·

Jeremy Allred

·

Jason Ostergard

·

[...]

·


Costimulatory blockade dampens Tfr cell differentiation after allogeneic kidney transplantation
a Schematic of kidney transplantation. Balb/c (allogeneic) or C57BL/6 (syngeneic) underwent kidney transplantation into FoxP3 reporter mice. Recipients were harvested 20 days post-transplantation to assess immune response and graft rejection. b The levels of DSA IgG in the serum were measured by flow cytometry and presented as MFI (n = 4 for the Syn and POD20 group, n = 5 for the POD10 group). POD: post-operative day. MFI: mean fluorescence intensity. c Representative histological images of kidney grafts at postoperative day 20. Magnification: 200×, scale bars: 50 μm. d. Gating strategy (left) and quantification (right) of follicular CXCR5⁺ T, Tfh (gated as CD4⁺CD19⁻CXCR5⁺FoxP3⁻), and Tfr (gated as CD4⁺CD19⁻CXCR5⁺FoxP3⁺) cells in the spleen and dLN. n = 4 for the Syn and POD20 group, n = 5 for the POD10 group. e Frequency of CD4⁺FoxP3⁺ Treg cells in total CD4+ T cells in the spleen and dLN (n=4 for the Syn and POD20 group, n = 5 for the POD10 group). f Gating strategy (left) was used to identify and quantify (right) CD19⁺GL7⁺FAS⁺ germinal center (GC) B cells from dLNs. n = 4 for the Syn and POD20 group, n = 5 for the POD10 group. g Schematic of CTLA4Ig treatment after kidney transplantation. h Representative histological images of transplanted kidneys from CTLA4Ig treated mice are shown, stained with hematoxylin and eosin (HE) (Magnification: 200×, scale bars: 50 μm) and ABMR marker C4d (Magnification: 100×, scale bars: 100 μm). i IgG DSA levels at indicated time points from the experiment as in (g). n = 4 mice per group. Student’s two-tailed unpaired T-test was used to compare two groups. Bonferroni correction was used to control for multiple comparisons. j Gating strategy (left) and frequency (right) of GC B cells from dLNs with (n = 4) or without CTLA4Ig treatment (n = 5). k Gating strategy (left) and quantification (right) of follicular CXCR5⁺ T, Tfh (gated as CD4⁺CD19⁻CXCR5⁺FoxP3⁻), and Tfr (gated as CD4⁺CD19⁻CXCR5⁺FoxP3⁺) cells from dLNs (n = 5 for the No treatment and n = 4 for the CTLA4Ig group). Data is from one experiment and is representative of two independent experiments. Error bars show mean ± SEM. Student’s two-tailed unpaired T test (j, k), One-way ANOVA with Tukey’s multiple-comparison test (b, d–f). Source data are provided as a Source Data file. Portions created in BioRender. Zhang, H. (2025) https://BioRender.com/b29q775.
The follicular to GC-like developmental transition in Tfr cells is sensitive to costimulatory blockade
a Diagram of single-cell RNA sequencing experiment. Balb/c kidneys were transplanted into Foxp3IRES-GFP mice with or without CTLA4Ig treatment. Tfr (CD4⁺CXCR5⁺FoxP3⁺) and Treg (CD4⁺CXCR5⁻FoxP3⁺) cells from dLN were sorted and scRNAseq performed. b UMAP plot showing unsupervised clustering of all post-filter cells. The total number of cells analyzed is shown. c Cells in UMAP plot marked by cell type. d Feature plots showing the expression levels of indicated genes of interest across clusters. e Clusters annotated based on gene expression states, including Naïve-like, Follicular-like, and GC-like. f Monocle pseudotime analysis utilizing cluster 0 as the starting node and indicating developmental trajectories by lines and overall pseudotime by color, plotted within UMAP space. g. UMAP plot showing clustering (top) and cell type (bottom) for cells in no treatment or CTLA4Ig treated mice. Number of cells per group is indicated. h Cluster distribution changes analyzed with miloR package. Red dots indicate a reduced cell population while blue dots indicate an increased cell population with the CTLA4Ig treatment. i Heatmap showing top marker genes for each cluster compared to all clusters. j Volcano plot showing differentially expressed genes (DEGs) between clusters 3 and 4. DEGs were identified using a two-sided Wilcoxon rank-sum test, as implemented by the Seurat FindMarkers function (logFC threshold=0.1, min.pct=0.1). p-values were adjusted for multiple comparisons using the Bonferroni method. k Venn diagram showing differentially expressed genes between Tfr and Treg cells in control or CTLA4Ig treated group. Portions created in BioRender. Zhang, H. (2025) https://BioRender.com/b29q775.
Costimulatory blockade restricts Tfr clonal expansion
a Clonal expansion indicated in UMAP space utilizing scRepertoire. Each dot represents an individual cell, with colors indicating the size of clonal expansion. Pink indicates singleton clones, while dark pink and red indicate small (1<x < =5) and medium (5<x < =20) clonal expansion, respectively. b Distribution of clonal expansion across experiment groups (left) and clusters (right). c Correlation of clonal expansion and sharing of clones between Tfr and Treg cells calculated by scRepertoire. Circled clones indicate shared clones. d Proportion of clonal sharing between cluster 0 and other clusters, with cluster 0 set as founder clones. The thickness of lines indicates magnitude of shared clones. e Circos plot showing sharing of expanded clones between clusters. Only expanded clones are included.
Tfr cells prevent allo-antibody formation and rejection of kidney allografts
a Schematic of Tfr cell deletion using Tfr-DTR mice. Control (Tfr-Con, Foxp3CreYFPCxcr5wt) or Tfr-DTR (Foxp3CreYFPCxcr5LoxSTOPLoxDTR) mice received diphtheria toxin (DT), CTLA4Ig (on day 2) and Balb/c kidneys as life-sustaining (b) or non-life-sustaining (c–g) transplants. b Survival of transplanted recipients not receiving CTLA4Ig (no-treatment), or CTLA4Ig treated Tfr-Con or Tfr-DTR mice. MST, median survival time. c Spleens and kidney grafts from Tfr-Con and Tfr-DTR mice harvested 20 days after transplantation. d Representative histological images of kidney grafts at postoperative day 20, including hematoxylin and eosin (HE), immunohistochemistry staining for B220, and immunofluorescence staining for C4d and IgG. scale bars: 50 and 100 μm. e IgG DSA levels in serum at indicated time points. MFI=mean fluorescence intensity. n = 5 mice per group. f Allo-reactive IgG to individual MHC molecules measured by single antigen bead. Syn= syngeneic. n = 5 mice per group. g Quantification of serum autoantibody levels. Representative images of serum from mice as in (a) incubated with Hep-2 cells (left) and quantification of cumulative, cytoplasm, and nucleus antibody signal. n = 5 mice per group. Scale bars: 50 μm. Data is from one experiment and is representative of two independent experiments. Statistics: Error bars show mean ± SEM. Student’s two-tailed unpaired T test for 2-group comparisons and Kaplan-Meier survival analysis and a log-rank test for survival analysis (b). Source data are provided as a Source Data file. Portions created in BioRender. Zhang, H. (2025) https://BioRender.com/b29q775.
Tfr cells restrain lymph node and intragraft germinal center B cell differentiation
a Schematic of Tfr cell deletion using Tfr-DTR mice. Tfr-Con (Foxp3CreYFPCxcr5wt) or Tfr-DTR (Foxp3CreYFPCxcr5LoxSTOPLoxDTR) mice received diphtheria toxin (DT), CTLA4Ig (on day 2) and Balb/c kidneys. b Gating strategy and quantification of CXCR5⁺ follicular T, Tfh (gated as CD4⁺CD19⁻CXCR5⁺FoxP3⁻), Tfr (gated as CD4⁺CD19⁻CXCR5⁺FoxP3⁺), and CXCR5⁻FoxP3⁺ T cells in total CD4⁺ T cells from the dLNs. n = 4 mice for the Tfr-Con group and n = 5 for the Tfr-DTR group. c Gating strategy (left) and quantification (right) of CD19⁺GL7⁺FAS⁺ GC B cells. n = 4 mice for the Tfr-Con group and n = 5 for the Tfr-DTR group. d. Gating strategy (left) and quantification (right) of Naïve B cells (CD38⁺IgG1⁻CD19⁺), memory-like B cells (CD38⁺IgG1⁺CD19⁺), and IgG1⁺ B cells in the dLN (n = 4 for Tfr-Con and n = 5 for Tfr-DTR). e Gating strategy and quantification of Tfh and Tfr cells in kidney allografts (n = 4 for Tfr-Con and n = 5 for Tfr-DTR). f Gating strategy and quantification of GC-like B cells (GL7⁺CD19⁺) in kidney allografts. n = 4 mice for each group. g Schematic of GC B single cell culture assays. Single GC B cells from dLN or grafts were sorted and cultured with NB21 feeder cells for 6 days. Culture supernatants were pre-screened for IgG positivity and further assessed for DSA reactivity. h DSA signal of individual IgG⁺ clones from LN GC B and graft B cells from either control or Tfr-DTR mice 20 days after transplantation. The dotted line indicates the level of detection threshold assessed by signal using syngeneic cells. i Frequency of DSA clones (from all IgG⁺ clones). Numbers indicate the total number of IgG clones analyzed. Data is from one experiment and is representative of two independent experiments. Statistics: Error bars show mean ± SEM. Student’s two-tailed unpaired T test for 2-group comparisons. Source data are provided as a Source Data file. Portions created in BioRender. Zhang, H. (2025) https://BioRender.com/b29q775.

+3

Follicular regulatory T cells restrain kidney allograft rejection in mice by suppressing alloreactive B cells
  • Article
  • Full-text available

March 2025

·

26 Reads

Pathogenic antibodies produced by alloreactive B cells mediate antibody-mediated rejection after kidney transplantation, but the mechanisms remain poorly understood. Follicular regulatory T (Tfr) cells modulate follicular helper T cell-mediated B cell responses, but the functions of Tfr in controlling alloreactive antibody are unknown. Here we study the developmental signals and functions of Tfr cells in mouse allogeneic kidney transplantation models, and show that costimulatory blockade alters the development of Tfr cells disproportionately by decreasing germinal center (GC)-like Tfr cells but increasing follicular-like Tfr cells. Functionally, global Tfr cell deletion results in accelerated graft rejection and increases in donor-specific B cells in both draining lymph nodes and kidney allografts. Mechanistically, Tfr cell deletion increases GC B cell expression of pro-inflammatory cytokines such as IL-15, while neutralization of IL-15 compensates for the loss of Tfr cells and prolongs the survival of mice receiving kidney transplants. Together our preclinical mouse data demonstrate how Tfr restrains kidney allograft rejection by limiting alloreactive B cell responses.

Download

Defining pathogenic IL-17 and CSF-1 gene expression signatures in chronic graft-versus-host disease

February 2025

·

6 Reads

Blood

Chronic graft-versus-host disease (cGVHD) remains the leading cause of non-relapse morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT). Effective therapeutics agents targeting dysregulated cytokines including IL-17 and CSF-1 have been defined in preclinical models of cGVHD, and efficacy in subsequent clinical trials has led to their recent FDA approval. Despite this, these agents are effective in only a subset of patients, expensive, difficult to access outside the US, and used in a trial-and-error fashion. The ability to readily discern druggable, dysregulated immunity in these patients is desperately needed to facilitate the selection of appropriate treatment and to potentially identify high-risk individuals for preemptive therapy. We used single cell sequencing-based approaches in our informative preclinical cGVHD models to "reverse engineer" temporal IL-17 and CSF-1 signatures in mouse blood that could be used to interrogate patients. We defined distinct, non-intuitive IL-17 and CSF-1 signatures in mouse blood monocytes that could be identified in relevant monocytes within 70% of patients at diagnosis of cGVHD and in half of patients at day +100 post-HCT who subsequently developed cGVHD. These signatures can now be evaluated prospectively in clinical studies to help delineate potential responder and non-responders to relevant therapeutics targeting these pathways.


Chronic Graft-versus-host Disease, Part 2: Clinical Success and Roadmap to the Future

February 2025

·

25 Reads

Transplantation

Chronic graft-versus-host disease (cGVHD) is an immune-mediated, heterogeneous, multiorgan complication affecting allogeneic hematopoietic cell transplantation recipients, leading to increased morbidity, mortality, and decline in health-related quality-of-life. Advances in understanding the complex disease pathophysiology, and collaborative efforts lead by the National Institutes of Health to standardize criteria for clinical trials, led to bench-to-bedside efforts resulting in the development of 4 US Food and Drug Administration-approved agents for the treatment steroids-refractory cGVHD since 2017. Despite the remarkable advances in the field of hematopoietic cell transplantation in prevention of cGVHD, and more treatment options, the outcome of patients with moderate-severe cGVHD remains suboptimal. Essential to successful cGVHD management is to recognize the disease at early stages before the onset of irreversible damage, allowing for personalized multidisciplinary specialized interventions that include pharmacologic therapies and additional supportive care measures. The aim of this review is to summarize key areas of active clinical research and new developments in cGVHD therapeutic approaches, with focus on (1) preemptive therapy, (2) upfront therapy beyond corticosteroids, (3) treatment refractory cGVHD novel agents, role of combination therapies, and organ-specific approaches, and (4) challenges, gaps, and future directions.


Targeting cell surface VISTA expression on allospecific naïve T cells promotes tolerance

February 2025

·

9 Reads

Blood

The success of allogeneic hematopoietic stem cell transplantation (allo-HSCT) can be limited by graft-versus-host disease (GVHD). T-cell activation is a key factor in GVHD progression. Costimulatory signals can be counterbalanced by co-inhibitory signals such as the checkpoint molecule VISTA (V-domain Ig-containing suppressor of T-cell activation)/PD-1H that restrains activation and maintains donor T-cell quiescence. A single dose of anti-VISTA mAb prevents acute GVHD lethality in multiple models. Naïve donor T-cells express moderate VISTA levels, which transiently increase in allo-HSCT recipients in association with TCR signaling, leading to heightened susceptibility to anti-VISTA mAb-mediated depletion, in contrast to donor T-cells transferred to syngeneic recipients. Anti-VISTA mAb donor T-cell depletion was compatible with rapamycin but incompatible with peri-transplant tacrolimus GVHD prophylaxis. Targeting VISTA exclusively on host cells or donor CD8+ T-cells was not protective against GVHD lethality. Instead, anti-VISTA mAb mediated deletion of alloreactive donor T-cells depended on targeting a third (non-T) cell type. Further mechanistic studies indicated that donor T-cells concurrently exposed to anti-VISTA mAb in vivo but not preincubated in vitro before adoptive T-cell transfer were eliminated via FcR-mediated phagocytosis. In a lymphoma challenge model, a graft-versus-lymphoma effect was fully retained when anti-human VISTA mAb exclusively targeted donor CD4+ T-cells and delayed but mostly retained when unseparated donor T-cells were infused. In a xenogeneic GVHD model, anti-human VISTA mAb reduced donor T-cell expansion, VISTA T-cell expression levels, and recipient lethality. Together, these data support a novel clinical translational pathway in which acute GVHD lethality can be mitigated without negating the GVL effect.


Activation-induced thrombospondin-4 works with thrombospondin-1 to build cytotoxic supramolecular attack particles

February 2025

·

32 Reads

Proceedings of the National Academy of Sciences

Cytotoxic attack particles released by CTLs and NK cells include diverse phospholipid membrane and glycoprotein encapsulated entities that contribute to target cell killing. Supramolecular attack particles (SMAPs) are one type of particle characterized by a cytotoxic core enriched in granzymes and perforin surrounded by a proteinaceous shell including thrombospondin (TSP)-1. TSP-4 was also detected in bulk analysis of SMAPs released by CTLs; however, it has not been investigated whether TSP-4 contributes to distinct SMAP types or the same SMAP type as TSP-1 and, if in the same type of SMAP, whether TSP-4 and TSP-1 cooperate or compete. Here, we observed that TSP-4 expression increased upon CD8 ⁺ T cell activation while, surprisingly, TSP-1 was down-regulated. Correlative Light and Electron Microscopy and Stimulated Emission Depletion microscopy localized TSP-4 and TSP-1 in SMAP-containing multicore granules. Superresolution dSTORM revealed that TSP-4 and TSP-1 are usually enriched in the same SMAPs while particles with single-positive shells are rare. Retention Using Selective Hooks assays showed that TSP-4 localizes to the lytic granules faster than TSP-1 and promotes its accumulation therein. TSP-4 contributed to direct CTL-mediated killing, as previously shown for TSP-1. TSP-4 and TSP-1 were both required for latent SMAP-mediated cell killing, in which released SMAPs kill targets after removal of the CTLs. Of note, we found that chronic lymphocytic leukemia (CLL) cell culture supernatants suppressed expression of TSP-4 in CTL and latent SMAP-mediated killing. These results identify TSP-4 as a functionally important component of SMAPs and suggest that SMAPs may be targeted for immune suppression by CLL.


Deficiency of T follicular helper cell Tet3 DNA demethylation inhibits pathogenic IgG2c class switching and chronic GVHD

February 2025

·

15 Reads

Blood

Chronic graft-versus-host disease (cGVHD) is the leading cause of morbidity and non-relapse associated mortality following allogeneic hematopoietic cell transplantation (aHSCT). Treating steroid resistant/refractory cGVHD remains challenging. Epigenetic regulators can have global transcriptional effects that control donor T-cell responses. We previously showed that inhibiting histone lysine motifs by chromatin-modifying enzymes can ameliorate murine cGVHD. Targeting donor T-cell DNA methyltransferases reduce acute GVHD. Here, we sought to investigate the DNA demethylase Tet (ten-eleven translocase) methylcytosine dioxygenases 2 (Tet2) and Tet3 in T follicular helper cell (TFH) dependent cGVHD. In a clinically relevant model of cGVHD that recapitulates pulmonary fibrosis from bronchiolitis obliterans, recipients of Tet2 deleted donor T-cells did not have improved pulmonary function tests in contrast to the markedly improved pulmonary function in Tet3 deleted donor T-cells. Tet3 deleted donor T-cells did not impair TFH-dependent germinal center (GC) formation. Unexpectedly, TET3 deficiency resulted in elevated GATA3 expression in and IL-4 production by TFH cells. TET3 deficient TFH cells supported GC B-cell immunoglobulin (Ig) class switching to nonpathogenic IgG1 but not pathogenic IgG2c allowing mice to escape cGVHD pulmonary fibrosis. Elevated GATA3 expression and disruption of IgG2c class switching was recapitulated in an in-vitro human GC culture system. These studies provide new insights into the function of Tet3 in TFH driven Ig class switching and suggest a new approach to mitigate cGVHD.



Organ-specific microenvironments drive divergent T cell evolution in acute graft-versus-host disease

January 2025

·

23 Reads

Science Translational Medicine

Tissue-specific T cell immune responses play a critical role in maintaining organ health but can also drive immune pathology during both autoimmunity and alloimmunity. The mechanisms controlling intratissue T cell programming remain unclear. Here, we leveraged a nonhuman primate model of acute graft-versus-host disease (aGVHD) after allogeneic hematopoietic stem cell transplantation to probe the biological underpinnings of tissue-specific alloimmune disease using a comprehensive systems immunology approach including multiparameter flow cytometry, population-based transcriptional profiling, and multiplexed single-cell RNA sequencing and TCR sequencing. Transcriptional profiling revealed substantial biological differences between T cells infiltrating the lung and liver during aGVHD. These included enrichment for transcriptional pathways controlling extracellular matrix remodeling and chemotaxis in the lung and enrichment for transcriptional pathways linked to nucleic acid metabolism and proliferation in the liver. Single-cell RNA sequencing and TCR sequencing substantiated divergent organ-specific transcriptional programing of tissue-infiltrating T cells, which was linked to clonal expansion, with expanded clones progressively enriched for C-X3-C motif chemokine receptor 1 ( CX3CR1 )–expressing CD8 effector T cells in the lung and eomesodermin ( EOMES )–expressing CD8 effector-memory T cells in the liver. This divergent evolution of T cells was maintained even for T cells sharing the same TCRs, indicating its independence from antigen specificity. Together, these results provide insights into the role that tissue microenvironment–derived signals play in local T cell transcriptional programming during alloimmune-mediated clonal expansion and suggest potential opportunities to develop tissue-specific therapeutics to curtail pathogenic immunity after transplant.


Five-Year Outcomes of the “Abatacept Combined with a Calcineurin Inhibitor and Methotrexate for Graft Versus Host Disease (GVHD) Prophylaxis: A Randomized Controlled Trial” (‘ABA2‘)

November 2024

·

12 Reads

Blood

Background: GVHD remains a major cause of non-relapse mortality (NRM) after hematopoietic cell transplant (HCT). Abatacept, a costimulatory blockade agent targeting the CD28:CD80/86 pathway, was FDA-approved in 2021 for GVHD prophylaxis in patients undergoing an 8/8 HLA-matched unrelated donor (MUD), or 7/8-HLA mismatched unrelated donor (MMUD) HCT for a hematologic malignancy, in part based on the ABA2 trial (NCT01743131). Here, we present 5-yr follow-up data for ABA2. Methods: ABA2 patient eligibility criteria (described in detail in PMID: 33449816) included patients ≥ 6 yr with heme malignancies undergoing MUD or MMUD HCT after intensive conditioning. MUD patients were enrolled on the randomized, double-blind placebo-controlled arm, with patients randomized to either 4 doses of abatacept (10mg/kg/dose on Day -1, +5, +14 +28) or placebo. MMUD patients all received 4 doses of abatacept. 185 patients were enrolled: 69 MUD-placebo, 73 MUD-abatacept, 43 MMUD-abatacept. GVHD prophylaxis also included a calcineurin inhibitor (CNI, continued through Day +100 followed by a wean) and methotrexate (MTX, 15mg/m2 on day +1 and 10mg/m2 on Day +3, +6, +11). Both bone marrow and PBSC grafts were used. All patient data were independently monitored through 5 yr by the NMDP CRO. Here we report cumulative incidence (CI) data at 2-, 3- and 5-yr for overall survival (OS), relapse, relapse-free survival (RFS), non-relapse mortality (NRM), systemic therapy (ST)-requiring chronic GVHD (cGVHD), and grade 3-4 acute GVHD (aGVHD)-free/ST-requiring cGVHD-free/relapse-free survival (GRFS). Results: The median follow-up was 53.8 months, 56.1 months and 60 months for the MUD-placebo, MUD-abatacept, and MMUD-abatacept cohorts, respectively. The 2-, 3- and 5-yr OS was 65.2%, 62.3% and 54.3% for MUD-placebo, 75.2%, 68.0% and 61.0% for MUD-abatacept, and 78.8%, 74.0% and 71.7% for MMUD-abatacept cohorts, respectively. The 2-, 3- and 5-yr CI of relapse was 25.7%, 27.5% and 27.5% for MUD-placebo, 21.7%, 25.0% and 25.0% for MUD-abatacept, and 9.7%, 9.7% and 12.6% for MMUD-abatacept cohorts, respectively. The 2-, 3- and 5-yr CI of NRM was 18.1%, 20.0%, 28.7% for MUD-placebo, 14.4%, 18.1% and 22.5% for MUD-abatacept, and 15.0%, 20.2% and 20.2% for MMUD-abatacept cohorts, respectively. The 2-, 3- and 5-yr CI of RFS was 60.9%, 58.0% and 51.7% for MUD-placebo, 67.0%, 61.4% and 58.1% for MUD-abatacept, and 76.7%, 72.1% and 69.8% for MMUD-abatacept cohorts, respectively. The 1-yr CI of grade 3-4 acute GVHD was 14.8% for MUD-placebo, 8.4% for MUD-abatacept, and 2.3% for MMUD-abatacept cohorts, respectively. The 2-, 3- and 5-yr CI of ST-requiring chronic GVHD was 49.6%, 52.0% and 52.0% for MUD-placebo, 54.1%, 58.5% and 58.5% for MUD-abatacept, and 72.5%, 75.2% and 78.0% for MMUD-abatacept cohorts, respectively. The 2-, 3- and 5-yr cumulative incidence of GRFS was 27.5%, 26.1%, and 26.1% for MUD-placebo, 27.4%, 21.9% and 21.9% for MUD-abatacept, and 23.3%, 20.9% and 16.3% for MMUD-abatacept cohorts, respectively. Conclusions: Here we demonstrate that the central outcomes previously reported for ABA2 at 2 yr post-HCT (PMID: 33449816) were durable at 3- and 5-yr, with relatively stable OS, relapse, NRM, cGVHD and RFS. These data are important given new CIBMTR comparisons demonstrating that, for both MUD and MMUD recipients at 2 yr, abatacept/CNI/MTX is superior for OS and RFS compared to both CNI/MTX and CNI/MTX/ATG, and is similar to post-transplant cyclophosphamide (PT-Cy, Blood 2024, PMID:39028876). Moreover, the 3-yr outcome data for MMUD patients presented here for ABA2 also appear similar to the recently reported 3-yr outcomes for myeloablative conditioning (MAC) and PT-Cy for the MMUD-15 trial (PMID: 36584941, 5-yr outcomes not yet reported for MMUD-15). Most notable is the fact that despite increased cGVHD in ABA2 compared to MMUD-15, the GRFS is similar in both trials (20.9% GRFS at 3 yr for ABA2 versus 16.9% for PT-Cy after MAC HCT). These similar composite endpoints may be driven by lower apparent relapse rates in ABA2 (9.7% at 3-yr for ABA2 and 50.5% at 3-yr for MAC PT-Cy in MMUD-15). Together, these data lead to two key conclusions: (1) That the addition of abatacept continues to abrogate historic disparities in OS and RFS for HLA-mismatched vs HLA-matched HCT (extending our previous comparison (PMID:34753172) to 5-yrs post-HCT). (2) That a head-to-head randomized trial of abatacept- versus PT-Cy-based prophylaxis is warranted.


Citations (44)


... d Tumour-induced immune suppression is mediated by factors such as TGFβ, IL-10, and IDO, which directly inhibit T cells and promote Treg development. e Tumour cells also establish privileged sites by creating physical barriers restricting immune system access(Taylor and Balko 2022;Rui et al. 2024;Banushi et al. 2023;Li et al. 2023;Scott et al. 2021) ...

Reference:

Current advancement of immune function paradox of tumour-infiltrating cells and their immunotherapeutic targets: a mini-review
Human OX40L–CAR-T regs target activated antigen-presenting cells and control T cell alloreactivity
  • Citing Article
  • October 2024

Science Translational Medicine

... This shift is critical for controlling excessive inflammation and promoting immune tolerance. Lactate also enhances Treg cell function and generation (102). One primary pathway is the modulation of MOESIN lactylation, which facilitates interaction with the transforming growth factor beta (TGF-b) receptor, enhancing TGF-b signaling and SMAD3 activation. ...

Lactate supports Treg function and immune balance via MGAT1 effects on N-glycosylation in the mitochondria

The Journal of clinical investigation

... Further, nonrelapse mortality (NRM) is high in patients with steroidrefractory aGVHD, for example it was reported at 49% at 18 months among patients randomized in the ruxolitinib arm of the REACH2 trial [2]. CGVHD is also a serious complication of alloHCT, typically occurring between 100 days and 2 years after transplantation and affecting around 50% of patients sometimes for many years [3]. Therefore, it is critical to further improve GVHD prophylaxis and therapy. ...

Chronic graft-versus-host disease: Unresolved complication or ancient history?
  • Citing Article
  • July 2024

Blood

... Interpretation was heavily influenced by single-cell RNA-seq (scRNA-seq) analyses on day 14 with subsequent protein analyses, performed principally on CD8 + 2C TCR-Tg T cells. In contrast, our findings were primarily in alloreactive CD4 + T cells, with validation of expansion kinetics using small numbers of H-Yspecific CD4 + TCR-Tg T cells (2). We noted preferential CD4 + central memory T cell (Tcm) generation in both polyclonal and HY-TCR-Tg T cells in the presence of a calcineurin inhibitor (CNI) and importantly, this expansion was dose and agent (tacrolimus > cyclosporin) dependent (2). ...

Calcineurin inhibition rescues alloantigen-specific central memory T cell subsets that promote chronic GVHD

The Journal of clinical investigation

... These findings were consistent with previous studies. [25][26][27][28][29][30][31][32] These immune factors could regulate AML development by modulating inflammatory microenvironment, activating signaling pathways, or altering metabolism. 28,[33][34][35] We also revealed that AML patients with high IL-6 levels exhibited poorer OS rates. ...

CD8+ T-cell Differentiation and Dysfunction Inform Treatment Response in Acute Myeloid Leukemia
  • Citing Article
  • May 2024

Blood

... Although gene correction efficiency was only 10.5% in the bulk population of cells, the formation of AFs was observed at the dermo-epidermal junction in mice grafted with skin equivalents consisting of RDEB patientderived keratinocytes and PE-corrected RDEBF. Steinbeck et al. 82 reported a twin PE (twinPE) strategy utilizing the PEmax and PE6 primer editors to replace the target exon with recombinase attachment sequences, subsequently reintroducing a wild-type copy of the target exon to restore the C7 production in RDEB skin cells. ...

Twin Prime Editing Mediated Exon Skipping/Reinsertion for Restored Collagen VII Expression in Recessive Dystrophic Epidermolysis Bullosa
  • Citing Article
  • May 2024

Journal of Investigative Dermatology

... This finding might be attributable to IBD therapy. Several studies indicated that drugs such as prednisone and mesalazine, which were part of IBD therapy utilized in our cohort study, have direct inhibitory effect on cell proliferation 45,46 . Moreover, ileal CD patients exhibited downregulation of the Wnt transcription factor TCF-4 which is critical for ISC proliferation 47 . ...

Corticosteroids impair epithelial regeneration in immune-mediated intestinal damage

The Journal of clinical investigation

... Another example of an adverse event that requires complex multicellular interactions is immune effector cell-associated neurotoxicity syndrome (ICANS) occurring after CAR-T cell and T cell bispecific antibody (TCB) therapy. Although the pathophysiology remains poorly understood, the interaction between the blood-brain barrier (BBB), neurons, microglia, and immune effector cells seems to be an important driver of toxicity (18,19). Several MPS models have successfully replicated the human BBB, some of which incorporate neural tissue as well. ...

Immune-Effector-Cell-Associated-Neurotoxicity-Syndrome (ICANS) Pathophysiology Is Mediated By Microglia TGF-β-Activated Kinase-1 Signaling
  • Citing Article
  • November 2023

Blood

... Increasing AMPK activity in primary human CD4+ T cells pushed these cells towards an exaggerated Th1 phenotype (with increased interferon gamma (IFNg) and TNF)) while also downregulating the production of the Th2-associated cytokines IL-4 and IL-5. Furthermore, this polarization occurred in both bulk and previously differentiated populations (123). However, work with murine T cells lacking AMPK demonstrated no difference in FAO when activated in vivo, despite an appreciable difference in oxidative metabolism (124). ...

Overexpression of AMPKγ2 increases AMPK signaling to augment human T cell metabolism and function

Journal of Biological Chemistry

... ROCK2 (The rho-associated coiled-coil-containing protein kinase-2 (ROCK2) signaling pathway regulates the Th17/regulatory T cells balance and controls profibrotic pathways) inhibition significantly diminished STAT3 phosphorylation and binding to IL-17 and IL-21 promoters and reduced interferon regulatory factor 4 and nuclear hormone RORyt protein levels in T cells derived from healthy subjects or rheumatoid arthritis patients. Simultaneously, selective ROCK2 inhibition with Belumosudil (KD025) KD025 also promoted the suppressive function of regulatory T cells through up-regulation of STAT5 phosphorylation [164]. KD025 has been shown to ameliorate cGVHD in multiple murine models and inhibit the secretion of IL-21, IL-17 and interferon y along with decreasing phosphorylated STAT3 and reduced protein expression of interferon regulatory factor 4 and B-cell lymphoma (BCL6) in human peripheral blood mononuclear cells purified from active cGVHD patients [165]. ...

Selectivity matters: selective ROCK2 inhibitor ameliorates established liver fibrosis via targeting inflammation, fibrosis, and metabolism

Communications Biology