Yani Peng's research while affiliated with UCSF University of California, San Francisco and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (7)


HLA-A2-specific regulatory T cells
  • Patent
  • Full-text available

February 2023

·

45 Reads

·

·

·

[...]

·

Yani Peng

The present disclosure relates to chimeric-antigen-receptor regulatory T cells (CAR-Tregs) that include an exogenous nucleic acid encoding a chimeric antigen receptor (CAR) comprising the complementarity determining regions (CDRs) of a human anti-HLA-A2 antibody grafted onto an antibody scaffold, a hinge domain, a transmembrane domain; and an intracellular domain, where the CAR-Treg expresses the CAR on the cell surface. Vectors and methods of making and using such CAR-Tregs are disclosed as well.

Download
Share

Fig. 2. ARTEMIS darT reg manufacturing outcome and correlative data. (A) T reg expansion during the 16-day darT reg manufacturing for ARTEMIS (n = 10) and non-ARTEMIS individuals (n = 8). The star at day 0 indicates sBC stimulation, and the arrowhead at day 11 indicates aCD3/28 bead stimulation. (B) Day 16 darT reg (D16T reg ) product yield is shown for ARTEMIS (n = 10) versus non-ARTEMIS individuals (n = 8). Mann-Whitney test was used to assess the statistical significance of the difference between the two groups. (C) Day 16 viability was measured in infused (n = 5) and not infused (n = 5) darT reg products in the ARTEMIS trial. Mann-Whitney test was used to assess the statistical significance of the difference between the two groups. (D) Correlation between number of T regs purified on day 0 of culture initiation and day 16 darT reg product yield. Spearman correlation was used to assess the significance of the correlation. (E) Absolute (Abs.) T reg numbers per microliter of blood were measured using the T reg TruCount assay, and results from ARTEMIS (n = 9) and participants in other trials [type 1 diabetes , n = 25; pre-islet transplant, n = 4; pre-kidney transplant (KTx), n = 3; post-KTx, n = 9; pre-liver transplant, n = 3; pemphigus, n = 5; total, n = 51] were compared. The statistical significance of the difference between the two groups was assessed using an unpaired t test. For ARTEMIS, black symbols indicate products that failed to meet the minimal infusible dose. White symbols are products that achieved the full target dose. Half white symbols are products that achieved a partial dose. (F) Correlation between T reg fold expansion during the 16 days of culture and day 16 darT reg product yield. Spearman correlation was used to assess the significance of the correlation. In (A), (B), (D), and (F), the shaded area is below the 100 × 10 6 minimal infusible dose, and the black dashed line indicates the target dose threshold of 300 × 10 6 . For all panels, ARTEMIS participants are represented in orange and non-ARTEMIS data points are shown in blue. (A), (B), (D), and (F) include all darT reg GMP products manufactured at the UCSF facility except four (three due to the use of cryopreserved instead of fresh PBMCs as starting materials and one due to outgrowth of contaminating CD4 + T conv cells). Data for ARTEMIS include 10 data points from nine participants because manufacturing was attempted twice for participant 3. (C) and (E) included all available data. Data in (B) and (C) are presented as boxand-whisker plots showing all data points. The spread of the whiskers represents the range, the height of the box represents the inner quartile range, and the line in the box represents the median of the dataset. Data in (E) are presented as truncated violin plots showing all data points. The median is indicated by the thick white line, and the inner quartile range is indicated by the thin white lines.
Fig. 3. Pharmacokinetics of T reg products after infusion. (A) Percent deuterium enrichment in the genomic DNA of peripheral blood T regs collected after T reg infusion. The numbers of infused darT regs are indicated in the legend. (B) The relationship between the number of cells infused and the peak of deuterium enrichment was assessed using simple linear regression. (C) The biphasic exponential decay model was used to calculate the clearance rates (T 1/2 ) and relative size of the two distinct kinetic pools (fast and slow) of darT regs for participants 2 and 4. Day 1 data points were excluded from curve fitting due to variable mixing and trafficking of the infused darT regs at this early time point. All available data are included in (A) and (B). Decay curve analyses in (C) exclude day 1 data points (white circles) based on the reasoning that the infused T regs may not have sufficient time to reach equilibrium. Tang et al., Sci. Transl. Med. 14, eabo2628 (2022) 2 November 2022 5 of 12
Fig. 5. T reg donor reactivity before and after liver transplantation. Liver transplant recipients' PBMCs banked from the AWISH study (n = 16) were labeled with CTD before stimulation with activated B cells from the liver donors or a panel of allogeneic ( pan allo) B cells. CTD dilution was measured on day 4 of culture by flow cytometry. (A) Representative flow cytometric histograms of CTD dilution and analysis gates for CTD low cells for CD4 + FOXP3 + HELIOS + T regs . (B) Violin plot summaries of percentages of T regs in the CTD low gate stimulated by donor or pan allo B cells. Individual data points are represented by white circles, the medians are shown as thick black lines, and the inner quartiles are marked with thin black lines. Mixed-effects analysis was used to assess the significance of the changes over time. (C) Paired analyses are shown comparing the percentages of CTD low T regs before transplant (Pre-Tx; n = 16) and at 6 months (n = 14) or 2 years (n = 12) after transplantation. Wilcoxon test was used to assess the statistical significance of the differences observed. (D) Frequencies of darT reg precursors were calculated and summarized. Mixed-effects analysis and Wilcoxon test were used to assess the statistical significance of the differences observed. (E) Summary of percentages of T regs in the CTD low gate that had five or more rounds of cell division (left) and those that had divided only once (right). Mixed-effects analysis and Tukey multiple comparison posttest were used to assess the significance of the changes over time. P values are listed above the graphs. In all panels, the 6-month time point excludes two samples and the 2-year time point excludes four other samples due to insufficient cells recovered after thawing. Details of samples used in the analyses can be found in data file S3.
Selective decrease of donor-reactive Tregs after liver transplantation limits Treg therapy for promoting allograft tolerance in humans

November 2022

·

143 Reads

·

29 Citations

Science Translational Medicine

Promoting immune tolerance to transplanted organs can minimize the amount of immunosuppressive drugs that patients need to take, reducing lifetime risks of mortality and morbidity. Regulatory T cells (Tregs) are essential for immune tolerance, and preclinical studies have shown their therapeutic efficacy in inducing transplantation tolerance. Here, we report the results of a phase 1/2 trial (ARTEMIS, NCT02474199) of autologous donor alloantigen-reactive Treg (darTreg) therapy in individuals 2 to 6 years after receiving a living donor liver transplant. The primary efficacy endpoint was calcineurin inhibitor dose reduction by 75% with stable liver function tests for at least 12 weeks. Among 10 individuals who initiated immunosuppression withdrawal, 1 experienced rejection before planned darTreg infusion, 5 received darTregs, and 4 were not infused because of failure to manufacture the minimal infusible dose of 100 × 106 cells. darTreg infusion was not associated with adverse events. Two darTreg-infused participants reached the primary endpoint, but an insufficient number of recipients were treated for assessing the efficacy of darTregs. Mechanistic studies revealed generalized Treg activation, senescence, and selective reduction of donor reactivity after liver transplantation. Overall, the ARTEMIS trial features a design concept for evaluating the efficacy of Treg therapy in transplantation. The mechanistic insight gained from the study may help guide the design of future trials.


Anti‐HLA‐A2‐CAR Tregs prolong vascularized mouse heterotopic heart allograft survival

April 2022

·

36 Reads

·

26 Citations

American Journal of Transplantation

Alloantigen‐specific regulatory T cell (Treg) therapy is a promising approach for suppressing alloimmune responses and minimizing immunosuppression after solid organ transplantation. Chimeric antigen receptor (CAR) targeting donor alloantigens can confer donor reactivity to Tregs. However, CAR Treg therapy has not been evaluated in vascularized transplant or multi‐MHC mismatched models. Here, we evaluated the ability of CAR Tregs targeting HLA‐A2 (A2‐CAR) to prolong survival of heterotopic heart transplants in mice. After verifying the in vitro activation, proliferation, and enhanced suppression of A2‐CAR Tregs in the presence of A2‐antigen, we analyzed the in vivo function of Tregs in C57BL/6 (B6) mice receiving A2‐expressing heart allografts. A2‐CAR Treg infusion increased the median survival of grafts from B6.HLA‐A2 transgenic donors from 23 days to 99 days, whereas median survival with polyclonal Treg infusion was 35 days. In a more stringent model of haplo‐mismatched hearts from BALB/cxB6.HLA‐A2 F1 donors, A2‐CAR Tregs slightly increased median graft survival from 11 days to 14 days, which was further extended to >100 days when combined with a 9‐day course of rapamycin treatment. These findings demonstrate the efficacy of CAR Tregs, alone or in combination with immunosuppressive agents, toward protecting vascularized grafts in fully immunocompetent recipients.


IL-6 and TNFα Drive Extensive Proliferation of Human Tregs Without Compromising Their Lineage Stability or Function

December 2021

·

195 Reads

·

28 Citations

Frontiers in Immunology

Frontiers in Immunology

Treg therapies are being tested in clinical trials in transplantation and autoimmune diseases, however, the impact of inflammation on Tregs remains controversial. We challenged human Tregs ex-vivo with pro-inflammatory cytokines IL-6 and TNFα and observed greatly enhanced proliferation stimulated by anti-CD3 and anti-CD28 (aCD3/28) beads or CD28 superagonist (CD28SA). The cytokine-exposed Tregs maintained high expression of FOXP3 and HELIOS, demethylated FOXP3 enhancer, and low IFNγ, IL-4, and IL-17 secretion. Blocking TNF receptor using etanercept or deletion of TNF receptor 2 using CRISPR/Cas9 blunted Treg proliferation and attenuated FOXP3 and HELIOS expression. These results prompted us to consider using CD28SA together with IL-6 and TNFα without aCD3/28 beads (beadless) as an alternative protocol for therapeutic Treg manufacturing. Metabolomics profiling revealed more active glycolysis and oxidative phosphorylation, increased energy production, and higher antioxidant potential during beadless Treg expansion. Finally, beadless expanded Tregs maintained suppressive functions in vitro and in vivo. These results demonstrate that human Tregs positively respond to proinflammatory cytokines with enhanced proliferation without compromising their lineage identity or function. This property can be harnessed for therapeutic Treg manufacturing.


TNFa and IL-6 promote ex-vivo proliferation of lineage-committed human regulatory T cells

August 2021

·

80 Reads

·

1 Citation

Treg therapy is being tested in clinical trials in transplantation and autoimmune diseases, however, the impact of inflammation on Tregs is unclear. In this study, we challenged human Tregs ex-vivo with pro-inflammatory cytokines, TNFa and IL-6. These cytokines enhanced Treg proliferation induced by anti-CD3 and anti-CD28 or CD28 superagonist (CD28SA) while maintaining high expression of FOXP3 and HELIOS, demethylated FOXP3 enhancer, and low expression of cytokines IFNg, IL-4 and IL-17. Blocking TNF receptor signaling using etanercept or deletion of TNF receptor 2 using CRISPR/Cas9 blunted Treg proliferation and attenuated FOXP3 and HELIOS expression, revealing the importance of TNFR2 signaling in Treg proliferation and lineage stability. The robust proliferation induced by CD28SA with IL-6 and TNFa may be adopted for the expansion of therapeutic Tregs. Metabolomics analysis showed that Tregs expanded with CD28SA plus cytokines had more active glycolysis and oxidative phosphorylation, increased energy production, and higher antioxidant potential. Finally, CD28SA plus cytokine-expanded Tregs had comparable suppressive activity in vitro and in vivo in a humanized mouse model of graft-versus-host-disease when compared to Tregs expanded using the conventional protocol. These results demonstrate that human Tregs positively respond to proinflammatory cytokines with enhanced proliferation without compromising their lineage identity or function.


A human mutation in STAT3 promotes type 1 diabetes through a defect in CD8+ T cell tolerance

June 2021

·

101 Reads

·

33 Citations

Naturally occurring cases of monogenic type 1 diabetes (T1D) help establish direct mechanisms driving this complex autoimmune disease. A recently identified de novo germline gain-of-function (GOF) mutation in the transcriptional regulator STAT3 was found to cause neonatal T1D. We engineered a novel knock-in mouse incorporating this highly diabetogenic human STAT3 mutation (K392R) and found that these mice recapitulated the human autoimmune diabetes phenotype. Paired single-cell TCR and RNA sequencing revealed that STAT3-GOF drives proliferation and clonal expansion of effector CD8+ cells that resist terminal exhaustion. Single-cell ATAC-seq showed that these effector T cells are epigenetically distinct and have differential chromatin architecture induced by STAT3-GOF. Analysis of islet TCR clonotypes revealed a CD8+ cell reacting against known antigen IGRP, and STAT3-GOF in an IGRP-reactive TCR transgenic model demonstrated that STAT3-GOF intrinsic to CD8+ cells is sufficient to accelerate diabetes onset. Altogether, these findings reveal a diabetogenic CD8+ T cell response that is restrained in the presence of normal STAT3 activity and drives diabetes pathogenesis.


A human mutation in STAT3 promotes type 1 diabetes through a defect in CD8+ T cell tolerance

February 2021

·

116 Reads

Naturally occurring cases of monogenic type 1 diabetes (T1D) provide rare opportunities to establish direct mechanisms that cause this complex autoimmune disease. A recently identified de novo germline gain-of-function (GOF) mutation in the transcriptional regulator signal transducer and activator of transcription 3 (STAT3) was shown to cause neonatal T1D at birth. To investigate the role of STAT3 hyperactivity in T1D, we engineered a novel knock-in (KI) mouse incorporating this highly diabetogenic human mutation (K392R) in the STAT3 gene. These mice developed accelerated diabetes with severe insulitis and insulin autoantibodies, thereby recapitulating the human autoimmune diabetes phenotype. Paired T cell receptor (TCR) and transcriptome (RNA) sequencing in single cells revealed that STAT3-GOF drives the proliferation and clonal expansion of highly cytotoxic effector CD8+ T cells that are resistant to terminal exhaustion. Single-cell ATAC-seq showed that these effector T cells are epigenetically distinct and revealed differential chromatin architecture induced by STAT3-GOF. Analysis of islet TCR clonotypes revealed an effector CD8+ T cell reacting against the known antigen IGRP, and STAT3-GOF in an IGRP-reactive TCR transgenic model demonstrated that STAT3-GOF intrinsic to CD8+ T cells is sufficient to accelerate diabetes onset. Taken together, these findings reveal a diabetogenic CD8+ T cell response that is restrained in the presence of normal STAT3 activity and drives diabetes pathogenesis.

Citations (4)


... In another groundbreaking study, Tang et al. explored the use of donor-alloantigen-reactive regulatory T cells (darT regs ) to induce graft-specific immunosuppression in liver transplant recipients, potentially reducing the need for broad immunosuppressive drugs (74). This phase I/II trial, named ARTEMIS (NCT02474199), aimed to test the safety and efficacy of darT regs in maintaining stable liver function while allowing significant reduction in immunosuppression. ...

Reference:

Immune modulation in transplant medicine: a comprehensive review of cell therapy applications and future directions
Selective decrease of donor-reactive Tregs after liver transplantation limits Treg therapy for promoting allograft tolerance in humans

Science Translational Medicine

... Consequently, HLA-A2-CAR T regs have been developed, and these demonstrated attenuated effector T cell proliferation and reduced histological signs of graft rejection compared to polyclonal T regs in preclinical models of GvHD and skin allotransplantation in humanized NSG mice [74,75]. Moreover, in a murine heterotopic heart transplantation model using species-hybrid C57BL/6 mice expressing human HLA-A2 (B6.A2) and wild-type B6 mice, HLA-A2-CAR T reg therapy extended allograft survival from 35 to 99 days compared to polyclonal T regs [76], indicating that HLA-A2-CAR therapy can have allograft prolonging outcomes. However, using B6.A2×BALB/c F1 hybrids as donors, HLA-A2-CAR T regs only modestly increased median graft survival from 11 to 14 days compared to no therapy, which could be extended beyond 100 days in combination with a 9 day course of rapamycin [76]. ...

Anti‐HLA‐A2‐CAR Tregs prolong vascularized mouse heterotopic heart allograft survival
  • Citing Article
  • April 2022

American Journal of Transplantation

... The direct effects of IL-6 on stable lineage-committed Tregs remain more controversial. Recent data indicate that under certain conditions, IL-6 might in fact enhance Treg proliferation in a TNFR2dependent manner 76,77 . In our model, IL-6 blockade resulted in only a modest increase in systemic Treg levels, which were already elevated after ATG induction. ...

IL-6 and TNFα Drive Extensive Proliferation of Human Tregs Without Compromising Their Lineage Stability or Function
Frontiers in Immunology

Frontiers in Immunology

... Previous studies of STAT3 GOF mouse models from our group and others have shown that STAT3 GOF causes T cell dysregulation and lymphoproliferation, similar to patients (Masle-Farquhar et al., 2022;Schmitt et al., 2022;Warshauer et al., 2021;Woods et al., 2022). We previously demonstrated an expansion of effector CD4 + and CD8 + T cells in older STAT3 GOF mice and Th1 skewing in a colitis model (Schmitt et al., 2022). ...

A human mutation in STAT3 promotes type 1 diabetes through a defect in CD8+ T cell tolerance
Journal of Experimental Medicine (JEM)

Journal of Experimental Medicine (JEM)