scholarly journals NY-ESO-1 Single Edited T Cells to Treat Multiple Myeloma without Inducing GvHD

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 308-308
Author(s):  
Sara Mastaglio ◽  
Pietro Genovese ◽  
Zulma Magnani ◽  
Elisa Landoni ◽  
Barbara Camisa ◽  
...  

Abstract Background. Gene transfer of T cell receptors (TCR) specific for tumor associated antigens is a promising approach of adoptive immunotherapy for cancer patients. However, T cells transduced with an exogenous TCR, have proved less effective than naturally occurring tumor specific T cells, with suboptimal clinical results. This could be due to the dilution of the tumor specific TCR that competes with endogenous TCRs for its expression on the T cell surface. Moreover, mispairing between endogenous and exogenous TCR a and b chains could generate new TCRs with unpredictable and potentially harmful specificities. To overcome these issues, our group developed a TCR gene editing procedure, based on the knockout of the endogenous TCR genes by transient exposure to a and b chain specific Zinc Finger Nucleases (ZFNs), followed by the introduction of tumor-specific TCR genes by lentiviral vectors (Provasi, Genovese et al, Nature Medicine 2102). The complete editing procedure requires multiple manipulation steps involving repeated cell activation cycles and four transduction procedures. Conversely, ‘single TCR editing’ (SE), based on the disruption of a single endogenous TCR chain, followed by transfer of the tumor specific TCR, generates redirected T cells fully devoid of their natural TCR repertoire, in a single round of cell activation. The aim of this project is to determine the efficacy and safety profile of SE T cells in vitro and in vivo. Results. We validated the SE protocol exploiting an HLA-A2 restricted TCR specific for the cancer testis antigen NY-ESO-1157-165 peptide. NY-ESO-1 is highly expressed by a large number of solid tumors and by the majority of monoclonal plasma cells of patients affected by high risk multiple myeloma, while its expression in healthy tissues is restricted to testicular germ cells. T cells harvested from healthy donors were activated with CD3/CD28 conjugated beads and cultured with low doses of IL7 and IL15. Two days after activation ZFNs were transiently delivered to T cells by mRNA transfection or by infection with adenoviral vectors, with similar efficacy. Sorted CD3- T cells were transduced at day 8 with a bidirectional lentiviral vector encoding both the a and b chains of the NY-ESO-1-specific TCR. The SE strategy allowed the rapid production of high numbers of extremely fit tumor specific T cells, enriched for cells with an early differentiated phenotype (CD45RA+/CD62L+/CD95+, namely stem memory T cells). NY-ESO-1 TCR expression, investigated through dextramer specific binding, was significantly higher in SE than in T cells undergoing TCR gene transfer (TR). To test the efficacy of NY-ESO-1 redirected T cells we took advantage of the NY-ESO-1+HLA-A2+ U266 myeloma cell line. When tested against U266 cells in different assays (co-cultures, g-IFN and 51Cr release), all NY-ESO-1 redirected T cells showed a strikingly high killing potential. However, when we compared the alloreactive potential of the different NY-ESO-1 specific T cell populations in mixed lymphocyte reactions against completely mismatched allogeneic targets, we interestingly observed that the lysis of the allogeneic target by TR cells was significantly higher than that of SE T cells (p=0.05). This suggests that the residual endogenous polyclonal TCRs and possibly mispaired TCRs expressed on the cell surface of TR T cells can lead to off-target recognition, while SE T cells are devoid of such reactivity. These results were validated in NSG mice, engrafted with U266 cells and subsequently infused with NY-ESO-1 specific T cells. All animals treated with tumor specific T cells were completely disease free at the time of sacrifice, demonstrating the powerful anti-tumor potential of the NY-ESO-1 TCR. However, a substantial proportion of mice administered with TR cells suffered from xenogeneic chronic graft versus host disease (GvHD), while all animals treated with SE T cells remained free of GvHD throughout the course of the study. Discussion. The single gene editing protocol enables the rapid generation of clinically relevant doses of highly performing tumor specific T cells, fully devoid of their endogenous TCR repertoire, and thus particularly appealing for a future clinical translation. Donor-derived SE T cells, with a significantly reduced alloreactive potential, could be especially suitable to treat patients with minimal residual disease after allogeneic hematopoietic stem cell transplantation. Disclosures Reik: Sangamo Bioscience: Employment. Holmes:Sangamo Bioscience: Employment. Gregory:Sangamo Bioscience: Employment.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3002-3002 ◽  
Author(s):  
Lorenz Jahn ◽  
Renate S. Hagedoorn ◽  
Pleun Hombrink ◽  
Michel G.D. Kester ◽  
Dirk M. van der Steen ◽  
...  

Abstract Therapeutic reactivity of CD20-specific monoclonal antibodies (mAb) or CD19-specific chimeric antigen receptor (CAR)-transduced T cells is exerted by targeting extracellular antigens. In contrast to mAbs and CARs, T cell receptors (TCRs) recognize antigen-derived peptides that are bound to human leukocyte antigen (HLA) molecules on the cell surface. Since HLA molecules constantly sample the entire endogenous proteome of a cell, extracellular and intracellular antigens are presented and can thus be recognized by a TCR. Here, we identified the intracellular transcription factor Bob1 encoded by gene POU2AF1 as a suitable target for immunotherapy. Bob1 is highly expressed in CD19+ B cells, acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), mantle cell lymphoma (MCL) and multiple myeloma (MM) and is absent in the non-B lineages including CD34+ hematopoietic progenitor cells (HPCs), T cells, fibroblasts, keratinocytes and gastrointestinal tract. Bob1 is localized intracellularly but HLA-presented Bob1-derived peptides are accessible on the cell surface to TCRs and can thus be recognized by T cells. From the HLA-presented ligandome (Mol Cell Proteomics, 2013;12:1829) we identified naturally processed Bob1-derived peptides displayed in HLA-A*0201 (HLA-A2) and in HLA-B*0702 (HLA-B7). Since auto-reactivity towards self-antigens such as Bob1 is prevented by depleting high-avidity T cells recognizing self-antigens in self-HLA, we exploited the immunogenicity of these peptides presented in allogeneic HLA. From a HLA-A2/B7-negative healthy individual we isolated T cell clone 4G11 demonstrating high sensitivity and specificity for Bob1-derived peptide Bob144 presented in HLA-B7. Bob1-dependent recognition was demonstrated by transduction of Bob1 into cell lines that otherwise lack Bob1 expression. No harmful toxicities of clone 4G11 were observed against a wide panel of Bob1-negative stimulator cells including HLA-B7-positive CD34+ HPCs, T cells, monocytes, immature and mature dendritic cells, and fibroblasts even under simulated inflamed conditions. Furthermore, stringent HLA-B7-restricted recognition was observed for clone 4G11 when tested against a stimulator panel expressing a wide range of common and rare HLA class I and II molecules. Clone 4G11 demonstrated clinical applicability by efficiently recognizing HLA-B7+ primary ALL, CLL and MCL. Furthermore, reproducible strong recognition of purified primary HLA-B7+ MM could be demonstrated. Therefore, the TCR of clone 4G11 may be used for immunotherapy by administering TCR-transduced T cells to patients suffering from B cell malignancies including multiple myeloma. Retroviral gene transfer of TCR 4G11 led to efficient cell surface expression demonstrated by binding of TCR-transduced CD8+ T cells to pMHC-tetramer composed of peptide Bob144 bound to HLA-B7. TCR-modified CD8+ T cells strongly recognized Bob1-expressing HLA-B7+ multiple myeloma cell lines U266 and UM9, and ALL cell lines. TCR-modified T cells efficiently lysed HLA-B7+ primary ALL, CLL and MCL at very low effector-to-target ratios. In addition, highly purified primary multiple myeloma samples were also readily lysed. Furthermore, TCR-transduced T cells strongly proliferated in an antigen-specific manner when stimulated with primary malignant cell samples including ALL, CLL, and MCL or MM cell lines. As expected, TCR-transduced T cells also lysed autologous primary and CD40L-stimulated B cells since these targets cells also express Bob1. In contrast, no lysis of Bob1-negative autologous primary and activated T cells, or monocytes was observed when co-cultured with TCR-transduced T cells. In summary, we identified the intracellular transcription factor Bob1 encoded by gene POU2AF1 as a suitable target for TCR-based immunotherapies of B cell malignancies. Bob1-specific T cell clone 4G11 efficiently recognized primary B cell leukemia and multiple myeloma. Gene transfer of TCR of clone 4G11 installed Bob1-reactivity and specificity onto recipient T cells shown here by cytolytic capacity and proliferation upon antigen encounter. TCR gene transfer approaches using this Bob1-specific TCR can bring novel treatment modalities and possibly curative therapy to patients with B cell malignancies including multiple myeloma. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2898-2898
Author(s):  
Sara Mastaglio ◽  
Pietro Genovese ◽  
Zulma Magnani ◽  
Elena Provasi ◽  
Angelo Lombardo ◽  
...  

Abstract The genetic transfer of tumor-specific T cell receptors (TCRs) into mature T lymphocytes enables T cell specificity to be redirected towards cancer cells, however the transfer of novel TCRs into polyclonal T cells, while overcoming tolerance barriers, may be limited by factors intrinsic to TCR biology. Specifically, the tumor-specific a and b TCR chains are expressed in lymphocytes that already bear an endogenous TCR on their cell surfaces. Gene-modified cells thus express at least two different TCRs that compete for binding to the CD3 complex, resulting in mutual TCR dilution and reduced avidity. Furthermore, since TCRs are heterodimers, the a and b chains of the endogenous TCR have the potential to mispair with the respective α and β chains of the transgenic TCR to produce a new hybrid TCR, with unpredictable and potentially harmful specificity. This represents a major concern in TCR transfer adoptive immunotherapy, both in autologous and allogeneic settings. To permanently eliminate the expression of the endogenous TCR and the risk of mispairing, our group recently developed a TCR gene editing approach. This technique is based on the transient transfer of zinc-finger nucleases (ZFN) to induce DNA double strand breaks in the constant regions of the endogenous TCR a and/or b chain genes, leading to permanent gene disruption. Upon lentiviral transfer of a tumor-specific TCR, such fully TCR-edited T cells express only the exogenous tumor-specific TCR transgenes at high levels (Provasi, Genovese et al., Nature Medicine 2012). While the complete editing procedure (both a and b TCR chains) currently requires multiple manipulation steps, ‘single TCR editing’, based on the ZFN-mediated knock-down of a single endogenous TCR chain (a or b) followed by the introduction of the tumor-specific TCR, enables the generation of redirected T cells devoid of their natural TCR repertoire during a single round of T cell activation, improving the feasibility of the clinical translation of this approach. This might be particularly useful to reduce the risk of GvHD after allogeneic hematopoietic stem cell transplantation. We exploited a HLA-A2 restricted TCR specific for NY-ESO-1, a cancer testis antigen expressed by solid tumors and hematological malignancies, to directly compare the safety and efficacy profile of unedited TCR transferred T cells (TR), single TCR edited (SE) lymphocytes and completely TCR edited (CE) T cells. We observed that gene editing does not detectably affect the phenotype, function or proliferative potential of engineered lymphocytes. Our protocols ensured the maintenance of the early differentiated memory phenotype, with enrichment in central memory and CD45RA+/CD62L+/CD95+ memory stem T (TSCM) cells. Upon lentiviral transfer of the NY-ESO-1-specific TCR, we observed significantly higher levels of the tumor-specific TCR expression, evaluated as NY-ESO-1 specific dextramer binding, in edited versus transferred T cells (relative fluorescence intensity to untransduced cells: CE: 37; SE: 31; TR: 19). Edited T cells were more efficient than unedited-TCR transferred T cells in killing NY-ESO-1-pulsed cell lines (half maximal effective peptide concentration in a 51Cr release assay: 310, 210, 186 nM for TR, SE and CE T cells respectively) and NY-ESO-1+ myeloma cell lines naturally processing the antigen. Importantly our SE and CE T cells displayed no activity against NY-ESO-1- targets. Importantly, in NSG mice, NY-ESO-1 redirected single edited and complete edited T cells completely eliminated an NY-ESO1+ HLA-A2+, WT1- myeloma cell line, that, on the contrary, expanded in bone marrow in the presence of WT1-redirected CE T cells. Our results demonstrate that the TCR single editing approach is effective in redirecting T cell specificity as evidenced by the potent anti-tumor effect observed while potentially eliminating the risk of GvHD associated with the infusion of donor-derived lymphocytes. Moreover, the relative speed and simplicity of the TCR single editing protocol should facilitate its clinical application to patients with hematological malignancies. Disclosures: Reik: Sangamo BioSciences: Employment. Holmes:Sangamo BioSciences: Employment. Gregory:Sangamo BioSciences: Employment.


2018 ◽  
Vol 215 (3) ◽  
pp. 985-997 ◽  
Author(s):  
Akiko Seki ◽  
Sascha Rutz

CRISPR (clustered, regularly interspaced, short palindromic repeats)/Cas9 (CRISPR-associated protein 9) has become the tool of choice for generating gene knockouts across a variety of species. The ability for efficient gene editing in primary T cells not only represents a valuable research tool to study gene function but also holds great promise for T cell–based immunotherapies, such as next-generation chimeric antigen receptor (CAR) T cells. Previous attempts to apply CRIPSR/Cas9 for gene editing in primary T cells have resulted in highly variable knockout efficiency and required T cell receptor (TCR) stimulation, thus largely precluding the study of genes involved in T cell activation or differentiation. Here, we describe an optimized approach for Cas9/RNP transfection of primary mouse and human T cells without TCR stimulation that results in near complete loss of target gene expression at the population level, mitigating the need for selection. We believe that this method will greatly extend the feasibly of target gene discovery and validation in primary T cells and simplify the gene editing process for next-generation immunotherapies.


Blood ◽  
2020 ◽  
Vol 135 (15) ◽  
pp. 1232-1243 ◽  
Author(s):  
Kodandaram Pillarisetti ◽  
Suzanne Edavettal ◽  
Mark Mendonça ◽  
Yingzhe Li ◽  
Mark Tornetta ◽  
...  

Abstract T-cell–mediated approaches have shown promise in myeloma treatment. However, there are currently a limited number of specific myeloma antigens that can be targeted, and multiple myeloma (MM) remains an incurable disease. G-protein–coupled receptor class 5 member D (GPRC5D) is expressed in MM and smoldering MM patient plasma cells. Here, we demonstrate that GPRC5D protein is present on the surface of MM cells and describe JNJ-64407564, a GPRC5DxCD3 bispecific antibody that recruits CD3+ T cells to GPRC5D+ MM cells and induces killing of GPRC5D+ cells. In vitro, JNJ-64407564 induced specific cytotoxicity of GPRC5D+ cells with concomitant T-cell activation and also killed plasma cells in MM patient samples ex vivo. JNJ-64407564 can recruit T cells and induce tumor regression in GPRC5D+ MM murine models, which coincide with T-cell infiltration at the tumor site. This antibody is also able to induce cytotoxicity of patient primary MM cells from bone marrow, which is the natural site of this disease. GPRC5D is a promising surface antigen for MM immunotherapy, and JNJ-64407564 is currently being evaluated in a phase 1 clinical trial in patients with relapsed or refractory MM (NCT03399799).


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2966-2966 ◽  
Author(s):  
Manisha Bhutani ◽  
David Foureau ◽  
Tammy Cogdill ◽  
Kyle Madden ◽  
Qing Zhang ◽  
...  

Abstract BACKGROUND: Lenalidomide is an immunomodulatory drug (IMiD) with co-stimulatory effects on immune effector cells in vitro and is an approved treatment for multiple myeloma (MM), although its mode of action in patients is not well defined. We studied the impact of lenalidomide maintenance therapy, following autologous stem cell transplant (ASCT), on NK and NK-T polarization (i.e. activating or inhibitory molecules) and, T cell activation (early vs. late activation) in patients with multiple myeloma. PATIENTS AND METHODS: In this ongoing prospective study with a targeted enrollment of 28 newly diagnosed multiple myeloma patients, blood samples are being collected at 2 to 3 months post ASCT, before starting lenalidomide maintenance therapy (baseline), and serially after 1, 3 and 6 months of treatment (T+1mo, T+3mo, T+6mo). Using a 9 color flow cytometry panel, peripheral blood samples were analyzed for expression of CD3 and CD56 to define NK (CD56+ CD3-), NKT (CD56+ CD3+), and T cell (CD56- CD3+) subsets. Killer 'inhibitory' Ig-like receptors, (KiR2DS4, KiR3DL1) natural killer group 2 proteins (NKG2a, NKG2D) and natural killer p46 protein (NKp46) expression were quantified to assess polarization of NK, and NK-T cells. Programmed death receptor 1 (PD-1) and T-cell Ig and mucin receptor 3 (Tim3) expression was quantified to assess T cell activation state. Flow cytometry data were acquired on a BD FACSAria II, and analyzed using FlowJo version X software. RESULTS: Samples from 11 patients have been collected and analyzed so far (11 baseline, 6 T+1mo, 4 T+3mo). At baseline lymphoid cells represent 12-46% of white blood cells (WBC), this heterogeneity being mainly driven by a wide range of T cell relative distribution among patients (30-74 % lymphoid cells). Phenotypically, NK cells at baseline mainly express natural cytotoxicity receptors (NKp46 and NKG2D), whereas NK-T cell also express NKG2D but approximately 1/3 also express PD-1 indicating they may be functionally defective. T cells at baseline express early T cell activation markers NKG2D and approximately 1/3 also stained positive for late T cell activation marker PD-1. Lymphoid cells relative distribution among WBC tends to normalize at T+1mo of treatment (15 to 35 % of WBC) before expanding at T+3mo (35 to 43 % of WBC). Phenotypically, across the 27 immune variables analyzed, each multiple myeloma patient displayed high level of immune homeostasis after 1 or 3 months of lenalidomide treatment. Noticeably, Nkp46 expression by NK cell and PD-1 expression by NK-T cells decreased in 4/6 patients and, NKG2D expression by T cell decreased in all but one patient during lenalidomide therapy. CONCLUSION: To our knowledge, this is the first study examining the influence of lenalidomide maintenance on the comprehensive immune repertoire in the post-ASCT setting in MM patients. The wide heterogeneity of NK, NK-T and T cell distribution observed at baseline among lymphoid cells indicates the potential effect of post-ASCT immune reconstitution and immunomodulatory the impact of lenalidomide. The capacity of lenalidomide to mediate effects on several immune cells raises the question as to which, if any, of these changes correlate with clinical responses. In our study, serially collected data from each patient, when completed would determine the impact of lenalidomide immunomodulatory effect of therapeutic efficacy and PFS duration in relation to immune reconstitution stage. Disclosures Cogdill: Millennium: Speakers Bureau; Onyx: Speakers Bureau; Celgene: Speakers Bureau; Novartis: Speakers Bureau. Ghosh:Celgene: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Pharmacyclics: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Usmani:Sanofi: Honoraria, Research Funding; Millennium: Honoraria, Speakers Bureau; Onyx: Honoraria, Research Funding, Speakers Bureau; Pharmacyclics: Research Funding; Celgene: Honoraria, Speakers Bureau; Janssen Oncology: Honoraria, Research Funding; Array BioPharma: Honoraria, Research Funding.


2018 ◽  
Author(s):  
Christopher Bricogne ◽  
Michael Fine ◽  
Pedro M. Pereira ◽  
Julia Sung ◽  
Maha Tijani ◽  
...  

AbstractTMEM16F, an ion channel gated by high cytoplasmic Ca2+, is required for cell surface phosphatidylserine exposure during platelet aggregation and T cell activation. Here we demonstrate in Jurkat T cells and HEK293 cells that TMEM16F activation triggers large-scale surface membrane expansion in parallel with lipid scrambling. Following TMEM16F mediated scrambling and surface expansion, cells undergo extensive membrane shedding. The membrane compartment that expands the cell surface does not involve endoplasmic reticulum or acidified lysosomes. Surprisingly, T cells lacking TMEM16F expression not only fail to expand surface membrane, but instead rapidly internalize membrane via massive endocytosis (MEND). The T cell co-receptor PD-1 is selectively shed when TMEM16F triggers membrane expansion, while it is selectively internalized in the absence of TMEM16F. Its participation in this trafficking is determined by its single transmembrane domain. Thus, we establish a fundamental role for TMEM16F as a regulator of Ca2+-activated membrane trafficking.


eLife ◽  
2018 ◽  
Vol 7 ◽  
Author(s):  
Jie Geng ◽  
John D Altman ◽  
Sujatha Krishnakumar ◽  
Malini Raghavan

When complexed with antigenic peptides, human leukocyte antigen (HLA) class I (HLA-I) molecules initiate CD8+ T cell responses via interaction with the T cell receptor (TCR) and co-receptor CD8. Peptides are generally critical for the stable cell surface expression of HLA-I molecules. However, for HLA-I alleles such as HLA-B*35:01, peptide-deficient (empty) heterodimers are thermostable and detectable on the cell surface. Additionally, peptide-deficient HLA-B*35:01 tetramers preferentially bind CD8 and to a majority of blood-derived CD8+ T cells via a CD8-dependent binding mode. Further functional studies reveal that peptide-deficient conformers of HLA-B*35:01 do not directly activate CD8+ T cells, but accumulate at the immunological synapse in antigen-induced responses, and enhance cognate peptide-induced cell adhesion and CD8+ T cell activation. Together, these findings indicate that HLA-I peptide occupancy influences CD8 binding affinity, and reveal a new set of regulators of CD8+ T cell activation, mediated by the binding of empty HLA-I to CD8.


2021 ◽  
Author(s):  
James Robert Byrnes ◽  
Amy M Weeks ◽  
Julia Carnevale ◽  
Eric Shifrut ◽  
Lisa Kirkemo ◽  
...  

Immunosuppressive factors in the tumor microenvironment (TME) impair T cell function and limit the anti-tumor immune response. T cell surface receptors that influence interactions and function in the TME are already proven targets for cancer immunotherapy. However, surface proteome remodeling of primary human T cells in response to suppressive forces in the TME has never been characterized systematically. Using a reductionist cell culture approach with primary human T cells and SILAC-based quantitative cell surface capture glycoproteomics, we examined how two immunosuppressive TME factors, regulatory T cells (Tregs) and hypoxia, globally affect the activated CD8+ surface proteome (surfaceome). Surprisingly, the CD8+/Treg co-culture only modestly affected the CD8+ surfaceome, but did reverse several activation-induced surfaceomic changes. In contrast, hypoxia dramatically altered the CD8+ surfaceome in a manner consistent with both metabolic reprogramming and induction of an immunosuppressed state. The CD4+ T cell surfaceome similarly responded to hypoxia, revealing a novel hypoxia-induced surface receptor program. Our findings are consistent with the premise that hypoxic environments create a metabolic challenge for T cell activation, which may underlie the difficulty encountered in treating solid tumors with immunotherapies. Together, the data presented here provide insight into how suppressive TME factors remodel the T cell surfaceome and represent a valuable resource to inform future therapeutic efforts to enhance T cell function in the TME.


2020 ◽  
Author(s):  
Daniel Peltier ◽  
Molly Radosevich ◽  
Guoqing Hou ◽  
Cynthia Zajac ◽  
Katherine Oravecz-Wilson ◽  
...  

ABSTRACTMechanisms governing allogeneic T-cell responses after allogeneic hematopoietic stem cell (HSC) and solid organ transplantation are incompletely understood. Long non-coding RNAs (lncRNA) do not code for, but control gene expression with tissue specificity. However, their role in T-cell alloimmunity is unknown. We performed RNA-seq on donor T-cells from HSCT patients and found that increasing strength of allogeneic stimulation caused greater differential expression of lncRNAs. The differential expression was validated in an independent patient cohort, and also following ex vivo allogeneic stimulation of healthy human T-cells. Linc00402, a novel, conserved lncRNA, was identified as the most differentially expressed and was enriched 88 fold in human T-cells. Mechanistically, it was mainly located in the cytoplasm, and its expression was rapidly reduced following T-cell activation. Consistent with this, tacrolimus preserved the expression of Linc00402 following T-cell activation, and lower levels of Linc00402 were found in patients who subsequently went on to develop acute graft versus host disease (GVHD). The dysregulated expression of Linc00402 was also validated in murine T-cells, both in vitro and in vivo. Functional studies using multiple modalities to deplete Linc00402 in both mouse and human T-cells, demonstrated a critical role for Linc00402 in the T-cell proliferative response to an allogeneic stimulus but not a non-specific anti-CD3/CD28 stimulus. Thus, our studies identified Linc00402 as a novel, conserved regulator of allogeneic T-cell function. Because of its T-cell specific expression and its impact on allogeneic T-cell responses, targeting Linc00402 may improve outcomes after allogeneic HSC and solid organ transplantation.One sentence summaryLncRNAs are differentially expressed by allogeneic antigen-stimulated T-cells, and the novel lncRNA, Linc00402, is a specific regulator of mouse and human allogeneic T-cells.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3202-3202
Author(s):  
Cameron S. Bader ◽  
Henry Barreras ◽  
Casey O. Lightbourn ◽  
Sabrina N. Copsel ◽  
Dietlinde Wolf ◽  
...  

Graft-versus-host disease (GVHD) remains a significant cause of morbidity and mortality in patients receiving allogeneic hematopoietic stem cell transplants (aHSCTs). Pre-HSCT conditioning typically consists of irradiation and drug administration resulting in the death of rapidly dividing cells and release of endogenous danger signals. These molecules drive the activation of antigen presenting cells (APCs) and the differentiation of allo-reactive donor T cells, leading to damage of particular host tissues characteristic of GVHD. Cell death following conditioning has promoted the hypothesis that sensors of cytoplasmic DNA damage in GVHD target tissues contribute to pro-inflammatory cytokine production. We identified a role for Stimulator of Interferon Genes (STING), an innate immune sensor, in GVHD using pre-clinical MHC-matched unrelated donor (MUD) aHSCT models. Here we show that STING rapidly promotes donor CD8+ T cell activation and recipient APC death early after aHSCT. To assess STING involvement immediately post-HSCT, cytokine mRNA expression was examined 48 hrs after transplant of MUD C3H.SW bone marrow (BM) + T cells into irradiated B6 wildtype (WT) or STING-/- recipients. Colon tissue from STING-/- recipients had >2x reduction in IFNβ, TNFα and IL-6 mRNA vs WT. MUD STING-/- HSCT recipients also experienced decreased weight loss, GVHD scores and skin pathology 6 wks post-HSCT vs WT. Double chimerism studies showed that the absence of STING in non-hematopoietic cells was responsible for GVHD amelioration. Conversely, a single dose of the highly specific STING agonist DMXAA given in vivo increased IFNβ, TNFα and IL-6 mRNA expression in WT, but not STING-/-, colon tissue 48 hrs after transplant and increased GVHD scores and lethality post-HSCT. Post-transplant cytoxan treatment abolished the ability of DMXAA to augment GVHD, supporting the notion that STING signaling increases donor T cell activation during aHSCT. To evaluate the potential impact of STING in the clinical setting, we transplanted C3H.SW BM + T cells into mice homozygous for a murine homologue of a human allele associated with diminished STING activity (STINGHAQ/HAQ) and found that these mice also exhibited diminished GVHD. Interestingly, our findings that STING deficiency ameliorates GVHD in MUD aHSCT contrasts to reported observations that STING deficiency can exacerbate GVHD after MHC-mismatched (MMUD) aHSCT (Fischer J, et al, Sci. Transl. Med. 2017). Since CD4+ and CD8+ T cells are central in MMUD and MUD GVHD, respectively, we hypothesized that STING's effect on the predominant T cell subset in each model may explain these seemingly paradoxical results in STING-/- vs WT recipients. Therefore, we transplanted MMUD BALB/c BM + CD8+ T cells into B6-WT and STING-/- mice and found that - in contrast to MMUD recipients of combined CD4+ and CD8+ T cells - STING-/- recipients developed lower GVHD clinical scores, reduced skin pathology and had lower frequencies of activated T cells 8 wks post-HSCT vs WT, supporting a role for STING in the promotion of CD8+ T cell-mediated GVHD. Next, we investigated if recipient APCs played a role in STING's enhancement of CD8+ T cell-mediatedGVHD. We found that STING-/- mice had greater frequencies and numbers of recipient splenic CD11b+CD11c+ APCs 1 day after MMUD B6 into BALB/c aHSCT (Fig. A). BALB/c-STING-/- APCs also expressed reduced MHC class I protein levels (Fig. B). Moreover, STING-/- recipient spleens contained lower numbers of donor CD8+ T cells producing IFNγ and TNFα (Fig. C). These data support the hypothesis that STING contributes to early activation of donor CD8+ T cells and elimination of recipient APCs. Next, to identify if the loss of host MHC II+ APCs affected subsequent donor CD4+ T cell activation, B6-Nur77GFP transgenic donor T cells were used to explicitly monitor T cell receptor signaling. Consistent with increased numbers of host MHC II+ APCs in the spleens of STING-/- recipients 1 day post-aHSCT, we found greater frequencies and numbers of donor Nur77GFP CD4+ T cells expressing GFP, CD69 and IFNγ in STING-/- spleens 6 days after transplant (Fig. D). In summary, our studies demonstrate that STING plays an important role in regulating aHSCT and provide one potential mechanism by which STING could promote CD8+ T cell-mediated GVHD yet diminish CD4+-mediated GVHD. Overall, our studies suggest this pathway can provide a target for new therapeutic strategies to ameliorate GVHD. Disclosures Blazar: BlueRock Therapeutics: Membership on an entity's Board of Directors or advisory committees; Childrens' Cancer Research Fund: Research Funding; KidsFirst Fund: Research Funding; Tmunity: Other: Co-Founder; Kamon Pharmaceuticals, Inc: Membership on an entity's Board of Directors or advisory committees; Regeneron Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Five Prime Therapeutics Inc: Co-Founder, Membership on an entity's Board of Directors or advisory committees; Magenta Therapeutics and BlueRock Therapeuetics: Membership on an entity's Board of Directors or advisory committees; Fate Therapeutics, Inc.: Research Funding; RXi Pharmaceuticals: Research Funding; Alpine Immune Sciences, Inc.: Research Funding; Abbvie Inc: Research Funding; Leukemia and Lymphoma Society: Research Funding. Levy:Heat Biologics: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pelican Therapeutics: Consultancy, Research Funding.


Sign in / Sign up

Export Citation Format

Share Document