Abstract 1570: STAT3 inhibition allows for TLR9-induced reprogramming of acute myeloid leukemia into antigen-presenting cells to generate T-cell mediated immune responses

Author(s):  
Yu-Lin Su ◽  
Dongfang Wang ◽  
Priyanka Duttagupta ◽  
Hiqing Li ◽  
Ya-Huei Kuo ◽  
...  
Leukemia ◽  
2021 ◽  
Author(s):  
Daniel T. Johnson ◽  
Jiarong Zhou ◽  
Ashley V. Kroll ◽  
Ronnie H. Fang ◽  
Ming Yan ◽  
...  

AbstractCancer vaccines are promising treatments to prevent relapse after chemotherapy in acute myeloid leukemia (AML) patients, particularly for those who cannot tolerate intensive consolidation therapies. Here, we report the development of an AML cell membrane-coated nanoparticle (AMCNP) vaccine platform, in which immune-stimulatory adjuvant-loaded nanoparticles are coated with leukemic cell membrane material. This AMCNP vaccination strategy stimulates leukemia-specific immune responses by co-delivering membrane-associated antigens along with adjuvants to antigen-presenting cells. To demonstrate that this AMCNP vaccine enhances leukemia-specific antigen presentation and T cell responses, we modified a murine AML cell line to express membrane-bound chicken ovalbumin as a model antigen. AMCNPs were efficiently acquired by antigen-presenting cells in vitro and in vivo and stimulated antigen cross-presentation. Vaccination with AMCNPs significantly enhanced antigen-specific T cell expansion and effector function compared with control vaccines. Prophylactic vaccination with AMCNPs enhanced cellular immunity and protected against AML challenge. Moreover, in an AML post-remission vaccination model, AMCNP vaccination significantly enhanced survival in comparison to vaccination with whole leukemia cell lysates. Collectively, AMCNPs retained AML-specific antigens, elicited enhanced antigen-specific immune responses, and provided therapeutic benefit against AML challenge.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A767-A767
Author(s):  
Yu-Lin Su ◽  
Marcin Kortylewski ◽  
Priyanka Duttagupta

BackgroundSignal transducer and activator of transcription factor 3 (STAT3) is commonly activated in acute myeloid leukemia (AML) and known for supporting cancer cell proliferation and survival. Recently, we demonstrated that STAT3 also plays a critical role ensuring AML immune evasion. Intravenous injections of bi-functional decoy oligodeoxyribonucleotides (CpG-STAT3dODN) blocked STAT3 activity and induced TLR9 signaling in Cbfb/MYH11/Mpl (CMM) AML cells, thereby resulting in immunogenic effects and T cell-mediated immune responses and leukemia regression.MethodsTo understand the molecular mechanisms of the CpG-STAT3 decoy-induced AML differentiation and immunogenicity, we performed global gene expression analysis on the in vivo treated AML cells using oligonucleotide strategy as well as an inducible STAT3 gene silencing.ResultsTranscriptional profiling revealed the upregulation of myeloid cell differentiation related genes, such as Irf8, Cebpa, and Gadd45A with reduction of oncogenic Runx1 and Run1t1 in CMM leukemic cells after CpG-STAT3dODN but not after control treatments. CpG-STAT3dODN treatment also upregulated set of antigen-presentation related genes, such as CIIta, Il12a, and Ifng in CMM AML cells. Importantly, the induction of Irf8 and Cebpa, with the concomitant suppression of Runx1 were found specifically in the subset of differentiated CD11b+ CMM cells but not in the bulk CD11b– leukemic cells. These effects were likely related to epigenetic reprogramming of AML cells as indicated by treatment-induced changes in the expression and protein levels of STAT3 regulated DNA methyltransferases, DNMT1 and DNMT3a/b. Furthermore, our initial studies suggest that STAT3 inhibition/TLR9 activation leads to immunogenic effects also in a xenotransplanted model of human FLT3-ITD MV4-11 leukemia in humanized mice. CpG-STAT3dODN alone or together with clinically-relevant demethylating agent (Decitabine) triggered differentiation of MV4-11 cells into CD11b+HLA-DR+CD86+ antigen-presenting cells (APCs) and increased ratio of CD8+ to regulatory T cells in the bone marrow, thereby reducing leukemia burden.ConclusionsOur results suggest that eliminating STAT3 permits the TLR9-driven reprogramming of AML cells into APCs to unleash T cell-mediated responses against leukemia.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A232-A232
Author(s):  
Haoxiao Zuo ◽  
Satwinder Kaur Singh ◽  
Marie-José Van Lierop ◽  
Jorn Kaspers ◽  
Remco Bos ◽  
...  

BackgroundDCP-001 is a cancer relapse vaccine derived from the DCOne® human leukemic cell line. During manufacturing, DCOne® cells are shifted towards a mature dendritic cell (mDC) phenotype, combining an endogenous tumor antigen repertoire (e.g. WT-1, RHAMM and PRAME) with a mDC costimulatory profile and providing the basis for the highly immunogenic vaccine DCP-001. In a phase I clinical study in acute myeloid leukemia (AML), DCP-001 demonstrated to be safe and to induce multifunctional antitumor immune responses.1 It has also been reported that DCP-001 induces antitumor immunity against multiple myeloma cells in peripheral blood mononuclear cells (PBMC) from multiple myeloma patients and that DCP-001 antigenic material is transferred to host antigen presenting cells (APC), possibly via extracellular vesicles.2 However, the possibility of direct interactions between DCP-001 and host APC has not yet been investigated.MethodsTo further elucidate the mode of action of DCP-001, we studied the interactions of DCP-001 with human PBMC and isolated immature monocyte-derived DCs (iMoDC) in in vitro co-culture studies. A human skin explant model was used to determine uptake of DCP-001 by migrating skin DCs after intradermal injection.ResultsWe found that DCP-001 stimulates the secretion of various proinflammatory cytokines (IL-1β, GM-CSF, IFN-γ, IL-2, TNF-α, and IL-6) and chemokines (IL-8 and RANTES) in PBMC. In addition, we demonstrate that DCP-001 is efficiently taken up by iMoDC via direct cell-cell interactions and that this phagocytic process is influenced by ”eat-me” and ”don’t eat me” signaling pathways. Blocking of the ”eat-me” signals calreticulin and phosphatidylserine inhibited the uptake of DCP-001, whereas blockade of the ”don’t eat me” signal CD47 enhanced DCP-001 uptake. After intradermal injection of DCP-001 in an ex-vivo human skin model, its uptake by skin-emigrating DCs was demonstrated as well as simultaneous activation of these DCs.ConclusionsOur data suggest a key role for host antigen presenting cells in the triggering of immune responses upon DCP-001 vaccination. In addition, the data provide rationale for potential combination therapies based on DCP-001 and inhibitors of the CD47 pathway.Referencesvan de Loosdrecht AA, et al. A novel allogeneic off-the-shelf dendritic cell vaccine for post-remission treatment of elderly patients with acute myeloid leukemia. Cancer Immunol Immunother 2018;67(10):1505–1518.Leaf RK, et al. DCOne as an allogeneic cell-based vaccine for multiple myeloma. J Immunother 2017;40(9):315–322.


Blood ◽  
2001 ◽  
Vol 97 (9) ◽  
pp. 2764-2771 ◽  
Author(s):  
Beth D. Harrison ◽  
Julie A. Adams ◽  
Mark Briggs ◽  
Michelle L. Brereton ◽  
John A. Liu Yin

Abstract Effective presentation of tumor antigens is fundamental to strategies aimed at enrolling the immune system in eradication of residual disease after conventional treatments. Myeloid malignancies provide a unique opportunity to derive dendritic cells (DCs), functioning antigen-presenting cells, from the malignant cells themselves. These may then co-express leukemic antigens together with appropriate secondary signals and be used to generate a specific, antileukemic immune response. In this study, blasts from 40 patients with acute myeloid leukemia (AML) were cultured with combinations of granulocyte-macrophage colony-stimulating factor, interleukin 4, and tumor necrosis factor α, and development to DCs was assessed. After culture, cells from 24 samples exhibited morphological and immunophenotypic features of DCs, including expression of major histocompatibility complex class II, CD1a, CD83, and CD86, and were potent stimulators in an allogeneic mixed lymphocyte reaction (MLR). Stimulation of autologous T-cell responses was assessed by the proliferative response of autologous T cells to the leukemic DCs and by demonstration of the induction of specific, autologous, antileukemic cytotoxicity. Of 17 samples, 11 were effective stimulators in the autologous MLR, and low, but consistent, autologous, antileukemic cytotoxicity was induced in 8 of 11 cases (mean, 27%; range, 17%-37%). This study indicates that cells with enhanced antigen-presenting ability can be generated from AML blasts, that these cells can effectively prime autologous cytotoxic T cells in vitro, and that they may be used as potential vaccines in the immunotherapy of AML.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4070-4070
Author(s):  
Marcin Kortylewski ◽  
Priyanka Duttagupta ◽  
Yu-Lin SU ◽  
Tomasz Adamus ◽  
Guido Marcucci

Abstract STAT3 transcription factor is persistently activated in cancer cells and in diverse tumor-associated immune cells being an important oncogene and an essential immune checkpoint regulator. It is a highly desirable but challenging therapeutic target for pharmacological intervention. We previously demonstrated that ligand for the intracellular receptor TLR9 (CpG oligonucleotide) allows for the uptake and cytoplasmic delivery of decoy oligodeoxynucleotides (dODNs) to inhibit transcriptional activity of STAT3 in a variety of human and mouse acute myeloid leukemia (AML) cells. Here, we demonstrate that CpG-STAT3dODN injected intravenously to mice engrafted with Cbfb/MYH11 AML, dramatically altered leukemic cell morphology, by reducing nucleus-cytoplasm ratio and increasing mitochondria size, and also resulting in metabolic shift from glycolysis to oxidative phosphorylation, resulting in increased mitochondrial ATP production. The transcriptional profiling of AML cells isolated from spleens of mice treated using CpG-STAT3dODN vs control CpG-scrODN or PBS, revealed the increased expression of genes regulating myeloid cell differentiation (Irf8, Cebpa, Gadd45a) and antigen-presentation (B2m, CIIta, IL-12a, Ifng), with decrease of leukemia-related regulators Runx1and Run1t1. Thein vivodifferentiation of leukemic cells was confirmed using flow cytometry by detecting appearance of the population of AML-derived CD11b+/MHCII+/CD86+myeloid cells after CpG-STAT3dODN but not control treatments. Furthermore, we confirmed that the differentiated AML cells have immunogenic effects, which drive expansion of Th1 effector T cell responses in Cbfb/MYH11 as well as C1498 AML models. Overall, our studies suggest that STAT3-inhibition can unleash immune-stimulatory potential downstream of TLR9 signaling transforming AMLcells into antigen-presenting cells. These findings support development of CpG-STAT3dODN strategy as a new bi-functional agent for AML immunotherapy. This project described was supported by the National Cancer Institute of the National Institutes of Health under award number R01CA213131 to M.K. Disclosures Kortylewski: N/A: Patents & Royalties: I am an inventor on the US patent 9,976,147.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 414-414
Author(s):  
Jochen Greiner ◽  
Michael Schmitt ◽  
Li Li ◽  
Krzysztof Giannopoulos ◽  
Katrin Bosch ◽  
...  

Abstract Several Tumor-associated antigens (TAAs) are expressed in acute myeloid leukemia (AML) and elicit specific immune responses of CD8 positive T cells. These specific T cell responses against leukemic blasts expressing TAAs might play a critical role in the control of minimal residual disease (MRD) in AML. Therefore, we investigated whether TAAs inducing specific immune responses in AML patients were associated with the clinical outcome. A DNA-microarray analysis of 116 AML samples was performed to correlate expression of TAAs to the clinical outcome. In these AML patients specific T cell responses to TAAs were assessed by ELISPOT analysis, tetramer staining and chromium release assays. Quantitative RT-PCR based validation of our results demonstrated the power of DNA microarray technology. We found a significant correlation of high mRNA expression of the TAA G250/CA9 with a longer overall survival (P=0.022), a trend for better outcome in patients with high expression levels of PRAME (P=0.103), and a hint for RHAMM/HMMR. In contrast, for other TAAs like WT1, TERT, PRTN3, BCL2, and LAMR1 we found no correlation with clinical outcome of AML patients. Moreover, co-expression of RHAMM/HMMR, PRAME and G250/CA9 provided a favorable prognostic effect (P=0.005). We found specific T cell responses at high frequency for these three antigens in AML patients. Positive immune reactions were detected in 8/17 (47%) AML patients for RHAMM/HMMR-R3-derived, in 7/10 (70%) for PRAME-P3-derived, and in 6/10 (60%) for newly characterized G250/CA9-G2-derived peptides. We detected a significant increased immune response of AML patients in complete remission compared to AML patients with refractory disease (P<0.001). Furthermore, we could demonstrate specific lysis of T2 cells and AML blasts presenting these epitope peptides RHAMM/HMMR-R3, PRAME-P3 and G250/CA9-G2. In conclusion, the expression of the TAAs RHAMM/HMMR, PRAME and G250/CA9 can induce strong anti-leukemic immune responses of CD8 positive T cells possibly enabling the control of MRD in AML patients. Thus, the antigens RHAMM/HMMR, PRAME and G250/CA9 represent interesting target structures for polyvalent immunotherapeutic approaches in AML.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1023-1023
Author(s):  
Victoria Y Ling ◽  
Denise Lee ◽  
Zoe K Mcquilten ◽  
Sharon Avery ◽  
Michael Low ◽  
...  

Abstract Acute myeloid leukemia with normal cytogenetics (CN-AML) is biologically and clinically heterogeneous. Better prognostication and therapeutic strategies are required to improve outcomes. Evasion of host immunity is an important hallmark of cancer. Conversely, robust immune responses have been associated with improved outcomes in solid organ and hematological malignancies, for example, Hodgkin lymphoma. Moreover, targeting immune responses specifically at leukemic cells utilizing bi-specific T cell-engaging antibodies and chimeric antigen receptor T cell immunotherapy demonstrates efficacy primarily in lymphoid malignancies. We therefore sought to explore the effect of immune response on outcomes in myeloid cancers by aiming to determine whether T cell numbers in CN-AML at diagnosis would predict survival. Using diagnostic trephine sections from patients with CN-AML at our institution between 2006 and 2013, we performed immunohistochemistry for CD3, CD8 and Granzyme B (GB) expression. CD4 was not assessable due to high background staining. For each trephine section, three representative fields at 200x magnification were analyzed with a mean of 4245 cells per field. Positive cells, enumerated using Fiji© image analysis software (v1.48o), were expressed as a percentage of total cells. The primary outcome was overall survival (OS). Cox regression was used for univariate and multivariate analyses. Survival was estimated by Kaplan-Meier analyses and categories compared using the log-rank test. Seventy-five patients (52% male, median age 61 years) were analyzed with a median follow-up of 15.9 months. Fifty-six (75%) patients were treated with curative intent with 21 (28%) proceeding to allogeneic transplant either in first complete remission (CR1) (11 patients) or CR2 (10 patients). Of the 33 (44%) patients who died, 18 never achieved CR and 15 relapsed including 3 patients who were allografted in CR1 and 3 patients allografted in CR2. OS was superior in patients with CD3% above the 75th centile (>11.89%) compared with those below (p = 0.0323) (Figure 1). Factors significantly associated with better OS on univariate analyses were younger age, allograft, absence of preceding myelodysplastic syndrome, absence of primary refractory disease, and FLT3-ITD negativity. CD3 (p=0.096), CD8, GB, gender, NPM1 positivity, relapse and blast percentage at diagnosis were not statistically significant. However, in a multivariate analysis of CD3 and the variables found to be significant in the univariate analyses, higher CD3 was found to be an independent predictor of OS (Hazard ratio 0.922 for death, 95% CI 0.851-0.998, p=0.045). Relapse-free survival was evaluable in 49 patients who achieved a CR and was not influenced by CD3, CD8 or GB expression. FLT3 and NPM1 status were available for 53 (71%) patients. Within molecular subgroups FLT3-ITD+ (n=20) and NPM1+/FLT3-ITD- (n=11), there was no survival difference between groups above and below the median for CD3, CD8 or GB. However, in FLT3-ITD-/NPM1- patients (n=22), CD3 > median (11.89%) (Figure 2A) and CD8 > median (10.66%) (Figure 2B) were associated with significantly superior OS whilst GB > median (1.17%) was not significantly associated (p=0.2330). Our findings show that in CN-AML, especially in the FLT3-ITD-/NPM1- subgroup, higher CD3 T cell numbers are associated with improved survival. This suggests baseline immune status may have prognostic value. These results provide impetus for studies into immune therapies in AML and prospectively assessing baseline immune status as a potential prognostic marker in CN-AML. Figure 1 Figure 1. Figure 2 Figure 2. Figure 3 Figure 3. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (6) ◽  
pp. 1282-1289 ◽  
Author(s):  
Jochen Greiner ◽  
Yoko Ono ◽  
Susanne Hofmann ◽  
Anita Schmitt ◽  
Elmar Mehring ◽  
...  

Abstract Mutations in the nucleophosmin gene (NPM1mut) are one of the most frequent molecular alterations in acute myeloid leukemia (AML), and immune responses may contribute to the favorable prognosis of AML patients with NPM1mut. In the present study, we were able to demonstrate both CD4+ and CD8+ T-cell responses against NPM1mut. Ten peptides derived from wild-type NPM1 and NPM1mut were subjected to ELISPOT analysis in 33 healthy volunteers and 27 AML patients. Tetramer assays against the most interesting epitopes were performed and Cr51-release assays were used to show the cytotoxicity of peptide-specific T cells. Moreover, HLA-DR–binding epitopes were used to test the role of CD4+ T cells in NPM1 immunogenicity. Two epitopes (epitopes #1 and #3) derived from NPM1mut induced CD8+ T-cell responses. A total of 33% of the NPM1mut AML patients showed immune responses against epitope #1 and 44% against epitope #3. Specific lysis of leukemic blasts was detected. To obtain robust immune responses against tumor cells, the activation of CD4+ T cells is crucial. Therefore, overlapping (OL) peptides were analyzed in ELISPOT assays and OL8 was able to activate both CD8+ and CD4+ T cells. The results of the present study show that NPM1mut induces specific T-cell responses of CD4+ and CD8+ T cells and therefore is a promising target for specific immunotherapies in AML.


2021 ◽  
Vol 14 (11) ◽  
pp. 1105
Author(s):  
Laura Jimbu ◽  
Oana Mesaros ◽  
Alexandra Neaga ◽  
Ana Maria Nanut ◽  
Ciprian Tomuleasa ◽  
...  

Tumor cells promote the suppression of host anti-tumor type 1 T cell responses by various mechanisms, including the upregulation of surface inhibitory molecules such as programmed death ligand (PD-L)-1, and the production of immunosuppressive cytokines such as interleukin-10 (IL-10). There are over 2000 trials investigating PD-L1 and/or its receptor programmed-death 1 (PD-1) blockade in cancer, leading to the approval of PD-1 or PD-L1 inhibitors in several types of solid cancers and in hematological malignancies. The available data suggest that the molecule PD-L1 on antigen-presenting cells suppresses type 1 T cell immune responses such as cytotoxicity, and that the cytokine IL-10, in addition to downregulating immune responses, increases the expression of inhibitory molecule PD-L1. We hypothesize that the manipulation of both the co-inhibitory network (with anti-PD-L1 blocking antibodies) and suppressor network (with anti-IL-10 blocking antibodies) is an attractive immunotherapeutic intervention for acute myeloid leukemia (AML) patients ineligible for standard treatment with chemotherapy and hematopoietic stem cell transplantation, and with less severe adverse reactions. The proposed combination of these two immunotherapies represents a new approach that can be readily translated into the clinic to improve the therapeutic efficacy of AML disease treatment.


Sign in / Sign up

Export Citation Format

Share Document