Tetrahydrouridine-decitabine for non-cytotoxic epigenetic therapy of NSCLC to enhance immunotherapeutic effect of anti-PD1 in vivo.

2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 11552-11552 ◽  
Author(s):  
Kai Kang ◽  
David S. Schrump ◽  
Anish Thomas ◽  
Kurt Alex Schalper ◽  
Yogen Saunthararajah ◽  
...  

11552 Background: NSCLC response to anti-PD1 therapy is ~20%, largely because most NSCLC avoids immune-recognition in the 1stplace, e.g., by epigenetics to suppress neo-antigen expression. DNA methyltransferase (DNMT1) mediates this repression and is depleted by decitabine (Dec). Unfortunately Dec has trivial distribution into solid cancer tissues because of rapid deamination by cytidine deaminase (CDA). Therefore, to improve tissue-distribution of Dec with the low Cmax/long Tmax profile needed for DNMT1-depletion without cytotoxicity, we combined Dec with a CDA inhibitor tetrahydrouridine (THU). Methods: C57/BL6 mice were inoculated with LL3-luc cells via tail vein. After documentation of lung invasion by live-imaging, mice (n = 5/group) were randomized to PBS, THU-Dec (10/0.1 mg/kg sc 3×/wk), anti-PD1 (5 mg/kg ip q5d, DX400 from Merck) or THU-Dec/anti-PD1 combination. Antigen presentation, PD, MDSCs, and T-cells were measured in blood and tumor. Results: THU-Dec or anti-PD1 alone decreased tumor by imaging and increased survival, however, THU-Dec/anti-PD1 combination extended median survival the most and completely regressed tumor in 2/5 mice (median survival days PBS 37, THU-Dec 56, anti-PD1 62, THU-Dec/anti-PD1 77). Rechallenge of cured mice with LL2-luc confirmed immune-memory effect, with no engraftment vs expected engraftment in controls. Consistent with the pharmacologic rationale, THU-Dec produced > 2-fold more DNMT1-depletion in tumor vs PBS. Consistent with non-cytotoxic effect, absolute lymphocyte counts were preserved with THU-Dec, while numbers of G-MDSC decreased (2.9 k/µL PBS vs 0.3 k/µL THU-Dec/anti-PD1, p < 0.01). Expression of neoantigens MAGE-A1 and MAGE-A3 increased > 4-fold with THU-Dec vs PBS (p < 0.01). THU-Dec/anti-PD1 increased tumor infiltrating lymphocytes 6-fold vs PBS (p < 0.01) and decreased regulatory T-cells 2.5-fold vs PBS (p < 0.01). IFNγ expression in tumor increased 2.4-fold with THU-Dec/anti-PD1 vs PBS (p < 0.01). Conclusions: THU-Dec/anti-PD1 produced marked survival improvements and cures in tumor-bearing mice, scientific validation of our clinical trial NCT02664181 combining THU-Dec/nivolumab in 2nd line for NSCLC.

2019 ◽  
Vol 47 (3) ◽  
pp. 244-253
Author(s):  
Mehmet Sahin ◽  
Emel Sahin

Naturally occurring regulatory T cells (nTregs) are produced under thymic (tTregs) or peripherally induced (pTregs) conditions in vivo. On the other hand, Tregs generated from naive T cells in vitro under some circumstances, such as treatment with transforming growth factor-β (TGFB), are called induced Tregs (iTregs). Tregs are especially characterized by FOXP3 expression, which is mainly controlled by DNA methylation. nTregs play important roles in the suppression of immune response and self-tolerance. The prostaglandin E2 (PGE2) pathway was reported to contribute to regulatory functions of tumor-infiltrating nTregs. In this study, we examined whether PGE2 contributes to the formation of iTregs treated with TGFB1 and 5-aza-2′-deoxycytidine (5-aza-dC), which is a DNA methyltransferase inhibitor. We found that the protein and gene expression levels of FOXP3 and IL-10 were increased in 5-aza-dC and TGFB1-treated T cells in vitro. However, the addition of PGE2 to these cells reversed these increments significantly. In CFSE-based cell suppression assays, we demonstrated that PGE2 decreased the suppressive functions of 5-aza-dC and TGFB1-treated T cells.


2010 ◽  
Vol 207 (8) ◽  
pp. 1791-1804 ◽  
Author(s):  
Elizabeth D. Thompson ◽  
Hilda L. Enriquez ◽  
Yang-Xin Fu ◽  
Victor H. Engelhard

Studies of T cell responses to tumors have focused on the draining lymph node (LN) as the site of activation. We examined the tumor mass as a potential site of activation after adoptive transfer of naive tumor-specific CD8 T cells. Activated CD8 T cells were present in tumors within 24 h of adoptive transfer and proliferation of these cells was also evident 4–5 d later in mice treated with FTY720 to prevent infiltration of cells activated in LNs. To confirm that activation of these T cells occurred in the tumor and not the tumor-draining LNs, we used mice lacking LNs. Activated and proliferating tumor-infiltrating lymphocytes were evident in these mice 24 h and 4 d after naive cell transfer. T cells activated within tumors acquired effector function that was evident both ex vivo and in vivo. Both cross-presenting antigen presenting cells within the tumor and tumor cells directly presenting antigen activated these functional CD8 effectors. We conclude that tumors support the infiltration, activation, and effector differentiation of naive CD8 T cells, despite the presence of immunosuppressive mechanisms. Thus, targeting of T cell activation to tumors may present a tool in the development of cancer immunotherapy.


2019 ◽  
Vol 1 (1) ◽  
Author(s):  
Yisel Rivera-Molina ◽  
Hong Jiang ◽  
Juan Fueyo ◽  
Teresa Nguyen ◽  
Dong Ho Shin ◽  
...  

Abstract Background Viroimmunotherapy is evolving as a strong alternative for the standard treatment of malignant gliomas. Promising results from a recent clinical trial testing the anticancer effect of Delta-24-RGD in patients with glioblastoma suggested the induction of antitumoral immunity after viral administration. To further enhance the anti-glioma immune effect, we have armed Delta-24-RGD with the costimulatory ligand GITRL (Delta-24-GREAT [Glucocorticoid Receptor Enhanced Activity of T cells]). Methods We tested the infectivity and replication of Delta-24-GREAT, and the expression of ectopic GITRL in human and murine glioma cell lines. In vivo experiments involved the intracranial implantation of glioma cells into an immunocompetent model to study the anticancer effect, and rechallenging experiments to study long-term protection. Phenotypic and functional characterization of lymphocyte populations were performed by FACS and ELISA for Th1 cytokines expression, respectively. Results Our results showed that Delta-24-GREAT infects and induces the expression of GITRL. Delta-24-GREAT prolonged the survival of glioma-bearing immunocompetent mice and resulted in both anti-viral and anti-glioma immune responses, including increased frequency of central memory CD8+ T cells. Rechallenging the surviving mice with a second implantation of glioma cells did not lead to tumor growth; however, the surviving mice developed lethal tumors when B16/F10 melanoma cells were implanted intracranially, strongly indicating that the immune response was specific for glioma antigens. Conclusions GITRL-armed Delta-24-RGD treatment results in an antigen-restricted antitumor memory, an enhanced anti-glioma effect, and the generation of central immune memory. Our results strongly indicate that this strategy represents a vertical advance in virotherapy designed to treat patients with malignant brain tumors.


eLife ◽  
2014 ◽  
Vol 3 ◽  
Author(s):  
Simone A Nish ◽  
Dominik Schenten ◽  
F Thomas Wunderlich ◽  
Scott D Pope ◽  
Yan Gao ◽  
...  

Innate immune recognition is critical for the induction of adaptive immune responses; however the underlying mechanisms remain incompletely understood. In this study, we demonstrate that T cell-specific deletion of the IL-6 receptor α chain (IL-6Rα) results in impaired Th1 and Th17 T cell responses in vivo, and a defect in Tfh function. Depletion of Tregs in these mice rescued the Th1 but not the Th17 response. Our data suggest that IL-6 signaling in effector T cells is required to overcome Treg-mediated suppression in vivo. We show that IL-6 cooperates with IL-1β to block the suppressive effect of Tregs on CD4+ T cells, at least in part by controlling their responsiveness to IL-2. In addition, although IL-6Rα-deficient T cells mount normal primary Th1 responses in the absence of Tregs, they fail to mature into functional memory cells, demonstrating a key role for IL-6 in CD4+ T cell memory formation.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3925-3925
Author(s):  
Anilkumar Gopalakrishnapillai ◽  
Colin Correnti ◽  
Anne Kisielewski ◽  
Allison Kaeding ◽  
Soheil Meshinchi ◽  
...  

Acute myeloid leukemia (AML) remains the type of pediatric leukemia with poorest outcome. Despite maximally intensive therapy, approximately 20% of patients experience recurrent disease. Novel targeted therapies are needed to improve survival. We recently identified that mesothelin, a well-validated target in some cancers, is also highly expressed in a subset of pediatric AML samples (Tarlock et al., Blood, 128:2873, 2016). Considering that it is not expressed in normal tissues in children (Fan et al., Blood, 130:3792, 2017), MSLN is a viable target for immunotherapies such as Bispecific T-cell Engaging antibodies (BiTEs) that combine antibody single chain variable (scFv) regions targeting a cancer antigen and the T-cell co-receptor CD3. We designed and tested the efficacy and specificity of BiTEs targeting MSLN in disseminated xenograft models of pediatric AML. Using scFv sequences derived from Amatuximab, which recognizes the N-terminal domain of the GPI-linked ectodomain of MSLN, targeting region 1 of MSLN, and from Blinatumomab/AMG-330 targeting CD3, we engineered and expressed two kinds of BiTE molecules - a canonical BiTE and an IgG BiTE, a larger molecule with improved serum half life in vivo. To evaluate the specificity and efficacy of canonical BiTEs, MV4;11-MSLN cell line was generated by lentiviral transduction of parental MV4;11 cells which do not constitutively express MSLN (Fig. 1A, B). These two cell lines were injected i.v. into NSG-SGM3 mice. Once engraftment was confirmed, purified human T cells (3 x 106) were injected to act as effector cells. Mice were then treated with the canonical αMSLN-αCD3 BiTE at a dose of 3 mg/kg/day daily for 6 days. A cohort of mice that were untreated or received BiTE or T-cell infusion only served as controls. Mice from both treated and untreated groups had to be euthanized when they presented with distended abdomens due to myeloid sarcomas and no significant differences in survival were observed. Post euthanasia, bone marrows were flushed and evaluated for the percentage of AML cells (human CD45+CD33+) and T cells (human CD45+CD3+). We observed that the αMSLN-αCD3 BiTE was effective in promoting T-cell activation (based on high T-cell counts compared to mice injected with T-cells alone) and greatly reducing leukemic burden in mice injected with MV4;11 cells engineered to express MSLN (Fig. 1C, D). Similar results were obtained using BiTEs targeting a different MSLN epitope. No T-cell expansion and anti-leukemic effect was observed in mice engrafted with parental MV4;11 cells. Although, there were no significant differences between the median survival of untreated and treated miceThese data highlight the specificity and efficacy of the aMSLN-CD3 BiTEs. Among a panel of 8 AML patient-derived xenograft (PDX) lines generated in the laboratory, NTPL-146 bearing MLL-ENL fusion was found to have endogenous MSLN expression (Fig. 1E). We evaluated the efficacy of αMSLN-αCD3 canonical BiTE (3 mg/Kg Qdx6) against NTPL-146 PDX line in NSG-B2m mice by transfusing human CD3+ T-cells to act as effector cells. A Kaplan-Meier survival plot based on the time when each mouse reached experimental end-point (reduced body weight greater than 20%, impaired mobility, hind limb paralysis) showed that the survival benefit for mice receiving BiTE in the presence of human T-cells (4/6 mice survived at the end of experiment) greatly exceeded the efficacy of T-cells alone (22-day improvement in median survival with no surviving mice), or BiTE treatment alone (no improvement in survival) compared to untreated mice (Fig. 1F, P<0.001). These data validate the efficacy of MSLN targeting BiTEs in a PDX model with endogenous MSLN expression. The efficacy of canonical vs IgG BiTEs was evaluated in MV4;11-MSLN xenografted mice. Mice were dosed Qd5x3 for canonical BiTE and Q7dx3 for IgG BiTE as shown (Fig. 1G). IgG BiTE treatment along with T-cell infusion significantly prolonged survival in mice transplanted with MV4;11-MSLN (Fig. 1H), suggesting that IgG BiTE was far more efficacious than canonical BiTEs (P<0.01). Taken together, these data indicate that MSLN-targeting BiTEs could be used as novel immunotherapy for pediatric AML with MSLN expression. Figure 1 Disclosures Kaeding: Celgene: Employment.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 424-424 ◽  
Author(s):  
Assaf Lask ◽  
Eran Ophir ◽  
Noga Or-Geva ◽  
Adva Cohen ◽  
Ran Afik ◽  
...  

Abstract Abstract 424 Generation of T cells endowed with graft versus leukemia (GVL) and depleted of graph versus host (GVH) activity represents a highly desirable goal in BM transplantation (BMT). Donor CTLs directed against 3rd party stimulators were shown to effectively enhance engraftment of BM allografts without causing GVHD. Recently, we demonstrated that activated anti 3rd party CD8 cells with central memory phenotype (Tcm), can further support and improve BM engraftment. This is likely due to the improved lymph node homing of the Tcms, their proliferative capacity and prolonged persistence in BMT recipients. Surprisingly, when tested in vitro, anti 3rd party human CTLs were found to eliminate B CLL and other lymphoma cell types. Investigating the mechanism of the cytotoxicity revealed a unique TCR independent effect mediated against both autologous and allogeneic targets. Inhibition studies using specific blocking Ab revealed initial binding of CTLs to their targets via a rapid ICAM1-LFA1 adhesion, followed by slow induction of apoptosis following interaction of the CD8 on the CTL with MHCI on the tumor cell. The killing did not involve classical CTL death molecules. To establish an appropriate mouse model, we initially verified that anti 3rd party Tcm derived from (B6xBALB/c)F1 exhibit TCR independent killing of A20 lymphoma cells of BALB/c origin (34.8±12.1% in 4 experiments, 5:1 Tcm/lymphoma cell ratio, in comparison to A20 cells incubated without Tcm, p<0.05). Moreover, after 16 hours of incubation with Tcm, AnnexinV staining of A20 cells was enhanced compared to basal staining (14.8±4.5% and 5.2±2.2% respectively, in three experiments, p<0.05), suggesting an apoptosis based mechanism as previously described for the killing of human lymphoma. Using luciferase expressing A20 cells, we were able to follow the fate of the malignant cells in vivo, and study the effect of added donor anti 3rd party Tcm to either syngeneic or allogeneic BMT in a model simulating minimal residual disease. In the syngeneic model, 3×106 Nude BALB/c BM cells were transplanted into lethally irradiated BALB/c mice together with 5000 A20 cells. On the next day, syngeneic Tcm were infused. As can be seen in Fig. 1, none of the untreated mice survived (0/7) 100 days post BMT (23 days median survival). Administration of 1×107 or 2×107 syngeneic Tcms led to significantly diminished tumor burden (Fig. 1A) and improved overall survival of 28% (2/7, P<0.0001) and 40% (2/5, P<0.002) with median survival of 49 and 80 days, respectively (Fig. 1B). As expected, further improvement of tumor eradication was found in the allogeneic setting, in which an additional GVL effect, mediated by residual alloreactivity in the absence of GVHD, occurred in addition to the TCR independent killing. In the allogeneic model, 3×106 allogeneic Nude B6 BM cells were transplanted together with 5000 A20 cells into lethally irradiated BALB/c mice, and mice were treated with donor type Tcm. Similar to the results in the syngeneic model, none of the untreated mice survived 100 days (0/8) (23 days median survival), while administration of 5×106 donor Tcms, led to remarkable overall survival of 100% (7/7) at 100 days post BMT (Fig. 2). Although the allogeneic Tcm displayed enhanced GVL activity compared to syngeneic Tcm, this effect was not associated with any manifestation of GVHD. Thus, as previously described, the weight and overall appearance of mice receiving allogeneic anti 3rd party Tcms were the same as that of mice in the control group, radioprotected with a transplant of Nude BM alone. Collectively, we demonstrate for the first time, by in vivo imaging the GVL reactivity of murine anti 3rd party Tcm. Considering our previous finding that anti 3rd party Tcm markedly enhance BM allografting, these results suggest that anti 3rd party Tcm can provide a ‘double supportive effect' by promoting BM engraftment, and concurrently inducing GVL reactivity without causing GVHD. Such cell therapy is highly attractive, in particular for elderly patients with B CLL and other B cell malignancies who might not tolerate aggressive conditioning. Figure 1: Syngeneic anti 3rd party Tcm inhibit tumor relapse after syngeneic BM transplantation (A) Tumor growth was monitored by weekly bioluminesence imaging (BLI); (B) Survival curves. Figure 1:. Syngeneic anti 3rd party Tcm inhibit tumor relapse after syngeneic BM transplantation . / (A) Tumor growth was monitored by weekly bioluminesence imaging (BLI); (B) Survival curves. Figure 2: Allogeneic anti 3rd party Tcm inhibit tumor relapse after allogeneic BM transplantation (A) Tumor growth was monitored by weekly BLI; (B) Survival curves. Figure 2:. Allogeneic anti 3rd party Tcm inhibit tumor relapse after allogeneic BM transplantation . / (A) Tumor growth was monitored by weekly BLI; (B) Survival curves. Disclosures: No relevant conflicts of interest to declare.


2009 ◽  
Vol 83 (8) ◽  
pp. 3528-3539 ◽  
Author(s):  
Keisha S. Mathurin ◽  
Gregory W. Martens ◽  
Hardy Kornfeld ◽  
Raymond M. Welsh

ABSTRACT The bacillus Calmette-Guerin (BCG) strain of Mycobacterium bovis is used in many parts of the world as a vaccine against Mycobacterium tuberculosis. Some epidemiological evidence has suggested that BCG immunization may have unpredicted effects on resistance to other pathogens. We show here in a mouse model that BCG immunization followed by antibiotic treatment to clear the host of the pathogen rendered three strains of mice partially resistant to infection with vaccinia virus (VV) but not to lymphocytic choriomeningitis virus (LCMV). VV-challenged BCG-immune mice developed a striking splenomegaly and elevated CD4 and CD8 T-cell responses by 6 days postinfection (p.i.). However, resistance to VV infection could be seen as early as 1 to 2 days p.i. and was lost after antibody depletion of CD4 T-cell populations. BCG- but not LCMV-immune memory phenotype CD4 T cells preferentially produced gamma interferon (IFN-γ) in vivo after VV challenge. In contrast, LCMV-immune CD8 T cells preferentially produced IFN-γ in vivo in response to VV infection. In BCG-immune mice the resistance to VV infection and VV-induced CD4 T-cell IFN-γ production were ablated by cyclosporine A, which inhibits signaling through the T-cell receptor. This study therefore demonstrates CD4 T-cell-mediated heterologous immunity between a bacterium and virus. Further, it poses the question of whether BCG immunization of humans alters resistance to unrelated pathogens.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii90-ii90
Author(s):  
Alexander Haddad ◽  
Jordan Spatz ◽  
Sara Collins ◽  
Matheus Pereira ◽  
Sabraj Gill ◽  
...  

Abstract BACKGROUND Severe local and systemic immune suppression in glioblastoma (GBM) contributes to the failure of single-agent immunotherapies in clinical trials. In this study, we evaluated the efficacy of locally delivered combination immunotherapy in a poorly immunogenic murine GBM model. METHODS Immunomodulators used in these studies included: IL-15 and IL-7 (T cell activation), LIGHT (T cell tumor infiltration), FLT3L (dendritic cell maturation/proliferation), a surface T cell engager (T cell killing of tumor cells), and a bispecific PD-L1/T cell engager (T cell killing targeted to PD-L1-expressing cells). We first assessed T cell-mediated cytotoxicity in vitro against SB28, a poorly immunogenic murine GBM cell line, after expressing these immunomodulators in combination. Next, tumor growth inhibition in vivo was evaluated in syngeneic C57BL/6 mice, initially by establishment of intracranial tumors with pre-transduced SB28 cells, and subsequently by delivering these immunomodulators to pre-established naïve SB28 tumors using neural stem cells (NSCs) and retroviral replicating vectors (RRV). RESULTS SB28 cells transduced with immunomodulators activated dose-dependent T cell-mediated cytotoxicity in vitro. Mice with pre-transduced intracranial SB28 gliomas showed significantly longer survival (minimum survival: 60 days, long-term survival in 57% of mice) vs. control mice (median survival: 20 days) (p&lt; 0.001), and significantly increased tumor infiltration of CD4+ and CD8+ T cells. NSC- and RRV-mediated immunomodulator delivery to pre-established SB28 gliomas also resulted in significantly increased survival of treated mice vs. controls (median survival: 31 days vs. 22 days, p&lt; 0.001). Immunomodulator-treated tumors again showed significantly increased infiltration of CD4+ and CD8+ T cells, along with decreased CD11b+ cell infiltration. CONCLUSIONS A novel combination therapy for GBM immunotherapy activated T cell killing of SB28 GBM cells in vitro and achieved a significant survival benefit in vivo, associated with anti-tumor alterations to the GBM tumor microenvironment. Further studies to optimize the efficiency of combinatorial immunomodulator delivery are currently underway.


2021 ◽  
Author(s):  
Jinhyuk F Chung ◽  
Zhisheng Her ◽  
Wai Mun Kong ◽  
Qingfeng Chen

Immune checkpoint inhibitors opened a new horizon in cancer therapy by enabling durable and complete responses in patients, but their wider application against general solid cancers has been hampered by the lack of a broadly acting anti-cancer immune response initiating agents. Parthanatos is a previously unexplored immunogenic programmed necrosis mechanism that is triggered by the hyperactivation of PARP DNA repair and executed by an efficient DNA-fragmentation mechanism. We developed a proprietary macromolecular zinc complex agent C010DS-Zn that efficiently induced parthanatos against 4T1 murine cancer cells in vitro, which was characterized as PARP-mediated necrotic death with massive DNA damages. Ex vivo screening of its cytotoxicity against a panel of 53 low-passage human solid cancer PDX tumor fragments demonstrated its consistent delivery of characteristically DNA-damaging cell death that was unseen in the corresponding apoptosis positive controls. Further characterization of its in vivo treatment effects versus the immunosuppressive 4T1-Balb/c and immunogenic CT26-Balb/c syngeneic cancer models showed that sufficiently high intravenous C010DS-Zn treatments led to robust initiation of the tumor-suppressed antitumor immune compartments such as T cells and macrophages. At lower non-anticancer doses, C010DS-Zn treatment still led to significantly reduced macrophage content and inflammation in the 4T1 tumor, suggesting its potential utility against macrophage-mediated inflammations such as those seen in MAS or COVID19. Given the observation of its low serum bioavailability in a rat pharmacokinetic study, these results suggest potential development opportunities for C010DS-Zn to become a widely applicable immune initiation agent with chemo-like broad applicability upon its pharmacokinetic improvements.


Sign in / Sign up

Export Citation Format

Share Document