scholarly journals OS7 - 145 Combination Immunotherapy for Glioma: Beyond PD 1 Inhibition

Author(s):  
W. Curry

Programmed Cell Death – 1 (PD1) inhibition activates tumor-specific T-lymphocytes and is an effective clinical therapy against some cancers. Preclinical data regarding immune checkpoint inhibitors against malignant glioma is scant, and interim analyses of clinical trials suggest modest effect in patients as single agents. We examined PD-1 inhibition in murine glioblastoma models in combination with other immunomodulatory agents. Methods – Syngeneic glioma tumors (GL261 and CT2A) were implanted intracranially in C57/Bl6 mice. In separate experiments, PD-1 inhibition was combined with antibody blockade of t-cell immunoglobulin and mucin protein (TIM3), ligation of OX40 on T-lymphocytes, or vaccination with irradiated GM-CSF expressing tumor cells. Systemic antitumor immunity and tumor infiltrating lymphocytes were analyzed by ELISPOT assay and flow cytometry, respectively. Results - In both syngeneic glioma models, day 3,6, and 9 systemic delivery of a monoclonal antibody against PD-1 led to increased survival vs. controls. In animals with GL261 intracranial tumors, survival was improved by combination of PD-1 blockade with subcutaneous injection of irradiated GM-CSF expressing GL261 tumor cells, with antibody blockade of t-cell immunoglobulin and mucin protein 3 (TIM3), or binding of OX40 on T-lymphocytes by an activating antibody. In most cases, ELISPOT analyses demonstrated enhanced Th1 immunity by combination immunotherapies. Vaccination was associated with an increased intratumoral CD8+ T lymphocyte / FoxP3+ T lymphocyte ratio. Conclusion –Blockade of PD-1 on T lymphocytes in glioma-bearing mice is active. Both antitumor immunity and survival can be enhanced by combination of PD-1 inhibition with agents that activate antitumor immunity by complementary mechanisms.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3246-3246
Author(s):  
Yosuke Yokota ◽  
Hiroyuki Inoue ◽  
Ayumi Watanabe ◽  
Chika Sakamoto ◽  
Megumi Narusawa ◽  
...  

Abstract Abstract 3246 GM-CSF (Granulocyte macrophage-colony stimulating factor) gene-transduced leukemia cell vaccine has been reported to be promising approach to enhance antitumor immune response in leukemia patients. The enhancement of this specific antitumor immunity is considered to be clinically beneficial to maintain complete remission for long time after chemotherapy without stem cell transplantion. Leukotriene B4 (LTB4) is an extremely potent lipid inflammatory mediator derived from membrane phospholipids, known to recruit and activate leukocytes including neutrophils, mediated by the same class of receptors, the GPCR (G-protein coupled receptor) superfamily, BLT1 and BLT2. So far, the role of LTB4 in tumor immunology is not well known. Previously, we demonstrated that the GM-CSF gene transduction into murine monocytic leukemia cell line of WEHI3B (WGM) eliminated the tumorigenicity in subcutaneous challenge model using wild type (WT) BALB/c mice. At day 50 after the challenge, we rechallenged WEHI3B cells into the opposite flank of both WT and BLT1-KO mice which had rejected WGM cells (WT/WGM, KO/WGM, respectively). Intriguingly, more KO/WGM mice re-rejected the rechallenged WEHI3B cells and showed significantly prolonged survival compared with WT/WGM mice. To clarify this unique mechanism of the long-lasting antitumor effects observed in KO/WGM mice, we performed following immunological assays. Our in vivo immune cell depletion assays (NK, CD4+ T, CD8+ T cell) showed that KO/WGM mice treated with anti-CD4 antibody displayed rather enhanced tumor growth compared with WT/WGM mice. Thus, we next compared the rates of different subsets of memory T cells, so called memory stem T cell subsets (TSCM), central memory T cell subsets (TCM) and effector memory T cell subsets (TEM) in TDLNs (Tumor draining lymph nodes) between WT/WGM mice and KO/WGM mice at the day 46 after tumor challenge. Expectedly, results showed that TDLNs harvested from KO/WGM mice exhibited higher subset ratios of CD44+CD62L+ (TCM) cell to CD4+ T cells as well as CD44+CD62L− or (TEM) cell to CD4+ T cells than those from WT/WGM mice. In the case of use of CD122 for memory marker, similar results were obtained. Of note, TDLNs from KO/WGM mice exhibited increased ratios of CD44+CD122+CD62L− (TSCM) cell. In addition, the cell numbers of immunosuppressive CD3+ CD4+ PD-1+, CD3+ CD4+ GITR+ and CD3+ CD4+ CTLA4+ cells were reduced in both TDLNs and spleen derived from KO/WGM mice compared with those from WT/WGM mice. Furthermore, our results of CBA (Cytometric Bead Array) assay using splenocytes harvested from day 2 to day 15 showed that the Th2 cytokine production level of IL-4 and IL-5 from splenocyte harvested from KO/WGM mice were higher than those from WT/WGM mice. In regard to IL-2 and IFN-g (Th1 cytokine), the production level at both day 7 and day10 from splenocytes harvested from KO/WGM mice were also higher than those from WT/WGM mice, implicating that loss of LTB4/BLT1 signaling promotes systemic activation of both tumor antigens specific Th1 and Th2 CD4+ T cell subsets. Finally, our flow cytometric analyses demonstrated that exogenously GM-CSF-driven upregulated MFI of CD40+, CD80+ and CD86+ DCs in TDLNs from KO/WGM mice were further significantly increased compared with those from WT/WGM mice (p<0.05), and more numerous number of CD86+ DCs that had phagocytosed TAAs of WEHI3B cells was detected in TDLNs harvested from KO/WGM mice than that from WT/WGM mice. In conclusion, we for the first time demonstrate that loss of LTB4/BLT1 axis can sustain long-lasting memory CD4+ T cell-dependent antitumor immunity against syngeneic tumor cells long after GM-CSF triggering tumor rejection, allowing us to expect the possibility that the strategy of blocking LTB4/BLT1signaling may be useful to maintain antitumor immunity induced by GM-CSF gene-transduced leukemia cell vaccine in clinical settings. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 9 (1) ◽  
pp. e001460 ◽  
Author(s):  
Xiuting Liu ◽  
Graham D Hogg ◽  
David G DeNardo

The clinical success of immune checkpoint inhibitors has highlighted the central role of the immune system in cancer control. Immune checkpoint inhibitors can reinvigorate anti-cancer immunity and are now the standard of care in a number of malignancies. However, research on immune checkpoint blockade has largely been framed with the central dogma that checkpoint therapies intrinsically target the T cell, triggering the tumoricidal potential of the adaptive immune system. Although T cells undoubtedly remain a critical piece of the story, mounting evidence, reviewed herein, indicates that much of the efficacy of checkpoint therapies may be attributable to the innate immune system. Emerging research suggests that T cell-directed checkpoint antibodies such as anti-programmed cell death protein-1 (PD-1) or programmed death-ligand-1 (PD-L1) can impact innate immunity by both direct and indirect pathways, which may ultimately shape clinical efficacy. However, the mechanisms and impacts of these activities have yet to be fully elucidated, and checkpoint therapies have potentially beneficial and detrimental effects on innate antitumor immunity. Further research into the role of innate subsets during checkpoint blockade may be critical for developing combination therapies to help overcome checkpoint resistance. The potential of checkpoint therapies to amplify innate antitumor immunity represents a promising new field that can be translated into innovative immunotherapies for patients fighting refractory malignancies.


2003 ◽  
Vol 21 (17) ◽  
pp. 3343-3350 ◽  
Author(s):  
Robert Soiffer ◽  
F. Stephen Hodi ◽  
Frank Haluska ◽  
Ken Jung ◽  
Silke Gillessen ◽  
...  

Purpose: Vaccination with irradiated, autologous melanoma cells engineered to secrete granulocyte-macrophage colony-stimulating factor (GM-CSF) by retroviral-mediated gene transfer generates potent antitumor immunity in patients with metastatic melanoma. Further clinical development of this immunization scheme requires simplification of vaccine manufacture. We conducted a phase I clinical trial testing the biologic activity of vaccination with irradiated, autologous melanoma cells engineered to secrete GM-CSF by adenoviral-mediated gene transfer.Patients and Methods: Excised metastases were processed to single cells, transduced with a replication-defective adenoviral vector encoding GM-CSF, irradiated, and cryopreserved. Individual vaccines were composed of 1 × 106, 4 × 106, or 1 × 107tumor cells, depending on overall yield, and were injected intradermally and subcutaneously at weekly and biweekly intervals.Results: Vaccines were successfully manufactured for 34 (97%) of 35 patients. The average GM-CSF secretion was 745 ng/106cells/24 hours. Toxicities were restricted to grade 1 to 2 local skin reactions. Eight patients were withdrawn early because of rapid disease progression. Vaccination elicited dense dendritic cell, macrophage, granulocyte, and lymphocyte infiltrates at injection sites in 19 of 26 assessable patients. Immunization stimulated the development of delayed-type hypersensitivity reactions to irradiated, dissociated, autologous, nontransduced tumor cells in 17 of 25 patients. Metastatic lesions that were resected after vaccination showed brisk or focal T-lymphocyte and plasma cell infiltrates with tumor necrosis in 10 of 16 patients. One complete, one partial, and one mixed response were noted. Ten patients (29%) are alive, with a minimum follow-up of 36 months; four of these patients have no evidence of disease.Conclusion: Vaccination with irradiated, autologous melanoma cells engineered to secrete GM-CSF by adenoviral-mediated gene transfer augments antitumor immunity in patients with metastatic melanoma.


2021 ◽  
Author(s):  
Luuk van Hooren ◽  
Alessandra Vaccaro ◽  
Mohanraj Ramachandran ◽  
Konstantinos Vazaios ◽  
Sylwia Libard ◽  
...  

AbstractGliomas are brain tumors characterized by immunosuppression. Immunostimulatory agonistic CD40 antibodies (αCD40) are in clinical development for solid tumors but are yet to be evaluated for glioma. Here, systemic delivery of αCD40 led to cytotoxic T cell dysfunction and impaired the response to immune checkpoint inhibitors in preclinical glioma models. This was associated with an accumulation of suppressive CD11b+ B cells. However, αCD40 also induced tertiary lymphoid structures (TLS). In human glioma, TLS correlated with increased T cell infiltration indicating enhanced immune responses. Our work unveils the pleiotropic effects of αCD40 therapy in glioma, which is of high clinical relevance.


2006 ◽  
Vol 19 (1) ◽  
pp. 205873920601900 ◽  
Author(s):  
S. Staibano ◽  
M. Mascolo ◽  
F. Tranfa ◽  
G. Salvatore ◽  
C. Mignogna ◽  
...  

Experimental and clinical evidence indicate that immunological mechanisms might be important in the clinical course of uveal malignant melanoma (UMM). We analyzed the amount and phenotype of tumor infiltrating lymphocytes (TIL) and the expression of the apoptosis-inducing molecule Fas and its ligand, FasL, on tumor cells and TIL in a selected series of UMM with the aim to establish if a correlation between their expression and the clinical behavior of UMM exists. TIL phenotype and Fas/FasL expression were evaluated by immunohistochemistry in 61 cases of formalin-fixed, paraffin-embedded UMM. Results were compared with the follow-up data of patients. Most of the UMM showed a prevalence of CD8+ CD3+ T lymphocytes, or CD4+ and CD8+ cells in equal amounts. UMM showed a variable expression of FasL, ranging from 0 to > 40% of neoplastic cells. Fas was always expressed in TIL, although with a variable extent. A subgroup of UMM showed in TIL a strongly reduced or even absent expression of TCR ζ-chain, involved in activation of T-lymphocytes. This subgroup was characterized by a worse outcome. We hypothesized that an impaired cytotoxic immune response due to the loss of the ζ-chain expression plays a primary role in the biological course of UMM. Our results indicate that the overcoming of the impairment of TCR function may represent a prerequisite for the development of new therapeutic strategies for managing UMM, suggesting that elimination of tumor cells may be possible by activation of cytotoxic cells present within ocular melanomas.


Cancers ◽  
2020 ◽  
Vol 12 (11) ◽  
pp. 3397
Author(s):  
Leyre Silva ◽  
Josune Egea ◽  
Lorea Villanueva ◽  
Marta Ruiz ◽  
Diana Llopiz ◽  
...  

Therapies based on immune checkpoint inhibitors (ICPI) have yielded promising albeit limited results in patients with hepatocellular carcinoma (HCC). Vaccines have been proposed as combination partners to enhance response rates to ICPI. Thus, we analyzed the combined effect of a vaccine based on the TLR4 ligand cold-inducible RNA binding protein (CIRP) plus ICPI. Mice were immunized with vaccines containing ovalbumin linked to CIRP (OVA-CIRP), with or without ICPI, and antigen-specific responses and therapeutic efficacy were tested in subcutaneous and orthotopic mouse models of liver cancer. OVA-CIRP elicited polyepitopic T-cell responses, which were further enhanced when combined with ICPI (anti-PD-1 and anti-CTLA-4). Combination of OVA-CIRP with ICPI enhanced ICPI-induced therapeutic responses when tested in subcutaneous and intrahepatic B16-OVA tumors, as well as in the orthotopic PM299L HCC model. This effect was associated with higher OVA-specific T-cell responses in the periphery, although many tumor-infiltrating lymphocytes still displayed an exhausted phenotype. Finally, a new vaccine containing human glypican-3 linked to CIRP (GPC3-CIRP) induced clear responses in humanized HLA-A2.01 transgenic mice, which increased upon combination with ICPI. Therefore, CIRP-based vaccines may generate anti-tumor immunity to enhance ICPI efficacy in HCC, although blockade of additional checkpoint molecules and immunosuppressive targets should be also considered.


2020 ◽  
Vol 217 (12) ◽  
Author(s):  
Isabelle C. Arnold ◽  
Mariela Artola-Boran ◽  
Alessandra Gurtner ◽  
Katrin Bertram ◽  
Michael Bauer ◽  
...  

The depletion of eosinophils represents an efficient strategy to alleviate allergic asthma, but the consequences of prolonged eosinophil deficiency for human health remain poorly understood. We show here that the ablation of eosinophils severely compromises antitumor immunity in syngeneic and genetic models of colorectal cancer (CRC), which can be attributed to defective Th1 and CD8+ T cell responses. The specific loss of GM-CSF signaling or IRF5 expression in the eosinophil compartment phenocopies the loss of the entire lineage. GM-CSF activates IRF5 in vitro and in vivo and can be administered recombinantly to improve tumor immunity. IL-10 counterregulates IRF5 activation by GM-CSF. CRC patients whose tumors are infiltrated by large numbers of eosinophils also exhibit robust CD8 T cell infiltrates and have a better prognosis than patients with eosinophillow tumors. The combined results demonstrate a critical role of eosinophils in tumor control in CRC and introduce the GM-CSF–IRF5 axis as a critical driver of the antitumor activities of this versatile cell type.


1987 ◽  
Vol 165 (3) ◽  
pp. 664-676 ◽  
Author(s):  
M L Plunkett ◽  
M E Sanders ◽  
P Selvaraj ◽  
M L Dustin ◽  
T A Springer

CD2, also known as LFA-2, T11, and the E rosette receptor, is a T lymphocyte surface protein functionally important in adhesion to target cells and T cell triggering. LFA-3 is a widely distributed cell surface protein that functions in adhesion on target cells. We find that LFA-3 is expressed on human E, and that CD2 is a receptor for LFA-3 that mediates T cell adhesion to human E. Pretreatment of T lymphocytes with CD2 mAb or of E with LFA-3 mAb inhibits rosetting. Purified CD2 molecules bind to human E and inhibit rosetting. 125I-CD2 binding to E is inhibited by LFA-3 mAb; reciprocally, binding of LFA-3 mAb to human E is inhibited by pretreatment with purified CD2. Higher concentrations of CD2 aggregate human E; aggregation is inhibited by mAb to LFA-3.


AIDS ◽  
2002 ◽  
Vol 16 (11) ◽  
pp. 1459-1465 ◽  
Author(s):  
Thomas W. McCloskey ◽  
Viraga Haridas ◽  
Rajendra Pahwa ◽  
Savita Pahwa

Sign in / Sign up

Export Citation Format

Share Document