scholarly journals Evaluation of tumor immunity after administration of conditionally replicative adenoviral vector in canine osteosarcoma patients

2019 ◽  
Author(s):  
Payal Agarwal ◽  
Elizabeth A Gammon ◽  
Maninder Sandey ◽  
Stephanie S Lindley ◽  
Jey W Koehler ◽  
...  

Abstract Background: Osteosarcoma is one among the most common neoplasms in dogs. Current treatments include leg-amputation or limb-sparing techniques along with chemotherapy that show limited efficacy and fail to prevent metastasis. Conditionally replicative adenoviruses (CRAd) are gene therapy tools that replicate exclusively in targeted tumor cells, causing a cytopathic effect and in turn release of new virus particles to infect additional cells. We proposed that OC-CAVE1 (CAV2 with the E1A promoter replaced with the osteocalcin promotor) may also enhance existing immunity against tumors by overcoming immune tolerance via exposure of new epitopes and cytokine signaling. A previous study indicated administration of OC-CAVE1 was safe and showed specific replication in osteosarcoma cells. Results: We enrolled eleven client-owned dogs with spontaneously occurring osteosarcomas in a pilot clinical-trial. All dogs were injected with OC-CAVE1 following amputation of the affected limb or limb-sparing surgery. Dogs were monitored for viremia and viral shedding. There was minimal virus shedding in urine and feces by the 6th day and no virus was present in blood after 4 weeks. CAV-2 antibody-titers increased in all of the patients, post-CRAd injection. OC-CAVE1 injections did not result in a statistically significant increase in life span, although 2 dogs in the study achieved survival times in excess of 1 year. Immunological assays were performed to monitor 1) humoral response against tumors, 2) levels of circulatory CD11c+ cells, 3) levels of regulatory T cells, and 4) cytotoxic activity of tumor specific T cells against autologous tumor cells between pre-CRAd administration and 4 weeks post-CRAd administration samples. Western blots indicated an increased humoral immune response against pre-existing antigens in some dogs, but flow cytometry did not support this apparent increase. Cytotoxic T-cell activity did not increase in a consistent pattern. Levels of circulatory regulatory T-cells did decrease statistically in all patients. Conclusions: Administration of the CRAd OC-CAVE1 resulted in alteration of some immune response parameters but did not appear to result in increased survival duration. Weak replication of OC-CAVE1 in metastatic cells and delay of chemotherapy following CRAd treatment may contribute to the lack of improvement in survival time of the clinical patients.

2011 ◽  
Vol 2011 ◽  
pp. 1-10 ◽  
Author(s):  
Uri Galili

Anti-Gal is the most abundant antibody in humans, constituting 1% of immunoglobulins. Anti-Gal binds specificallyα-gal epitopes (Galα1-3Galβ1-4GlcNAc-R). Immunogenicity of autologous tumor associated antigens (TAA) is greatly increased by manipulating tumor cells to expressα-gal epitopes and bind anti-Gal. Glycolipids withαgal epitopes (α-gal glycolipids) injected into tumors insert into the tumor cell membrane. Anti-Gal binding to the multiple α-gal epitopesde novopresented on the tumor cells results in targeting of these cells to APC via the interaction between the Fc portion of the bound anti-Gal and Fcγ; receptors on APC. The APC process and present immunogenic TAA peptides and thus, effectively activate tumor specific CD4+ helper T cells and CD8+ cytotoxic T cells which destroy tumor cells in micrometastases. The induced immune response is potent enough to overcome immunosuppression by Treg cells. A phase I clinical trial indicated thatα-gal glycolipid treatment has no adverse effects. In addition to achieving destruction of micrometastases in cancer patients with advance disease,α-gal glycolipid treatment may be effective as neo-adjuvant immunotherapy. Injection ofα-gal glycolipids into primary tumors few weeks prior to resection can induce a protective immune response capable of destroying micrometastases expressing autologous TAA, long after primary tumor resection.


2003 ◽  
Vol 77 (20) ◽  
pp. 10872-10880 ◽  
Author(s):  
Stéphane Vigouroux ◽  
Eric Yvon ◽  
Hans-Joachim Wagner ◽  
Ettore Biagi ◽  
Gianpietro Dotti ◽  
...  

ABSTRACT In mice, activation of the Notch pathway in T cells by antigen-presenting cells overexpressing Notch ligands favors differentiation of regulatory T lymphocytes responsible for antigen-specific tolerance. To determine whether this mechanism operates in human T cells, we used Epstein-Barr virus-positive lymphoblastoid cell lines (EBV-LCL) as our (viral) antigen-presenting cells and overexpressed the Notch ligand Jagged-1 (EBV-LCL J1) by adenoviral transduction. The EBV-LCL J1s were cocultured with autologous T cells, and the proliferative and cytotoxic responses to EBV antigens were measured. Transduction had no effect on EBV-LCL expression of major histocompatibility complex (MHC) antigens or of costimulatory molecules CD80, CD86, and CD40. However, we observed a 35% inhibition of proliferation and a >65% reduction in cytotoxic-T-cell activity, and interleukin 10 production was increased ninefold. These EBV-LCL J1-stimulated T lymphocytes act as antigen-specific regulatory cells, since their addition to fresh autologous T cells cultured with autologous nontransduced EBV-LCL cells significantly inhibited both proliferation and cytotoxic effector function. Within the inhibitory population, CD4+CD25+ and CD8+CD25− T cells had the greatest activity. This inhibition appears to be antigen-specific, since responses to Candida and cytomegalovirus antigens were unaffected. Hence, transgenic expression of Jagged-1 by antigen-presenting cells can induce antigen-specific regulatory T cells in humans and modify immune responses to viral antigens.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3172-3172
Author(s):  
Aaron Foster ◽  
Fatma Okur ◽  
Gianpietro Dotti ◽  
Jessica Shafer ◽  
An Lu ◽  
...  

Abstract Side-population (SP) cells, defined on the basis of their ability to rapidly expel Hoechst dye, may be precursor cells for normal and malignant tissues and are more resistant to chemotherapy. We have now characterized a malignant SP subset in the peripheral blood of B-CLL patients and show how vaccination with CD40L/IL-2 expressing autologous tumor cells can induce a T cell immune response that reduces or eliminates this tumor subpopulation. Hoechst 33342 staining combined with CD5/CD19 antibody labeling of peripheral blood mononuclear cells showed a distinct CD5+CD19+ SP phenotype (median=0.22%, range; 0.2–2.17%) in 85% (18/21) of B-CLL patients (Stage I–III). We sorted SP and non-SP (NSP) cells and extracted RNA and performed a comprehensive gene expression analysis by microarray (n=9 donor SP and NSP pairs), finding 85 genes overexpressed (>2-fold log ratio) in SP compared to NSP, and 228 genes higher in NSP versus SP. The analysis showed that compared to NSP cells from the same patient, the SP subset consistently and significantly overexpressed genes associated with cellular activation, proliferation and survival including eIF4A1 (p<.05), Wnt10a (p<.05), Bcl2L12 (p<.05) and Rel A (p=.07), a difference confirmed by qPCR. Conversely, NSP significantly overexpressed GSK3β (p<.05) compared to SP. CD40 mediated activation of B-CLL tumor cells further enhanced relative expression of these genes in the SP fraction, and induced overexpression of the proliferation-associated genes Ki-67 and PCNA in SP compared to NSP cells by 8-fold and 132-fold, respectively. We found SP cells were more resistant to exposure to chemotherapeutic agents including 50 mM fludarabine than NSP. Analysis of results in patients who had received an autologous B-CLL cell tumor vaccine expressing CD40L and IL-2, however, showed that B-CLL SP cells were vulnerable to a T cell mediated immune response. Hence, 3–8 subcutaneous injections CD40L/IL-2 B-CLL cells had no immediate effect on total B-CLL cell numbers in the peripheral blood, but Hoechst analysis showed a disappearance of SP cells in 7/8 immunized patients, associated with a rise in B-CLL-reactive T cells. CD8+ T cell clones isolated and expanded with CD40-activated autologous B-CLL tumor cells specifically responded to autologous SP cells, suggesting that SP depletion is mediated vaccine induced B-CLL-specific T cells. In 6/7 patients SP cells returned within a few weeks of completing the course of vaccination, coincident with a decline in T cell immunity. In one patient, however, immunity was longer lived and in this patient control of SP B-CLL cells persisted for more that 12 months. In this individual there was a delayed (by 6 months), but progressive disappearance of the bulk (non-SP) tumor cells. These results are consistent with the existence of an SP tumor precursor population of B-CLL, that can be targeted by immune T cells in vivo, and removal of which leads to later disappearance of their non-SP progeny.


Author(s):  
Yuanliang Yan ◽  
Shuangshuang Zeng ◽  
Zhicheng Gong ◽  
Zhijie Xu

AbstractGliomas, especially glioblastomas, represent one of the most aggressive and difficult-to-treat human brain tumors. In the last few decades, clinical immunotherapy has been developed and has provided exceptional achievements in checkpoint inhibitors and vaccines for cancer treatment. Immunization with cellular vaccines has the advantage of containing specific antigens and acceptable safety to potentially improve cancer therapy. Based on T cells, dendritic cells (DC), tumor cells and natural killer cells, the safety and feasibility of cellular vaccines have been validated in clinical trials for glioma treatment. For TAA engineered T cells, therapy mainly uses chimeric antigen receptors (IL13Rα2, EGFRvIII and HER2) and DNA methylation-induced technology (CT antigen) to activate the immune response. Autologous dendritic cells/tumor antigen vaccine (ADCTA) pulsed with tumor lysate and peptides elicit antigen-specific and cytotoxic T cell responses in patients with malignant gliomas, while its pro-survival effect is biased. Vaccinations using autologous tumor cells modified with TAAs or fusion with fibroblast cells are characterized by both effective humoral and cell-mediated immunity. Even though few therapeutic effects have been observed, most of this therapy showed safety and feasibility, asking for larger cohort studies and better guidelines to optimize cellular vaccine efficiency in anti-glioma therapy.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii214-ii214
Author(s):  
Anupam Kumar ◽  
Katharine Chen ◽  
Claudia Petritsch ◽  
Theodore Nicolaides ◽  
Mariarita Santi-Vicini ◽  
...  

Abstract The determinants of the tumor-associated immune response in brain tumors are poorly understood. Using tumor samples from two molecularly distinct subtypes of lower grade glioma, MAPK-driven glioma with biallelic inactivation of CDKN2A (n=30) and IDH-mutant, 1p/19q-intact astrocytoma (n=29), we demonstrate qualitative and quantitative differences in the tumor-associated immune response and we investigate the molecular mechanisms involved. Histologically the MAPK-driven gliomas were comprised of pleomorphic xanthoastrocytoma (PXA) (n=11) and anaplastic PXA (n=19). Seven patients had paired samples from two sequential surgeries. Immune cell populations and their activity were determined by quantitative multiplex immunostaining and Digital Spatial Profiling and gene expression was analyzed by Nanostring. Functional studies were performed using established cell lines and two new patient-derived lines from MAPK-driven LGGs. MAPK-driven tumors exhibited an increased number of CD8+ T cells and tumor-associated microglial/macrophage (TAMs), including CD163+ TAMs, as compared to IDH-mutant astrocytoma. In contrast, IDH-mutant tumors had increased FOXP3+ immunosuppressive T regulatory cells. Transcriptional and protein level analyses in MAPK-driven tumors suggested an active cytotoxic T cell response with robust expression of granzyme B, present on 27% of CD8+ T cells, increased MHC class I expression, and altered cytokine profiles. Interestingly, MAPK-driven tumors also had increased expression of immunosuppressive molecules, including CXCR4, PD-L1, and VEGFA. Expression differences for cell surface and secreted proteins were confirmed in patient-derived tumor lines and functional relationships between altered chemokine expression and immune cell infiltration was investigated. Our data provide novel insights into the immune contexture of MAPK driven LGGs and suggest MAPK driven gliomas with biallelic inactivation of CDKN2A may be particularly vulnerable to immunotherapeutic modulation


BMC Cancer ◽  
2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Satu Salmi ◽  
Anton Lin ◽  
Benjamin Hirschovits-Gerz ◽  
Mari Valkonen ◽  
Niina Aaltonen ◽  
...  

Abstract Background FoxP3+ Regulatory T cells (Tregs) and indoleamine-2,3-dioxygenase (IDO) participate in the formation of an immunosuppressive tumor microenvironment (TME) in malignant cutaneous melanoma (CM). Recent studies have reported that IDO expression correlates with poor prognosis and greater Breslow’s depth, but results concerning the role of FoxP3+ Tregs in CM have been controversial. Furthermore, the correlation between IDO and Tregs has not been substantially studied in CM, although IDO is known to be an important regulator of Tregs activity. Methods We investigated the associations of FoxP3+ Tregs, IDO+ tumor cells and IDO+ stromal immune cells with tumor stage, prognostic factors and survival in CM. FoxP3 and IDO were immunohistochemically stained from 29 benign and 29 dysplastic nevi, 18 in situ -melanomas, 48 superficial and 62 deep melanomas and 67 lymph node metastases (LNMs) of CM. The number of FoxP3+ Tregs and IDO+ stromal immune cells, and the coverage and intensity of IDO+ tumor cells were analysed. Results The number of FoxP3+ Tregs and IDO+ stromal immune cells were significantly higher in malignant melanomas compared with benign lesions. The increased expression of IDO in melanoma cells was associated with poor prognostic factors, such as recurrence, nodular growth pattern and increased mitotic count. Furthermore, the expression of IDO in melanoma cells was associated with reduced recurrence˗free survival. We further showed that there was a positive correlation between IDO+ tumor cells and FoxP3+ Tregs. Conclusions These results indicate that IDO is strongly involved in melanoma progression. FoxP3+ Tregs also seems to contribute to the immunosuppressive TME in CM, but their significance in melanoma progression remains unclear. The positive association of FoxP3+ Tregs with IDO+ melanoma cells, but not with IDO+ stromal immune cells, indicates a complex interaction between IDO and Tregs in CM, which demands further studies.


1991 ◽  
Vol 174 (4) ◽  
pp. 891-900 ◽  
Author(s):  
S M Friedman ◽  
M K Crow ◽  
J R Tumang ◽  
M Tumang ◽  
Y Q Xu ◽  
...  

While all known microbial superantigens are mitogenic for human peripheral blood lymphocytes (PBL), the functional response induced by Mycoplasma arthritidis-derived superantigen (MAM) is unique in that MAM stimulation of PBL consistently results in T cell-dependent B cell activation characterized by polyclonal IgM and IgG production. These immunostimulatory effects of MAM on the humoral arm of the human immune system warranted a more precise characterization of MAM-reactive human T cells. Using an uncloned MAM reactive human T cell line as immunogen, we have generated a monoclonal antibody (mAb) (termed C1) specific for the T cell receptor V beta gene expressed by the major fraction of MAM-reactive human T cells, V beta 17. In addition, a V beta 17- MAM-reactive T cell population exists, assessed by MAM, induced T cell proliferation and cytotoxic T cell activity. mAb C1 will be useful in characterizing the functional properties of V beta 17+ T cells and their potential role in autoimmune disease.


Sign in / Sign up

Export Citation Format

Share Document