CMV-Specific T-Cells Are More Effective Than WT-1-Specific T-Cells in Eradicating Clonogenic Tumor Cells Co-Expressing CMVpp65 and WT-1 in-Vivo and Both Types of T-Cells Are Functionally Augmented in the Presence of IL-15/IL-15Rα

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 646-646 ◽  
Author(s):  
Aisha Hasan ◽  
Dana Bakalar ◽  
Annamalai Selvakumar ◽  
Gloria C Koo ◽  
Ekaterina Doubrovina ◽  
...  

Abstract Abstract 646 Adoptively transferred virus or tumor antigen-specific T-cells have demonstrated efficacy in initial clinical trials, but while clearance of viral infection is sustained, responses to tumor-specific T-cells are usually short-lived. Therefore, current efforts are focused on distinguishing attributes of virus-specific T-cells that contribute to their persistence and formulating strategies to sustain anti-tumor effects of T-cell (TC) therapies. We have developed an in-vivo model to compare the relative efficacy of T-cells specific for a tumor antigen (WT-1) versus T-cells specific for a viral antigen (CMVpp65) using human colon carcinoma cells that express the oncofetal protein WT-1 which were transduced to co-express CMVpp65 (WT-1[+] cocapp65) as a surrogate system. Groups of 6 NOD/Scid-IL2Rgc-KO/J mice (NSG) were each subcutaneously injected with 3 × 105 WT-1 [+] cocapp65 on the R flank. Each animal was also injected with 3 × 105 cells from a WT-1[+] ovarian carcinoma cell line (SKOV3-A2) on the L shoulder to compare the efficacy of WT-1 specific T-cells (WT1-CTLs) against different WT-1 [+] tumor cell types. Expression of WT-1 protein is lower in SKOV3-A2 than in cocapp65 cells. Both tumors are HLA A0201[+] and were transduced to express a GFP-firefly luciferase gene. T-cells were administered intravenously 5 days after tumor injection to enable vascularization and tumor growth was quantitated using bioluminescence. These experiments evaluated (1) the relative capacity of CMVpp65 specific T-cells (CMV-CTLs) versus WT-1 CTLs to eradicate WT1[+] cocapp65 cells that co-express a viral and tumor antigen (2) the relative efficacy of WT-1 CTLs against 2 different HLA A0201 [+] WT-1 expressing tumors; an ovarian carcinoma and a colon carcinoma, and (3) the contribution of IL-15/15Rα complex in augmenting the efficacy of antigen specific T-cells by using intraperitoneally (i.p) injected Baf-3 cells transduced to express human IL-15/15Rα complex. The treatment groups were as follows: (1) Control – no T-cells + IL-2 (2000 U) (2) Control – no T-cells + IL-15/IL-15Rα (5 × 106 baf-3 cells) (3) WT1 CTLs + IL-2 (2000 U) (4) CMV-CTLs + IL-2 (2000 U) (5) WT1 CTLs + IL-15/IL-15Rα (5 × 106 baf-3 cells) (6) CMV-CTLs + IL-15/IL-15Rα (5 × 106 baf-3 cells). IL-2 and irradiated baf-3 cells were administered intraperitoneally twice weekly. When the doses of antigen specific interferon gamma (IFNg) [+] T-cells were equivalent in the infused CMVpp65 and WT1 specific T-cells, the CMV-CTLs induced greater, and more sustained suppression of the growth of the WT-1[+] cocapp65 cells in-vivo than the WT-1 CTLs (Fig.1). The anti-tumor activity of the WT-1 CTLs was greater against WT-1[+] cocapp65 than against the WT-1[+] ovarian carcinoma (SKOV3-A2), potentially reflecting the higher expression of WT-1 in cocapp65. The SKOV3-A2 tumor began to re-grow by 24 days post T-cell infusion approaching the size of control tumors by day 38, while the WT-1[+] cocapp65 still demonstrated slower growth through day 38. The addition of IL-15/15Rα increased the efficacy of the transferred T-cells, the difference being more pronounced for the anti-tumor activity of WT-1 CTLs. Fig. 1 Comparative Efficacy of CMV and WT-1 CTLs against Human Tumor Targets Co-expressing CMVpp65 and WT-1 Fig. 1. Comparative Efficacy of CMV and WT-1 CTLs against Human Tumor Targets Co-expressing CMVpp65 and WT-1 These studies demonstrate that equivalent doses of IFNg[+] WT-1 CTLs can also suppress WT-1[+] cocapp65 tumor xenografts, but are less effective than CMV-CTLs, and that IL-15 supplementation augments the cytotoxic activity of the CTLs in-vitro and enhances the duration of the anti-tumor effects in-vivo. This model permits side by side comparisons of the anti-tumor activity of human T-cells directed against viral and tumor antigens expressed on the same clonogenic human tumor target. Because both responses are directed against the same cells, this model could thereby facilitate identification of the distinguishing features of T-cells specific for viral or tumor antigens as well as differences in the presentation of viral and oncofetal “self” antigens by tumor cells that contribute to disparities in their anti-tumor activity and persistence in-vivo. Disclosures: No relevant conflicts of interest to declare.

2002 ◽  
Vol 195 (11) ◽  
pp. 1397-1406 ◽  
Author(s):  
Helen Y. Wang ◽  
Juhua Zhou ◽  
Kuichun Zhu ◽  
Adam I. Riker ◽  
Francesco M. Marincola ◽  
...  

CD4+ T cells play an important role in orchestrating host immune responses against cancer, particularly by providing critical help for priming and extending the survival of CD8+ T cells. However, relatively little is known about major histocompatibility complex class II–restricted human tumor antigens capable of activating CD4+ T cells. Here, we describe the identification of a mutated fibronectin (FN) as a tumor antigen recognized by human histocompatibility leukocyte antigen-DR2–restricted CD4+ T cells. Deoxyribonucleic acid (DNA) sequencing analysis indicated that this gene contains a mutation that results in the substitution of lysine for glutamic acid and gives rise to a new T cell epitope recognized by CD4+ T cells. Tumor cells harboring the mutant FN resulted in the loss of FN matrix formation and the gain of metastatic potential based on the migration pattern compared with that of tumor cells that express wild-type FN. Additional experiments using cell lines stably expressing the mutated FN cDNA demonstrated that the point mutation in FN was responsible for the loss of FN staining in extracellular matrices and the enhancement of tumor cell migration. These findings represent the first demonstration that a mutated gene product recognized by CD4+ T cells is directly involved in tumor metastasis, which indicates the importance of CD4+ T cells in controlling the spread of tumor cells to distant anatomic sites.


2017 ◽  
Vol 7 (3) ◽  
pp. e1407897 ◽  
Author(s):  
Tiphaine Delaunay ◽  
Mathilde Violland ◽  
Nicolas Boisgerault ◽  
Soizic Dutoit ◽  
Virginie Vignard ◽  
...  

2001 ◽  
Vol 194 (9) ◽  
pp. 1339-1348 ◽  
Author(s):  
Xingluo Liu ◽  
Xue-Feng Bai ◽  
Jing Wen ◽  
Jian-Xin Gao ◽  
Jinqing Liu ◽  
...  

B7H/B7RP (hereby called B7H) is a new member of the B7 family of costimulatory molecules and interacts with inducible costimulatory molecule (ICOS). Its function for CD8 T cells has not been reported. We report here that expression of B7H on the tumor cells reduced tumorigenicity and induced immunity to subsequent challenge with parental tumor cells. The immune protection correlates with an enhanced cytotoxic T lymphocyte (CTL) response against P1A, the major tumor antigen expressed in the J558 tumor. To understand the mechanism of immune protection, we adoptively transferred transgenic T cells specific for tumor antigen P1A into mice that bore P1A-expressing tumors. We found that while the transgenic T cells divided faster in mice bearing the B7H+ tumors, optimal B7H-induced clonal expansion of P1CTL required costimulation by B7–1 and B7–2 on the endogenous host antigen-presenting cells (APCs). Interestingly, when B7H+ and B7H− tumors were coinjected, P1CTL selectively eliminated the B7H+ tumor cells. Moreover, B7H expressed on the tumor cells made them highly susceptible to destruction by CTL in vivo, even if the CTL was administrated into mice with large tumor burdens. Tumors that recurred in the P1CTL-treated mice lost transfected B7H and/or H-2Ld, the class I molecule that presents the P1A peptide. Taken together, our results reveal that B7H costimulates clonal expansion of, and cognate destruction by CD8+ T lymphocytes in vivo.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii91-ii91
Author(s):  
Dalia Haydar ◽  
Zhongzhen Yi ◽  
Haley Houke ◽  
Martine F Roussel ◽  
Chris DeRenzo ◽  
...  

Abstract BACKGROUND We and others have identified B7-H3 (CD276) as a promising target for CAR T-cell-based immunotherapies for pediatric brain tumors. So far, B7-H3-CAR T cells have only been studied in xenograft models for brain tumors, which do not recapitulate the immunosuppressive tumor microenvironment (TME). To overcome this obstacle, we decided to adapt the immune competent GL261 murine glioma model which mimics human disease and host immune barriers. METHODS To evaluate their safety and efficacy, murine B7-H3-CAR T-cells were generated using retroviral particles encoding a 2nd generation B7-H3-CAR with a CD28.z signaling domain. Expansion, persistence, and anti-tumor activity were evaluated in vitro and in vivo. Components of the brain TME were then evaluated using flow cytometry and immunostaining. RESULTS B7-H3-CAR T cells only killed B7-H3+ tumor cells, secreted significant levels of IFNγ and IL-2 in an antigen-dependent manner and expanded an average of 85-fold in repeat stimulation assay with B7-H3+ tumor cells in contrast to control CAR T-cells. In vivo, intratumoral (2x106) or systemic (3x106) injection of syngeneic B7-H3-CAR T-cells into mice with orthotopic GL261 glioma induced complete regression in 60% of treated mice resulting in a significant survival advantage. Mice showed no evidence of acute or long-term toxicities related to CAR T-cell infusions. We confirmed this encouraging safety profile by systemic administration of a high dose (1x107) B7-H3-CAR T-cells and performing histological analyses of all major organs on day 14 post T-cell injection, which showed no notable signs of injury or on-target/off-tumor toxicities. CONCLUSIONS We successfully generated syngeneic B7-H3-CAR T-cells and have demonstrated that these cells have potent anti-tumor activity in the immune competent GL261 glioma model via local or systemic delivery without apparent toxicities. Our study paves the way for future testing of B7-H3-CAR T-cells in early phase clinical studies.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi128-vi129
Author(s):  
Polly Chuntova ◽  
Akane Yamamichi ◽  
Ryosuke Naka ◽  
Yafei Hou ◽  
Hideho Okada

Abstract Despite the recent successes in cancer immunotherapy, brain tumors continue to present a significant challenge due to the unique relationship between the CNS and the immune system. Adoptive cell transfer of chimeric antigen receptor T-cells (CAR-T cells) targets tumor-associated and tumor-specific antigens. Multiple clinical trials testing CAR-T cells in the brain tumor setting have demonstrated the feasibility and safety, but also highlighted the limitations and continued need for treatment improvement. These challenges include recurrence of tumor cells that do not express the target antigens and induction of immuno-regulatory mechanisms, such as PD-L1 expression within the tumor. In this study, we used a murine GBM cell line (SB28), engineered to express the tumor-specific neoantigen human epidermal growth factor receptor variant III, to study the in vivo effects of EGFRvIII-CAR T-cells in a novel fully immunocompetent mouse model. SB28 cells have been shown to recapitulate key characteristics of human GBM, such as aggressive growth, low mutation burden, and resistance to immune checkpoint blockade therapy, and therefore, allow us to investigate many clinically relevant questions and variations in treatment protocol. Intravenous administration of CAR-transduced murine T-cells in EGFRvIII-expressing SB28 tumors showed survival benefit only when preceded by a lymphodepleting regimen. Despite prolonging survival, EGFRvIII-CAR-T cells were not curative, and the recurrent tumors displayed profound antigen loss, similar to post-treatment human tumor samples. Preliminary mechanistic studies revealed upregulation of IFNγ-inducible molecules, such as PD-L1 on tumor cells and tumor-infiltrating myeloid cells, as well as the emergence of a small, but discreet population of activated antigen presenting cells. Currently ongoing studies are aimed at delineating the exact molecular and cellular changes in the tumor microenvironment in response to the EGFRvIII-CAR T-cell treatment, in combination with clinically relevant therapies targeting the largely immunosuppressive tumor myeloid compartment


Blood ◽  
2008 ◽  
Vol 111 (3) ◽  
pp. 1504-1511 ◽  
Author(s):  
Suzanne N. Franki ◽  
Kristopher K. Steward ◽  
David J. Betting ◽  
Kamran Kafi ◽  
Reiko E. Yamada ◽  
...  

Abstract The in vitro priming of tumor-specific T cells by dendritic cells (DCs) phagocytosing killed tumor cells can be augmented in the presence of antitumor monoclonal antibody (mAb). We investigated whether DCs phagocytosing killed lymphoma cells coated with tumor-specific antibody could elicit antitumor immunity in vivo. Irradiated murine 38C13 lymphoma cells were cocultured with bone marrow–derived DCs in the presence or absence of tumor-specific mAb. Mice vaccinated with DCs cocultured with mAb-coated tumor cells were protected from tumor challenge (60% long-term survival), whereas DCs loaded with tumor cells alone were much less effective. The opsonized whole tumor cell–DC vaccine elicited significantly better tumor protection than a traditional lymphoma idiotype (Id) protein vaccine, and in combination with chemotherapy could eradicate preexisting tumor. Moreover, the DC vaccine protected animals from both wild-type and Id-negative variant tumor cells, indicating that Id is not a major target of the induced tumor immunity. Protection was critically dependent upon CD8+ T cells, with lesser contribution by CD4+ T cells. Importantly, opsonized whole tumor cell–DC vaccination did not result in tissue-specific autoimmunity. Since opsonized whole tumor cell–DC and Id vaccines appear to target distinct tumor antigens, optimal antilymphoma immunity might be achieved by combining these approaches.


2021 ◽  
Vol 12 ◽  
Author(s):  
Xuemei Yang ◽  
Jialing Sun ◽  
Bin Wen ◽  
Yu Wang ◽  
Mingjia Zhang ◽  
...  

Tumor-infiltrating CD8+T lymphocytes are mostly associated with a favorable prognosis in numerous cancers, including hepatocellular carcinoma (HCC). Biejiajian Pill (BJJP) is a common type of traditional Chinese medicine that is widely used in the treatment of HCC in China. Previous studies showed that BJJP suppressed the growth of HCC cells both in vivo and in vitro, by exerting direct cytotoxic effects on tumor cells. The present study demonstrated that in addition to direct cytotoxicity, BJJP inhibits the growth of tumor cells by promoting the infiltration of CD8+T cells into the tumor in H22-bearing mice. Mechanistically, chemokine ligand 5 (CCL5) was identified as one of the most highly expressed chemokines by tumor cells in vivo after treatment with BJJP. Additionally, CCL5 was knocked down in H22 cells and the results showed that knockdown of the gene significantly impaired the infiltration of CD8+T cells in vivo. Furthermore, the effects of BJJP on human HCC cell lines were assessed in vitro. Similarly, cells treated with BJJP had higher expression of CCL5 mRNA, which was consistent with increased levels of CCL5 protein in human tumor cells. These findings provide new insights into the anticancer effects of BJJP, which regulated the expression of CCL5 and the infiltration of CD8+T cells. The results, therefore, suggest that BJJP has great potential application in clinical practice.


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. 3063-3063
Author(s):  
R. Conforti ◽  
Y. Ma ◽  
Y. Morel ◽  
C. Paturel ◽  
M. Terme ◽  
...  

3063 Background: Several tumor cells express toll-like receptors (TLRs) which constitute putative therapeutic targets. Methods: To dissect the clinical outcome of the host versus cell autonomous effects of the TLR3 agonist poly(A:U), we took advantage of two murine TLR3 expressing tumor models (melanoma and glioblastoma) that produced large amounts of CCL5 (a CCR5 ligand) and CXCL10 (a CXCR3 ligand) in response to the poly(A:U) and type I IFN in vitro and in vivo. We tested in vivo a triple combination based on vaccine against tumor antigens (V), chemotherapy (C), and TLR3 agonist (poly[A:U]) (T). Results: Single agents (V, C, or T) and combinations of two agents (VC, VT, or CT) failed to improve tumor progression. However, the sequential tritherapy (VCT) significantly retarded tumor growth and prolonged the survival of tumor-bearing C57BL/6 mice. The antitumor effects of VCT (immunochemotherapy) failed to be observed in nu/nu and TRIF-/- mice, indicating the contribution of T cells and TRIF-dependent signals to the therapeutic effect. Knockout of CCR5 improved the efficacy of immunochemotherapy. The VCT combination induced the recruitment of IFNγ+/CD8+/CXCR3+ T lymphocytes supporting the notion that this subset of T cells contributes to the antitumor efficacy of immunochemotherapy. Knockout of the CXCR3 gene annihilated the beneficial effects of VCT. The source of the deleterious CCL5 was the TLR3-activated tumor cells in that stable inhibition of the chemokine production by targeted shRNA CCL5 ameliorated the efficacy of VCT tritherapy. Altogether, 11% of wild-type mice were completely protected from melanoma by VCT sequential regimen. Conclusions: This study supports the notion that poly(A:U) can act on tumor epithelia to promote the release of beneficial CXCL10 for the recruitment of intratumoral CTLs but also the release of deleterious CCL5 acting on host immunosuppressors. Uncoupling chemokine release and/or prior vaccination may enable the CXCR3L-dependent CTL to overrule the CCR5-dependent suppression and may be integrated in future trials using TLR3 agonists. [Table: see text]


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. e14510-e14510
Author(s):  
Eric Zeng ◽  
Tailan Zhan ◽  
Yang Wu ◽  
Meili Chen ◽  
Junzheng Wang ◽  
...  

e14510 Background: CD4 is highly and uniformly expressed T-cell lymphomas (TCL) including PTCL and CTCL, suggesting its potential as a surface target for CAR-T therapy. However, there is a significant risk of potential antigen masking by CAR introduced into tumor cells, which thereby leads to escape of tumor cells from recognition by CAR-T cells. For example, Ruella M et al ( Nat Med, 2018) reported that contaminating malignant B cells transduced with anti-CD19 CAR (called CAR-B cells) in the manufacturing process led to the CD19 antigen masking by CAR molecules. Hence, CAR-B tumor cells could not be recognized by CAR-T cells, which resulted in relapse of CAR expressing B lymphoma cells. A similar antigen masking effect might occur for the anti-CD4 CAR-T product. Methods: To identify single-chain variable fragment (scFv) without antigen-masking effect, large panels of fully human monoclonal antibodies were converted to format of CAR modality with the scFv-4-1BB-CD3z structure and introduced into CD4+ and CD8+ T cells. After lentiviral transduction, residual CD4+ T cells were quantified and the CARs with complete elimination of CD4+ T cells were selected for further validation. The selected CAR constructs were then introduced into CD4+ TCL cells HH (CAR-HH cells) to mimic the potential risk of introducing CAR into contaminating malignant T cells in the manufacturing process. Thereafter, CAR-HH cells were subject to in vitro killing assay by LB1901. Last, identified CAR constructs were further tested for in vitro and in vivo anti-tumor efficacy and off-target binding and killing. Results: Introduction of LB1901 CAR into CD4+ and CD8+ T cells led to complete elimination of CD4+ T cells, suggesting no masking effect of CAR on CD4 antigen. Furthermore, the introduction of CAR into CD4+ HH cells did not protect HH cells from being recognized and eliminated by LB1901, further confirming that the CAR modality of LB1901 does not mask CD4 antigen. An in vivo anti-tumor efficacy study showed that LB1901 exhibited dose-dependent anti-tumor activity without significant adverse effect. As low as 0.3 million CAR+ cells completely suppressed tumor growth, suggesting the potent anti-tumor activity by LB1901. Immunohistochemical analysis of normal tissues with LB1901 scFV binder showed no off-target binding. Furthermore, no killing toward CD4- cell lines and primary cells derived from vital organs or antigen-independent cytokine release was observed in vitro. Conclusions: Altogether, the in vitro and in vivo studies showed that LB1901 did not “mask” the CD4 antigen but exhibited potent anti-tumor activity without off-target effects. A phase 1 study of LB1901 CAR-T in patients with relapsed or refractory PTCL or CTCL is ongoing in the US to assess the safety and tolerability of LB1901 CAR-T (NCT04712864).


2020 ◽  
Vol 22 (Supplement_3) ◽  
pp. iii360-iii360
Author(s):  
Dalia Haydar ◽  
Zhongzhen Yi ◽  
Chris DeRenzo ◽  
Stephen Gottschalk ◽  
Giedre Krenciute

Abstract BACKGROUND We and others have identified B7-H3 (CD276) as a promising target for CAR-based immunotherapies for pediatric brain tumors. So far, B7-H3-CAR T cells have only been studied in xenograft models for brain tumors, which do not recapitulate the immunosuppressive tumor microenvironment (TME). To overcome this obstacle, we decided to adapt the immune-competent GL261 murine glioma model which mimics human disease and host immune barriers. METHODS To evaluate the safety and efficacy of antigen-specific CAR T cells, murine B7-H3-CAR T cells were generated using retroviral particles encoding 2nd generation B7-H3-specific CD28.z CAR. Expansion, persistence, and anti-tumor activity were evaluated in vitro and in vivo. Components of the brain TME were then evaluated using flow cytometry and immunostaining. RESULTS B7-H3-CAR T cells only killed B7-H3+ tumor cells, secreted significant levels of IFNγ and IL-2 in an antigen-dependent manner and expanded an average of 33-fold in repeat stimulation assay with B7-H3+ tumor cells in contrast to control CAR T cells. In vivo, intratumoral injection of B7-H3-CAR T cells into orthotopic GL261 glioma induced complete regression in 60% of treated mice. Preliminary studies show numerous infiltration of suppressive tumor-associated macrophages within the tumor and its periphery. CONCLUSIONS In summary, we successfully generated murine B7-H3-CAR T cells and have demonstrated that these cells have potent anti-tumor activity in the immune-competent GL261 glioma model. However, it is likely that the tumor-associated macrophages are mediating immunosuppressive effects on B7-H3-CAR T cells. Therefore, studies focusing on TME/CAR T cell interactions are in progress.


Sign in / Sign up

Export Citation Format

Share Document