Usefulness of IL-21, IL-7, and IL-15 conditioned media for expansion of antigen-specific CD8+ T cells from healthy donor-PBMCs suitable for immunotherapy

2021 ◽  
Vol 360 ◽  
pp. 104257
Author(s):  
Julián A. Chamucero-Millares ◽  
David A. Bernal-Estévez ◽  
Carlos A. Parra-López
2008 ◽  
Vol 20 (8) ◽  
pp. 1087-1096 ◽  
Author(s):  
V. Voelter ◽  
N. Rufer ◽  
S. Reynard ◽  
G. Greub ◽  
R. Brookes ◽  
...  

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. e14511-e14511
Author(s):  
Amani Makkouk ◽  
Xue (Cher) Yang ◽  
Taylor Barca ◽  
Anthony Lucas ◽  
Mustafa Turkoz ◽  
...  

e14511 Background: Autologous αβ chimeric antigen receptor (CAR) T cell therapy has shown promising clinical results in hematologic malignancies but limited success in solid tumors. Allogeneic αβ T cell therapy may overcome several challenges faced by autologous therapy but carries the risk of graft-versus-host disease (GvHD) and does not readily recognize multiple tumor-associated antigens. Gamma delta (γδ) T cells are highly cytolytic effectors that can recognize and kill tumor cells in an MHC-unrestricted manner without causing GvHD. The Vδ1 subset is preferentially localized in peripheral tissue and is critical for tumor immunosurveillance. Engineering Vδ1 T cells with CARs can further enhance antitumor activity and represents an attractive and safe approach to treating solid tumors. However, their clinical use has been hindered by the limited number of circulating Vδ1 T cells. Here, we describe the development of the first allogeneic Vδ1 T cells that have been expanded from healthy donor PBMCs and genetically modified to secrete IL-15 (sIL15) and express a CAR targeting glypican-3 (GPC3), a rational target for hepatocellular carcinoma (HCC). Methods: Vδ1 T cells in healthy donor PBMCs were activated by a Vδ1-specific monoclonal antibody and transduced with 41BBζ or 41BBζ-sIL15 GPC3-CARs prior to cell expansion, αβ T cell depletion and cryopreservation. In vitro characterization included: 1) co-culture assays with GPC3-expressing HCC targets HepG2 and PLC/PRF/5, 2) phenotypic analysis by flow cytometry, and 3) cytokine production by multiplexed immunoassay. For in vivo assessment of tumor control, immunodeficient NSG mice were subcutaneously injected with HepG2 cells and treated with a single dose of 41BBζ or 41BBζ-sIL15 GPC3-CAR Vδ1 T cells. Additionally, tissues were harvested 7 days post transfer and analyzed by flow cytometry for Vδ1 T cell tissue homing and proliferation, or at end of study and analyzed for GvHD by immunohistochemistry. Results: Vδ1 T cells expanded over 10,000-fold and routinely reached >80% purity. Expanded Vδ1 T cells showed a primarily naïve-like phenotype (CD45RA+CD27+) with minimal exhaustion receptor expression and displayed robust proliferation, cytokine production, and cytotoxic activity against HCC cell lines expressing low and high GPC3 levels in vitro. In a HepG2 mouse model, GPC3-CAR Vδ1 T cells primarily accumulated and proliferated in the tumor, and a single dose was able to efficiently control tumor burden without causing GvHD. Importantly, 41BBζ-sIL15 GPC3-CAR Vδ1 cells displayed enhanced tumor-specific proliferation that resulted in better tumor control without any toxicity. Conclusions: Our results show that expanded Vδ1 T cells engineered with GPC3-CAR and sIL-15 represent a promising platform for safe and effective off-the-shelf treatment of HCC.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3605-3605 ◽  
Author(s):  
Haitham Abdelhakim ◽  
Luis M. Cortez ◽  
Meizhang Li ◽  
Mitchell Braun ◽  
Barry S. Skikne ◽  
...  

Background: Acute Myeloid Leukemia (AML) is an aggressive hematologic malignancy with known immune dyregulation. In addition to their capacity to rapidly divide, AML cells directly inhibit the activation and proliferation of immune cells in culture. Immunosuppressive features observed in the bone marrow of AML patients include upregulation of Tregs and production of immunosuppressive cytokines (e.g., TGFβ). Irradiating AML cells diminishes their immunosuppressive capacity while maintaining antigen presentation, leading to increased activation of T cells in co-culture. We subsequently identified the immune checkpoint LAG3 as an important mediator of AML-induced immunosuppression and LAG3 modulation as potential treatment strategy. Methods: Normal PBMC were isolated from healthy donors. PBMC were co-cultured with non-irradiated and irradiated (40 Gy) human AML cell lines (Kasumi1 (K1), THP1) separately at a 1:2 ratio. On day 3 of co-culture, immunophenotypic characterization of T cells was performed on a flow cytometer using the following surface markers: CD3, CD4, CD8, CD25, CD137, CD154, PD-1, TIM3, TIGIT, and LAG3 and intracellular IFNg and FOXP3. Supernatant from co-culture media were analyzed for cytokine (IL-2, IL-6, IL-10 & TGFβ) secretion by ELISA. CFSE-labeled AML cells were incubated with healthy donor PBMCs in the presence or absence of LAG3, then viability was measured by 7-ADD on flow cytometry. PBMCs were also isolated from AML patients' peripheral blood and mononuclear cells were isolated from their respective bone marrow samples. Primary AML cultures were established in RPMI complete media with 20% FBS. CFSE-7-ADD killing assay was conducted after incubation of AML cells with autologous PBMCs. Results: Healthy donor PBMC co-cultured with irradiated K1 AML cells showed higher intracellular IFNg expression (11.8% ± 3.1 v. 7% ± 3.3; n=7, P=0.012) and higher CD137 expression (9.3% ± 1.21 v. 5.7% ± 3.4; n=7, P<0.001) on CD8+ T cells, and higher CD154 expression on CD4+ cells (44.7% ± 20.3 v. 26.3% ± 14.2; n=5, P=0.002) when compared to the non-irradiated K1-PBMC co-cultures. There were fewer Tregs (CD4+ CD25+ FOXP3+) in the PBMC co-cultured with irradiated K1 cells (1.96% ± 0.37 v. 3.39% ± 0.58; n=4, P=0.03) compared to the non-irradiated K1-PBMC co-cultures. LAG3 expression on CD8+ T cells co-cultured with irradiated K1 was decreased (11.8% ± 2.4 v. 17.5% ± 2.5; n=4, P=0.002) compared to the PBMC co-cultured with non-irradiated K1 cells. No other changes in checkpoint expression on CD8+ T cells were observed. No changes were observed in MHCI or PDL1 expression on non-irradiated K1 AML cells before or after co-culture with PBMC. We observed similar findings with healthy donor PBMC co-cultured with a different AML cell line, THP1; CD137 expression was higher on CD8+ T cells (17.6% v. 6.5%; P=0.02, n=3). ELISA of the supernatant of culture media showed higher mean OD for secreted TGFβ in the non-irradiated AML co-cultures compared to the irradiated AML co-cultures at 6 hours (2.5 v. 2.0, P=0.03, n=3) and 72 hours (7.9 v. 5.3, P=0.04, n=3). Adding anti-LAG3 antibody (3DS223H; 100 µg/ml) to PBMC co-cultured with non-irradiated AML cells resulted in higher IFNg (16.3% v. 6.6%, P=0.01, n=4) and CD137 expression (6.5% v. 4.1%, p=.007, n=4) on CD8+ cells and fewer Tregs (1.7% v. 3.8%, P=.04, n=4) compared to no antibody added. Healthy donor PBMC (n=3) were incubated with CFSE labeled AML cells (K1 and THP1) separately at an effector:target ratio of 5:1. The addition of anti-LAG3 antibody lead to increased killing of both K1 and THP1 AML cells at 4 and 24 hours (Figure 1A). To eliminate the HLA mismatch effect, we incubated PBMC from AML patients with autologous AML cells in the presence or absence of anti-LAG3 (Figure 1B). MHC-I blocking (W6/32, 30 µg/ml) lead to inhibition of cell mediated killing in the presence of anti-LAG3 (Figure 1B). Conclusion: In this in vitro model, AML cells showed immunosuppressive features with decreased activation of CD8+ T cells, upregulation of Tregs, increased secretion of TGFβ and higher expression of LAG3 on CD8+ T cells. Antibody blocking of LAG3 mitigated this effect, resulting in increased activation of T cells, fewer T regs and improved MHC-I-mediated killing against AML cells. These results demonstrate that the immunosuppressive effects of AML cells can be modulated through inhibition of LAG-3, suggesting a potential strategy for future combination therapy in AML. Disclosures Lin: Jazz Pharmaceuticals: Honoraria; Pfizer: Membership on an entity's Board of Directors or advisory committees.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A106-A106
Author(s):  
David Langan ◽  
Jourdain Lemaster ◽  
Lauren Suarez ◽  
Pratima Kunwar ◽  
Sojung Kim ◽  
...  

BackgroundNexImmune is developing highly differentiated immunotherapies to target, activate and expand tumor antigen-specific T cells using the proprietary Artificial Immune Modulation (AIM™) nanotechnology platform. The AIM nanoparticle (AIM-np) technology functions as synthetic dendritic cells capable of directing a specific T cell-mediated immune response. By mimicking natural T cell biology, NexImmune’s cellular therapy product candidates (AIM ACT) are designed to combine the attributes of cellular precision, potency, and persistence with reduced potential for undesired toxicities. Human papilloma virus (HPV) is responsible for >45,000 cancers yearly in the United States, according to the CDC. From 2013–2017 an estimate 79% of cervical, vulva, penis, vaginal, anus, and oropharyngeal cancers were attributed to HPV and of these about 80% were associated with high-risk HPV types 16 and 18. Although multivalent vaccines against high-risk HPV infections exist, significant clinical challenges remain. A limited vaccination rate means many remain vulnerable and vaccination does not treat pre-existing HPV infections or malignancies.MethodsTherefore, NexImmune is employing its AIM-np technology to generate an adoptive cell therapy (ACT) using its proprietary enrichment and expansion (E+E) ex vivo process to expand clinically relevant numbers of CD8+ T cells that recognize the HPV16 and HPV18 oncogenic antigens (i.e., E6 and E7) expressed by malignant cells of head and neck, cervical, and anal cancers. Using the Immune Epitope Database and Analysis Resource (IEDB), 44 HLA-A2 restricted peptides were identified as potential immunogenic targets for preclinical screening. Using PBMCs from healthy donor-derived apheresis material, different combinations of these peptides were used in the E+E process to expand HPV-cancer specific CD8+ T cells.ResultsAfter multiple E+E experiments were concluded, 5 peptides were identified that consistently elicited the strongest T cell responses. Furthermore, these CD8+ T cells were predominantly from the central memory (CD62L+CD45RA-) and effector memory (CD62L-CD45RA-) phenotype (sum total 82.18 ± 8.29 [Mean ± SEM]) suggesting their in vivo functionality and persistence will combine anti-tumor activity with long-term immunologic memory.ConclusionsA similar E+E screening is being conducted with PBMCs isolated from HPV+ cancer patients. A comparison of the CD8+ T cell responses from healthy donor and cancer patient cells will provide critical preclinical data to support a planned FIH trial for HPV-associated malignancies. The current study demonstrates the ability for high-throughput peptide screening to identify clinically relevant peptide cocktails capable of expanding multi-antigen tumor-specific CD8+ T cell populations within 2 weeks.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 226-226
Author(s):  
Athalia Rachel Pyzer ◽  
Dina Stroopinsky ◽  
Jacalyn Rosenblatt ◽  
Kristen Anna Palmer ◽  
Maxwell Douglas Coll ◽  
...  

Abstract Introduction: Myeloid-derived suppressor cells (MDSCs) are a critical component of the immunosuppressive milieu of the tumor microenvironment and play an important role in promoting immune tolerance and disease growth. They are comprised of granulocytic and monocytic compartments defined by a unique immunophenotypic signature. Importantly, the mechanism by which tumor cells evoke the expansion of MDSCs has not been well elucidated. In the present study, we examined the interaction of MDSCs with AML cells, a setting in which the presence and function of MDSC has not been well described. Methods and Results: Peripheral blood mononuclear cells (PBMCs) were isolated from patients with active AML and granulocytic (CD33+/CD11b+/HLADR-/CD15+) and monocytic (CD33+/CD11b+/HLADR-/CD15-) MDSCs were quantified by multichannel flow cytometry. AML patients had a significantly higher mean granulocytic MDSC population of 17.2% (n=3) compared to healthy controls 1.9%, (n=10) p=0.0083 and a mean monocytic MDSC population of 6.5% (n=3), which was similar to healthy controls (monocytic MDSCs 4.1%, n=10). MDSCs isolated from an AML patient exhibited immunosuppressive effects as measured by the suppression of dendritic cell mediated stimulation of T cells. The addition of AML derived MDSCs resulted in a 40% reduction in CD4+T cell production of IFNϒ and an 11 fold increase IL-10 secretion by CD4 and CD8 T cells following coculture with allogenic DC stimulation. The ability of AML blasts to directly induce the expansion of MDSC was assessed in vitro. Healthy donor PBMCs were co-cultured for 6 days with or without the AML cell lines MOLM-14 and THP-1 at a ratio of 100:1. MDSCs were quantified after 6 days. Coculture with MOLM-14 and THP-1 induced a 2.35 and 8.2 fold increase in MDSCs respectively (n=4). MUC1 is a critical oncogene expressed on leukemic blasts and leukemia initiating cells and plays an important role in the tumor microenvironment promoting tumor growth and immune escape. In the present study, we demonstrated that silencing of MUC1 via shRNA significantly diminishes AML recruitment and expansion of MDSCs in vitro. MOLM-14 cells underwent lentiviral transfection to silence MUC1-C expression which was confirmed by Western Blot. MOLM-14 wild type, MUC1 silenced, and control vector treated cells were co-cultured with healthy PBMCs for 6 days in a ratio of 100:1. Of note, MUC1-C silenced MOLM-14 and THP-1 cells exhibited decreased capacity to expand MDSCs upon co-culture with healthy donor PBMCs, as compared to the control vector (2.4 fold higher expansion of MDSCs with control vector MOLM-14 compared to MUC1-C silenced MOLM-14, n=4, 1.92 fold higher expansion of MDSCs with control vector THP-1 compared to MUC-1C silenced THP-1, n=4). In an in vivo model, NSG mice were irradiated and inoculated with THP-1 control and THP-1 MUC1 silenced cells. Following establishment of disease, mice were sacrificed and spleens were FACS analysed for MDSC quantification. Mice inoculated with THP-1 MUC1 silenced cells had mean MDSCs of 7.5%, compared to 16.25% in mice innoculated with THP-1 Wildtype cells (n=4). In conclusion, the data demonstrates that MDSCs are increased in the circulation of patients with AML, and that leukemic blasts directly induce the expansion of MDSCs. MUC1 expression on AML blasts contributes to the immunosuppressive milieu, and notably, silencing of MUC1 in AML cells blunts their capacity to induce the expansion of MDSCs. Incorporating strategies to inhibit the expansion of MDSC in AML, and reverse their immunosuppressive phenotype has the potential to improve response to therapy in AML. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Manuel Reithofer ◽  
Sandra Rosskopf ◽  
Judith Leitner ◽  
Claire Battin ◽  
Barbara Bohle ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document