scholarly journals 164 Potent tumor organoid infiltration and killing by PBMC-derived effector cells

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A174-A174
Author(s):  
Frank Borriello ◽  
Joshua Keegan ◽  
James Lederer

BackgroundAlloplex Biotherapeutics has developed a novel autologous cellular therapy for cancer that uses ENgineered Leukocyte ImmunoSTimulatory cell lines called ENLIST cells to activate and expand a heterogeneous population of tumor killing effector cells from human peripheral blood mononuclear cells (PBMCs). The 2-week manufacturing process from PBMCs consistently results 300-fold expansion of NK cells, CD8+ T cells, gamma/delta T cells, NKT cells and some CD4+ T cells, collectively called SUPLEXA therapeutic cells. SUPLEXA cells will be delivered back to cancer patients via intravenous administrations on a weekly schedule as an autologous adoptive cellular immunotherapy for cancer. In this study, we tested SUPLEXA cells developed from normal healthy volunteer PBMCs for their ability to infiltrate and kill patient-derived tumor organoids (PDO) as a pre-clinical assessment for potency against 2 different types of tumor organoids.MethodsTumor organoids derived from colorectal cancer (CRC) or non-small cell lung carcinoma (NSCLC) patients were labeled with cell-trace red dye and plated at equal density in a 96-well plate. After 3 days culture, SUPLEXA cells were thawed (82.8% viable), labeled with cell-trace violet dye, and added to PDO at 1:2 serial diluted numbers ranging from 2 million to 7,800 cells per well. Fluorescent images were captured at 24 hours after adding SUPLEXA cells to PDO models to measure PDO size, tumor infiltration, and PDO killing.ResultsAdding SUPLEXA cells to PDO from CRC and NSCLC resulted in significant infiltration and killing of organoids by 24 hours as shown by the fluorescent images and the organoid size plot for the CRC PDO model (figure 1). Significant reduction in PDO size was observed by adding 31,240 SUPLEXA cells. Similar results were observed with the NSCLC PDO model with significant reduction in PDO size by adding 15,600 SUPLEXA cells. Obvious organoid infiltration was observed in both PDO models and organoid fluorescence was significantly reduced by addition of SUPLEXA cells in both PDO models to suggest that SUPLEXA cells were able to reduce tumor burden (figure 2).Abstract 164 Figure 1CRC organoid infiltration and killing by SUPLEXA. A representative fluorescent image of CRC organoid killing with addition of increasing SUPLEXA cell numbers and a plot showing statistical analysis of 6 replicate wells for changes in CRC organoid size in relation to SUPLEXA cell number additionsAbstract 164 Figure 2Dose-dependent killing in CRC and NSCLC PDO models. CRC and NSCLC organoids were detected by total red fluorescence at 24 hours after adding the indicated numbers of SUPLEXA cells. Loss of red fluorescence after adding SUPLEXA is a measure of overall tumor cell killing/burden in organoids. Data is plotted as mean ± SEM for n=6 replicates per group.ConclusionsSUPLEXA cells infiltrated and killed tumor cells in patient-derived organoids within 24 hours of culture at low cell concentrations indicating potent tumor killing activity. The observed activity in both colorectal and lung cancer organoid models support broad anti-tumor killing activity by SUPLEXA. These results provide further evidence that PBMCs from cancer patients can be activated and expanded by our approach as a novel autologous cellular immunotherapy for cancer.

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 7517-7517
Author(s):  
Joshua W. Keegan ◽  
Frank Borriello ◽  
Stacey M. Fernandes ◽  
Jennifer R. Brown ◽  
James A. Lederer

7517 Background: Alloplex Biotherapeutics has developed a cellular therapeutic that uses ENgineered Leukocyte ImmunoSTimulatory cell lines called ENLIST cells to activate and expand populations of tumor killing effector cells from human peripheral blood mononuclear cells (PBMCs). This process leads to a 300-fold expansion of NK cells, CD8+ T cells, NKT cells, and TCRγδ T cells that are called SUPLEXA cells, which will be cryopreserved and transferred back into patients as an autologous immune cell therapy for cancer. In this study, PBMCs from CLL patients were used to generate SUPLEXA cells as a first approach to comparatively profile SUPLEXA cells from cancer patients and normal healthy volunteers (NHVs). Methods: ENLIST cell lines were engineered by expressing curated immunomodulatory proteins in the SK-MEL-2 melanoma cell line. Two million (M) PBMCs from 10 CLL patients or 2 NHVs were incubated with 0.4 M freeze/thaw killed ENLIST cells for 5 days in XVIVO-15 medium with 2% heat-inactivated human AB serum (XAB2) and then split 1:15 in XAB2 containing IL-7 and IL-15 to expand. After 9 days, SUPLEXA cells were harvested and cryopreserved. Results: Original PBMCs and matched SUPLEXA cells from each donor were thawed and characterized by mass cytometry (CyTOF) using a 47-marker antibody panel. CyTOF staining results of PBMCs from CLL patients demonstrated approximately 95% leukemia cells and few T cells, NK cells, B cells, and monocytes. CyTOF staining of SUPLEXA cells from all 10 CLL patients showed expansion of NK cells (17%), CD8 T cells (11%), and CD4 T cells (7.5%) that were similar in phenotype to SUPLEXA cells from NHVs showing high expression of granzymes and perforin that are indicative of potent tumor cell killing activity. Cancer cells in the original CLL PBMC samples were reduced to 0.78%. However, a population of non-T/non-B cells (60% ± 9.5%) was detected in SUPLEXA cells from all CLL patients that require further characterization. Next, SUPLEXA cells from CLL and NHV patients were comparatively tested for tumor cell killing activity at 2:1, 1:1, and 1:2 effector to target cell (MEL-14 melanoma cells expressing RFP) ratios. Percent killing of tumor cells by SUPLEXA cells prepared from CLL patients (77.8% ± 2.6% at 2:1) and NHVs (81.5% ± 0.3% at 2:1) were nearly identical at all effector to target ratios. Conclusions: We demonstrate for the first time that PBMCs from CLL patients can be converted into SUPLEXA cells despite low numbers of normal immune cells at baseline and the known immunologic impairment present in CLL patients. Importantly, SUPLEXA cells derived from CLL patients acquire potent tumor killing activity that is indistinguishable from SUPLEXA cells prepared from NHVs. Taken together, these findings support the feasibility of converting PBMCs from CLL patients with low percentages of NK and T cells into an autologous cellular therapy for cancer.


Blood ◽  
2009 ◽  
Vol 114 (6) ◽  
pp. 1141-1149 ◽  
Author(s):  
Ilona Kryczek ◽  
Mousumi Banerjee ◽  
Pui Cheng ◽  
Linhua Vatan ◽  
Wojciech Szeliga ◽  
...  

Abstract Th17 cells play an active role in autoimmune diseases. However, the nature of Th17 cells is poorly understood in cancer patients. We studied Th17 cells, the associated mechanisms, and clinical significance in 201 ovarian cancer patients. Tumor-infiltrating Th17 cells exhibit a polyfunctional effector T-cell phenotype, are positively associated with effector cells, and are negatively associated with tumor-infiltrating regulatory T cells. Tumor-associated macrophages promote Th17 cells through interleukin-1β (IL-1β), whereas tumor-infiltrating regulatory T cells inhibit Th17 cells through an adenosinergic pathway. Furthermore, through synergistic action between IL-17 and interferon-γ, Th17 cells stimulate CXCL9 and CXCL10 production to recruit effector T cells to the tumor microenvironment. The levels of CXCL9 and CXCL10 are associated with tumor-infiltrating effector T cells. The levels of tumor-infiltrating Th17 cells and the levels of ascites IL-17 are reduced in more advanced diseases and positively predict patient outcome. Altogether, Th17 cells may contribute to protective human tumor immunity through inducing Th1-type chemokines and recruiting effector cells to the tumor microenvironment. Inhibition of Th17 cells represents a novel immune evasion mechanism. This study thus provides scientific and clinical rationale for developing novel immune-boosting strategies based on promoting the Th17 cell population in cancer patients.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4973-4973
Author(s):  
Hong-Hanh Nguyen ◽  
Dong-hwan Kim ◽  
Hyun-Ju Lee ◽  
Sung-Hoon Jung ◽  
Hyeoung-Joon Kim ◽  
...  

Abstract (Background) Adoptive T cell therapy using tumor-infiltrating lymphocytes (TILs) is a promising treatment for cancer patients unresponsive to conventional therapies. However, clonal expansion of T cells easily faced with T cell exhaustion or terminal differentiation which related to suppression of the tumor-killing function in tumor microenvironment. Naïve CD8+T cells have an astounding capacity to react to antigens by massive expansion and differentiation into cytotoxic effector cells. However, it is the main key factor that how can increase the limited number of tumor-specific naïve CD8 T cells due to the negative selection during T cell thymic development. (Methods) In human, naïve, memory T cells and TILs were activated with CD3/CD28 dynabeads and culture on CD2-coated plate to generate various effector T cell populations (Teff N, Teff M and activated TILs). Those different effectors were analyzed the expression of surface exhaustion phenotypes, intracellular transcription factors (T-bet/Eomes) and granzyme/perforin secretions using FACS and western blot assay. Telomere length of three kinds of effector cells was analyzed. High-dose IL-2 (1ug/mL) activated naïve CD8 T cells were stimulated with tumor antigen-loaded dendritic cells (DCs) and then, ELISPOT/cytokine ELISA assay was performed to evaluate tumor-specific (TA) CTL function. In murine model, we also checked the functional difference of each effector cells as like the same methods. In addition, tumor challenging test using EG7-EL4 cell line (OVAp expression) was performed in C57BL/6 mice which adoptively transferred with OT-I thy1.1 Teff N, Teff M and activated TILs. (Results) In vitro expansion of all human naïve, memory and TIL CD8+ T cells was induced successfully. After 3-5 days of expansion, effectors from different progenitors were assessed for the several activation markers CD44, OX40 and CD27 and were considered as the CD62LlowCD44highOX40highCD27high populations. Population frequency of Teff N was significantly higher than Teff M (p < 0.05) or activated TILs (p < 0.005). Telomere length, which correlates with replicative capacity, was greatest in TeffN, shorter in TeffM and shortest in aTILs. When compared the T cell exhaustion phenotypes in three different effector cells, TeffN showed the very low expression of inhibitory markers including PD-1, CTLA-4, and KLRG-1.aTILs expressed most high exhaustion phenotypes with shorter telomere length. Moreover, the secretion of cytotoxic granules such as granzyme B and perforin gradually increased in all effector cells but, among the fully effector cells status, TeffN possess the highest expression level compared to TeffM and aTILs wheras similar level of IFN-r. To further confirm expression profile of T-box transcription factors, the expression of both T-bet and Eomes in TeffN increased at relatively early time point (D+3) and sustained high expression levels during effector status (D+5 and D+8). High expression of T-bet in TeffN promoted the full effector generation and Eomes expression linked to formation of long-term tumor-specific memory population. In CTL function, high-dose IL2-activated naïve T cells were successfully increased and generated the tumor-specific CTLs co-cultured with TA DCs, which inducing the outstanding CTL function compared to those from memory CD8 T cells or aTILs. (Discussion) High-dose IL-2 could increase the tumor-specific naïve CD8 T cells without resulting in clonal exhaustion and could generate the potent tumor-specific CTLs with overcoming the tumor-derived immune suppression compared with CD8+ TILs or secondary effectors from memory T cells. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2329-2329
Author(s):  
Deok-Hwan Yang ◽  
Jae-Sook Ahn ◽  
Seo-Yeon Ahn ◽  
Sung-Hoon Jung ◽  
Je-Jung Lee ◽  
...  

Introduction Terminally differentiated effector memory (TEMRA) cells have been recent revealed as sub-population of CD8 T cells, and they have been expected to act like memory-like properties in peripheral blood. However, the precise characteristics or function of these cells have not been defined. Particularly, these cells have been found even in cancer tissues including tumor-infiltrating effector CD8+ T cells (TILEM). The study investigated the phenotypic or genomic difference of CD8+ TEMRA cells which extracted from not only peripheral blood of healthy and cancer patients but also tumor tissues of cancer patients. Material and methods Peripheral blood or tumor tissues from normal donors and patients with lung cancer or Hodgkin lymphoma were collected. Three different sources of CD8+ TEMRA cells, which from healthy peripheral blood (hTEMRA), patient peripheral blood (pTEMRA) and TIL (TIL TEMRA), were obtained and then analyzed for phenotypic and genomic differences. CD8+ T cells were isolated using magnetic-activated cell sorting (MACS), and sub-type cells were isolated using FACS (fluorescence-activated cell sorting) based on phenotypic factors (TEMRA; CD8+ CCR7- CD45RA+). Tumor tissues from patients with lung cancer were digested with enzyme and preceded to the same manner as peripheral blood. Next-generation sequencing (NGS) was used to characterize the TEMRA by using Hiseq2000 (Illumina) on cDNA synthesized from RNA extracted from sorted cells (TEM; CD8+ CCR7- CD45RA-, TEMRA; CD8+ CCR7- CD45RA+). All experiments were performed after obtaining the informed consent approved by the institutional ethical committee (IRB). Results CD8+ TEMRA exist with similar proportion in cancer tissues as well as peripheral blood in lung cancer patients. TIL TEMRA was distinguished from regulatory T cells without expression of CD25 and foxp3. All healthy or patient TEMRA cells including TIL TEMRA showed similar expression level of exhaustion phenotypes (CTLA4, PD-1 and Tim3) (p>0.05). However, TIL TEMRA cells exhibited significantly less amounts of T cell exhaustion markers (PD-1 and Tim3) than those of TILEM cells. Genomic background of blood-derived TEMRA differs from tumor tissue-derived TEMRA based on NGS assessment. Moreover, unlike general CD8+ TILEM, which consist mostly of exhausted effector cells due to tumor-derived immunosuppression, CD8+ TIL TEMRA showed the different genomic background. The transcriptional factors (T-bet and Eomes) or genomic patterns of CD8+ TIL TEMRA represented less exhausted, proliferative and cytotoxic than CD8+ TILEM. However, CD8+ TIL TEMRA might be induced into secondary effector cells after TCR re-stimulation. Conclusion Genomic and functional characteristics of CD8+ TIL TEMRA distinct from not only TIL TEM but also peripheral TEMRA.in cancer patients. This research could define the different subset of CD8+ TILs in cancer and give a clue for different response or modulating therapeutic response of immune checkpoint inhibitor by using CD8+ TIL TEMRA cells. Figure Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2390-2390
Author(s):  
Kristen Snyder ◽  
Hua Zhang ◽  
Barbara Vance ◽  
Carl H. June ◽  
Robert Vonderheide ◽  
...  

Abstract Background: Most tumor antigens are self antigens, therefore an effective immune response to cancer must break self tolerance. Current models hold that lymphopenia results in proliferation toward self antigens. We sought to determine whether the frequency of T cells specific for self antigens, both tumor associated (TA) and non-tumor associated (NTA) were increased in cancer patients, before or after the induction of lymphopenia. Further, we sought to expand T cells specific for self antigens using artificial APCs which signal via CD3 and 4-1BB. Methods: Using a broad panel of HLA-A2 tetramers, we used flow cytometry to enumerate circulating CD8+ T cells which recognize viral (CMV, EBV, flu), NTA self antigens (PR-1, WT1, MART-1 and CEA) and TA self antigens (NY-ESO1, PRAME, Ofa/iLRP, CYP239/190, Survivin and hTERT) in normal donors (ND) (n=13), cancer patients with Ewings sarcoma studied prior to chemotherapy (n=7) and following cytotoxic chemotherapy (n=5). To exclude non-specific tetramer binding, two tetramers with differing fluorochromes were included in each tube and cells simultaneously binding both tetramers were excluded as non-specific and binding frequency to control tetramers was subtracted. Results: The frequency of T cells with specificity for viral antigens (range 0.0%–0.7%) were similar between ND and cancer patients, with no significant increase seen following lymphopenia. For NTA self antigens frequencies prior to chemotherapy were similar between ND and cancer patients respectively, with the lowest mean frequency seen in CEA tetramer specific cells (0.084% ND vs. 0.088% pts), then PR-1 (0.19% ND vs. 0.019% pts), then WT-1 (0.43% ND vs. 0.20% pts) then MART-1 CD8+ specific tetramer cells (0.82% ND vs. 0.83% pts). Remarkably, the frequency of MART-1 specific cells approximates that seen for CMV in both patients and normal donors. For TA self antigens, patients showed trends toward increased frequencies compared to ND but these were not statistically significant. Following lymphopenia, there was no consistent or statistically significant increase in the frequency of cells with specificity for either TA or NTA self antigens. In an attempt to increase the frequency of tumor reactive T cells for potential use in adoptive cellular immunotherapy, artificial APCs were used to deliver a signal via CD3 and 4-1BB and changes in antigen specificity via tetramers were monitored. This method consistently increased the frequency of viral and self antigen specific (TA and NTA) cells with some cultures demonstrating frequencies for individual TA self antigens which approached 10%. Conclusions: 1. Both normal donors and cancer patients demonstrate sizable repertoires of self-reactive T cells, with the frequency of MART-1 specific cells similar to that seen toward CMV. 2. Patients with cancer retain cells which recognize tumor antigens. However, cells recognizing TA self antigens are only marginally increased compared to that found in normal individuals, and are found at frequencies similar to NTA self antigens implicating inefficient priming by the tumor. 3. Lymphopenia does not significantly augment the frequency of cells responding to TA or NTA self antigens. 4. Sizable expansions of T cells responding to TA self antigens can be induced using CD3/4-1BB based expansion.


2004 ◽  
Vol 22 (14_suppl) ◽  
pp. 2612-2612
Author(s):  
F. Schuetz ◽  
K. Ehlert ◽  
G. Bastert ◽  
A. Schneeweiss ◽  
V. Schirrmacher ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document