Th17 responses to autologous dendritic cell vaccine.

2019 ◽  
Vol 37 (8_suppl) ◽  
pp. 132-132
Author(s):  
Gabriel I. Nistor ◽  
Aleksandra J. Poole ◽  
Candace Hsieh ◽  
Hans S. Keirstead ◽  
Robert O. Dillman

132 Background: Dendritic cells (DC) loaded with autologous tumor antigens in vitro, can induce a Th17 phenotype preceding a Th1 type response. We examined this in patients with metastatic melanoma who had been treated in a randomized trial with either DC loaded with autologous tumor antigens (ATA) from self-renewing tumor cells (DCV) or irradiated tumor cells (TCV). Methods: Blood samples were obtained at baseline, one week before three weekly vaccinations (week-0), and at week-4, (one week after the third vaccination). They were analyzed for growth factors, tumor markers and biomarkers using a quantitative, multiplex ELISA. Monocytes, lymphocytes, loaded and unloaded DC were analyzed for cytokine production and intercellular responses using mixed lymphocyte co-cultures (MLC). Results: When compared to baseline or the TCV arm, DCV-treated patients showed reductions in tumor markers, inflammation and angiogenesis in parallel with Th17 pathway activation, IL12 p70 heterodimer expression was detected in monocytes and DC only after antigen loading. The IL12 p40 fraction was present after differentiation and was enhanced after antigen loading. Similarly, the p19 component of IL23 was expressed before DC loading and later enhanced. MLC showed induction of Th17, Th1 and CD8 subpopulations. In the presence of antigen-loaded DCs, the FOXp3 cells were reduced in number, an effect that was canceled by neutralizing the IL12p40 subunit or by the adding tumor cells alone in the MLC. Conclusions: DC produced ex vivo can activate multiple cytotoxic pathways depending on IL12 subunits. Expression of IL23p19 and the shared IL12p40 fraction are responsible for Th1 priming (IL12 incomplete) and Th17 induction (IL23 complete), determinants of the reactive phenotype of naïve DCs. IL12 p70 heterodimer expression after DC loading enables a targeted Th1 type response while neutralizing the IL12 subunit p40, enhances Foxp3 positive T-regs. DC vaccines produced ex vivo could induce a Th1/Th17 cytotoxic response against tumors. In the absence of antigens, however, The Th17 phenotype can become tolerizing in the microenvironment. The findings are important for applications in settings in which tumor infiltration with Th17 correlates with survival, such as in advanced ovarian cancer. Clinical trial information: NCT00436930.

1998 ◽  
Vol 188 (9) ◽  
pp. 1641-1650 ◽  
Author(s):  
Pedro Romero ◽  
P. Rod Dunbar ◽  
Danila Valmori ◽  
Mikaël Pittet ◽  
Graham S. Ogg ◽  
...  

Characterization of cytolytic T lymphocyte (CTL) responses to tumor antigens has been impeded by a lack of direct assays of CTL activity. We have synthesized reagents (“tetramers”) that specifically stain CTLs recognizing melanoma antigens. Tetramer staining of tumor-infiltrated lymph nodes ex vivo revealed high frequencies of tumor-specific CTLs which were antigen-experienced by surface phenotype. In vitro culture of lymph node cells with cytokines resulted in very large expansions of tumor-specific CTLs that were dependent on the presence of tumor cells in the lymph nodes. Tetramer-guided sorting by flow cytometer allowed isolation of melanoma-specific CTLs and confirmation of their specificity and their ability to lyse autologous tumor cells. Our results demonstrate the value of these novel reagents for monitoring tumor-specific CTL responses and for generating CTLs for adoptive immunotherapy. These data also indicate that strong CTL responses to melanoma often occur in vivo, and that the reactive CTLs have substantial proliferative and tumoricidal potential.


Cancers ◽  
2021 ◽  
Vol 13 (15) ◽  
pp. 3818
Author(s):  
Maud Plantinga ◽  
Denise A. M. H. van den Beemt ◽  
Ester Dünnebach ◽  
Stefan Nierkens

Induction of long-lasting immunity by dendritic cells (DCs) makes them attractive candidates for anti-tumor vaccination. Although DC vaccinations are generally considered safe, clinical responses remain inconsistent in clinical trials. This initiated studies to identify subsets of DCs with superior capabilities to induce effective and memory anti-tumor responses. The use of primary DCs has been suggested to overcome the functional limitations of ex vivo monocyte-derived DCs (moDC). The ontogeny of primary DCs has recently been revised by the introduction of DC3, which phenotypically resembles conventional (c)DC2 as well as moDC. Previously, we developed a protocol to generate cDC2s from cord blood (CB)-derived stem cells via a CD115-expressing precursor. Here, we performed index sorting and single-cell RNA-sequencing to define the heterogeneity of in vitro developed DC precursors and identified CD14+CD115+ expressing cells that develop into CD1c++DCs and the remainder cells brought about CD123+DCs, as well as assessed their potency. The maturation status and T-cell activation potential were assessed using flow cytometry. CD123+DCs were specifically prone to take up antigens but only modestly activated T-cells. In contrast, CD1c++ are highly mature and specialized in both naïve as well as antigen-experienced T-cell activation. These findings show in vitro functional diversity between cord blood stem cell-derived CD123+DC and CD1c++DCs and may advance the efficiency of DC-based vaccines.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 929-929
Author(s):  
Matthew J Goldstein ◽  
Bindu Varghese ◽  
Ranjani Rajapaksa ◽  
Joshua Brody ◽  
Shoshana Levy ◽  
...  

Abstract Abstract 929 Background: Recently, we have investigated adoptive cell therapy for treating lymphoma. The efficacy of this maneuver has been demonstrated by curing large established tumors. Specifically, we use active immunization to generate anti-tumor T cells in vivo and transfer these T cells into lymphodepleted recipient mice (Brody J, Goldstein MJ, Czerwinski DK, and Levy R; Blood, 2009). A major challenge in adoptive therapy is the method of generating anti-tumor T cells. Traditionally, tumor-specific T cells are expanded to large numbers ex vivo. Herein, we describe a new, whole-cell vaccine that is effective in inducing anti-tumor T cells in vivo. This vaccine combines tumor antigens with an immune stimulant: irradiated-tumor cells (a source of tumor antigens) are loaded with the TLR agonist CpG (an immune stimulant). Our vaccine approach has several potential advantages: (1) anti-tumor immunity generated by our CpG-loaded, whole-cell vaccine is poly-antigenic and thus, not limited by the expression of a single antigen target on tumor cells; (2) ex vivo expansion may generate large numbers of effector T cells that can induce tumor regression in the short-term, but have a limited ability to maintain a persistent anti-tumor response. Our model avoids ex vivo manipulation of anti-tumor T cells and thus, may preserve and enhance a memory T cell population that sustains the anti-tumor response. Methods: We derived a new pre-B cell lymphoma cell line in the C57BL/6 background. Primary bone marrow cells were isolated from C57BL/6 donor mice and transfected with a recombinant retrovirus containing the Bcr-Abl oncogene. The emerging transformed cell line was designated H11. This cell line expressed the B lineage marker CD19 but was negative for MHC II and surface Ig. Irradiated H11 tumor cells were pre-loaded with CpG for 24 hours and administered to donor mice by daily, sub-cutaneous injections for five days. Donor splenocytes were harvested seven days following vaccination and adoptively transferred into lethally irradiated recipient mice that were subsequently challenged with a lethal dose of H11 tumor cells. Results: Vaccination with CpG-loaded H11 tumor cells (CpG-H11) generated anti-tumor T cells that are effective in adoptive cell therapy. 100% of mice receiving adoptive therapy with vaccine-induced T cells were protected from tumor challenge. In contrast, vaccination of donor mice with untreated H11 tumor was insufficient for generating anti-tumor T cells. Only 20% of mice treated with T cells from these donors were protected from tumor challenge. In spite of the H11 tumor being MHC Class II−, we observed that anti-tumor immunity generated by the CpG-H11 vaccine was CD4 T cell mediated. CD4 T cells were isolated from CpG-H11 vaccinated donors by flow cytometry. Fewer than 1.8×106 CD4 T cells were sufficient to protect 80% of recipient mice from tumor challenge. In contrast, equivalent numbers of donor CD8 T cells provided no benefit. These results strongly suggest that the CpG-H11 vaccine induced cross-presentation of tumor antigens by antigen-presenting cells (APCs). We have demonstrated that CpG-loaded H11 tumor cells can leak CpG into the immediate environment activating nearby APCs. These APCs have greater phagocytic potential and express higher levels of co-stimulatory molecules such as CD40. Ongoing studies will determine whether APCs which encounter the CpG-H11 vaccine but not untreated H11 tumor cells, can stimulate proliferation of anti-tumor T cells. Conclusions: Here we describe a novel, whole-cell vaccine approach that induces anti-tumor T cells for adoptive therapy to treat lymphoma. This vaccine is superior to vaccination with tumor cells alone. We are currently developing this therapy for evaluation in a clinical trial to treat mantle cell lymphoma. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii38-ii39
Author(s):  
Daniela Bota ◽  
David Piccioni ◽  
R LaRocca ◽  
Christopher Duma ◽  
Santosh Kesari ◽  
...  

Abstract GBM standard treatment is associated with poor survival. Adjunctive therapy with patient-specific vaccines may improve outcomes by enhancing anti-GBM immune responses. A multi-institutional phase II clinical trial was designed with a primary objective of 75% survival 15 months after intent-to-treat enrollment. IL-4 and GM-CSF were used to generate dendritic cells (DC) from monocytes. DC were incubated with autologous tumor antigens (ATA) from the lysate of cultured GBM cells to produce each patient-specific DC-ATA vaccine. Each dose was admixed with 500 mcg GM-CSF at the time of subcutaneous injections at weeks 1, 2, 3, 8, 12, 16, 20 and 24. Enrollment has been completed in April 2020 (n=60). Three patients withdrew from the study prior to starting treatment leaving 57 patients for whom data is available. So far 57 patients have received 344 doses; 27 have completed all 8 doses, 11 received fewer than 8 doses at the time they discontinued treatment, 19 are currently in treatment. No patient has discontinued treatment because of toxicity. 9 pt had died and the preliminary 12 months overall survival is 74%. In a preliminary serologic analysis 12 of 16 patients (75%) had an increase in markers associated with Th1/NK, Th2/immunoglobulins, and Th2 hypersensitivity (eotaxins, IgE and IL17F) by week-3; 9 of 15 (60%) had a decrease in angiogenesis factors, growth factors, and tumor markers by week-8. Immunologic data for all 55 patients who received at least two injections will be available November 2020. This patient-specific DC-ATA immunotherapy approach is feasible, is associated with changes in serologic markers, and may be increasing intratumor inflammation that may be associated with on-target toxicity and efficacy. A interim survival analysis will be conducted in mid-October 2020, 15 months after the 28th patient was enrolled; results will be available November 2020 [Clinicaltrials.gov NCT03400917].


2009 ◽  
Vol 29 (3) ◽  
pp. 265-270
Author(s):  
Dan-feng XU ◽  
Yi GAO ◽  
Yu-shan LIU ◽  
Xin-gang CUI ◽  
Jian-ping CHE ◽  
...  

Cancers ◽  
2021 ◽  
Vol 13 (7) ◽  
pp. 1577
Author(s):  
Matteo Tanzi ◽  
Michela Consonni ◽  
Michela Falco ◽  
Federica Ferulli ◽  
Enrica Montini ◽  
...  

The limited efficacy of Natural Killer (NK) cell-based immunotherapy results in part from the suboptimal expansion and persistence of the infused cells. Recent reports suggest that the generation of NK cells with memory-like properties upon in vitro activation with defined cytokines might be an effective way of ensuring long-lasting NK cell function in vivo. Here, we demonstrate that activation with IL-12, IL-15 and IL-18 followed by a one-week culture with optimal doses of Interleukin (IL-2) and IL-15 generates substantial numbers of memory-like NK cells able to persist for at least three weeks when injected into NOD scid gamma (NSG) mice. This approach induces haploidentical donor-derived memory-like NK cells that are highly lytic against patients’ myeloid or lymphoid leukemia blasts, independent of the presence of alloreactive cell populations in the donor and with negligible reactivity against patients’ non-malignant cells. Memory-like NK cells able to lyse autologous tumor cells can also be generated from patients with solid malignancies. The anti-tumor activity of allogenic and autologous memory-like NK cells is significantly greater than that displayed by NK cells stimulated overnight with IL-2, supporting their potential therapeutic value both in patients affected by high-risk acute leukemia after haploidentical hematopoietic stem cell transplantation and in patients with advanced solid malignancies.


2010 ◽  
Vol 8 (1) ◽  
pp. 52 ◽  
Author(s):  
Laura Fiammenghi ◽  
Valentina Ancarani ◽  
Tilman Rosales ◽  
Jay R Knutson ◽  
Massimiliano Petrini ◽  
...  

2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi174-vi174
Author(s):  
Bianca Walter ◽  
Denis Canjuga ◽  
Simge G Yuez ◽  
Michael Ghosh ◽  
Przemyslaw Bozko ◽  
...  

Abstract Glioblastoma are incurable aggressive tumors and remain a therapeutic challenge. Glioblastoma frequently harbor alterations in the retinoblastoma pathway with subsequent cell cycle abnormalities. Here, we aimed to investigate the anti-glioma activity of the cell cycle-stabilizing compound Argyrin F and its potential treatment-induced vulnerabilities to exploit possibilities for novel combination therapies. We investigated cell viability, clonogenic survival, cell cycle status and immunoblots of human and murine glioma cells treated with Argyrin F. Moreover, we established an ex vivo glioma model using residual freshly resected tissue from patients, i.e. patient-derived microtumors (PDMs). Additionally, we extracted autologous tumor infiltrating lymphocytes (TILs) to perform co-culturing experiments. We performed mass spectrometry-based immunopeptidomics and used the orthotopic syngeneic SMA560/VM/Dk glioma mouse model. Argyrin F displayed anti-glioma efficacy in glioma cell lines in vitro and in PDM models ex vivo. Moreover, Argyrin F treatment induced cell cycle arrest, reduced clonogenic survival in vitro and prolonged survival in vivo. Argyrin F-treated SMA560 glioma displayed 4.6-fold more glioma-infiltrating CD8+ T cells. We discovered a distinctive treatment-induced immunopeptidome. Combination of Argyrin F plus PD-1 antibody increased cellular toxicity in PDM/TILs co-cultures ex vivo and prolonged overall survival compared with monotherapies in vivo. We conclude that our experimental data suggest a novel combination of Argyrin F plus PD-1 blockade and its clinical translation.


Author(s):  
Libuše Janská ◽  
Libi Anandi ◽  
Nell C. Kirchberger ◽  
Zoran S. Marinkovic ◽  
Logan T. Schachtner ◽  
...  

There is an urgent need for accurate, scalable, and cost-efficient experimental systems to model the complexity of the tumor microenvironment. Here, we detail how to fabricate and use the Metabolic Microenvironment Chamber (MEMIC) – a 3D-printed ex vivo model of intratumoral heterogeneity. A major driver of the cellular and molecular diversity in tumors is the accessibility to the blood stream that provides key resources such as oxygen and nutrients. While some tumor cells have direct access to these resources, many others must survive under progressively more ischemic environments as they reside further from the vasculature. The MEMIC is designed to simulate the differential access to nutrients and allows co-culturing different cell types, such as tumor and immune cells. This system is optimized for live imaging and other microscopy-based approaches, and it is a powerful tool to study tumor features such as the effect of nutrient scarcity on tumor-stroma interactions. Due to its adaptable design and full experimental control, the MEMIC provide insights into the tumor microenvironment that would be difficult to obtain via other methods. As a proof of principle, we show that cells sense gradual changes in metabolite concentration resulting in multicellular spatial patterns of signal activation and cell proliferation. To illustrate the ease of studying cell-cell interactions in the MEMIC, we show that ischemic macrophages reduce epithelial features in neighboring tumor cells. We propose the MEMIC as a complement to standard in vitro and in vivo experiments, diversifying the tools available to accurately model, perturb, and monitor the tumor microenvironment, as well as to understand how extracellular metabolites affect other processes such as wound healing and stem cell differentiation.


Sign in / Sign up

Export Citation Format

Share Document