Fig 6 - uploaded by Andreas Eigler
Content may be subject to copyright.
Tumor lysate-pulsed DCs induce CTLs specific for Panc-1 carcinoma cells. MNC were removed from the cocultures with unpulsed, lysate-pulsed, or lysate-plus KLH-pulsed DCs and were incubated with 51 Cr-labeled Panc-1 tumor cells at the indicated ratios. The HLA-A2 gastric carcinoma cell line Kato-III served as a negative control. Data represent means SE of five independent experiments. 

Tumor lysate-pulsed DCs induce CTLs specific for Panc-1 carcinoma cells. MNC were removed from the cocultures with unpulsed, lysate-pulsed, or lysate-plus KLH-pulsed DCs and were incubated with 51 Cr-labeled Panc-1 tumor cells at the indicated ratios. The HLA-A2 gastric carcinoma cell line Kato-III served as a negative control. Data represent means SE of five independent experiments. 

Source publication
Article
Full-text available
Dendritic cells (DCs) are potent antigen-presenting cells and play a pivotal role in T cell-mediated immunity. DCs have been shown to induce strong antitumor immune responses in vitro and in vivo, and their efficacy is being investigated in clinical trials. Compared with vaccination strategies directed against a single tumor antigen, tumor-cell lys...

Context in source publication

Context 1
... After four weekly stimulations with unpulsed, lysate-pulsed, or lysate-plus KLH-pulsed DCs, we removed the nonadherent cells from the cocul- tures and incubated them with 51 Cr-labeled Panc-1 tumor cells or KATO-III gastric carcinoma cells (HLA-A2 ). MNC from the cocul- ture with lysate-pulsed DCs were able to specifically lyse Panc-1 tumor cells (Fig. 6). Lytic activity was enhanced in the cocultures with lysate-plus KLH-pulsed DCs. KATO-III was lysed to a minor extent only, and the lysis was comparable with that of Panc-1 by MNC that had been cocultured with unpulsed DCs. To test whether tumor cell lysis was MHC class I-restricted, we preincubated the tumor cells with a MHC class ...

Citations

... A diverse array of target antigens is included in TCL, a combination of proteins generated by the induced lysis of tumor cells [158]. In addition, knowledge of important tumor-associated antigens (TAAs) and targets, including possibly undiscovered TAAs, is not a prerequisite for the TCL method of immunization [159,160]. TCL is an attractive vaccination candidate due to its simplicity of production and storage, absence of host-specific constraints, and unnecessity of prior knowledge about particular TAAs [161]. However, it should be noted that TCL also comprises naturally present immunosuppressants in cancer cells, including hyaluronan, which is recognized for its ability to induce tolerogenic maturation of DCs and Mφ rather than immunogenic maturation [162]. ...
Article
Full-text available
Antigenic cell fragments, pathogen-associated molecular patterns, and other immunostimulants in bacterial lysates or extracts may induce local and systemic immune responses in specific and nonspecific paradigms. Based on current knowledge, this review aimed to determine whether bacterial lysate has comparable functions in infectious diseases and cancer treatment. In infectious diseases, including respiratory and urinary tract infections, immune system activation by bacterial lysate can identify and combat pathogens. Commercially available bacterial lysates, including OM-85, Ismigen, Lantigen B, and LW 50020, were effective in children and adults in treating respiratory tract infections, chronic obstructive pulmonary disease, rhinitis, and rhinosinusitis with varying degrees of success. Moreover, OM-89, Uromune, Urovac, Urivac, and ExPEC4V showed therapeutic benefits in controlling urinary tract infections in adults, especially women. Bacterial lysate-based therapeutics are safe, well-tolerated, and have few side effects, making them a good alternative for infectious disease management. Furthermore, a nonspecific immunomodulation by bacterial lysates may stimulate innate immunity, benefiting cancer treatment. “Coley’s vaccine” has been used to treat sarcomas, carcinomas, lymphomas, melanomas, and myelomas with varying outcomes. Later, several similar bacterial lysate-based therapeutics have been developed to treat cancers, including bladder cancer, non-small cell lung cancer, and myeloma; among them, BCG for in situ bladder cancer is well-known. Proinflammatory cytokines, including IL-1, IL-6, IL-12, and TNF-α, may activate bacterial antigen-specific adaptive responses that could restore tumor antigen recognition and response by tumor-specific type 1 helper cells and cytotoxic T cells; therefore, bacterial lysates are worth investigating as a vaccination adjuvants or add-on therapies for several cancers.
... The intracellular contents of ADSC spheroids were extracted using a simple freezing-thawing approach [18][19][20][21] . ADSC spheroids were harvested on day 6 of incubation. ...
Article
Full-text available
Immunomodulatory properties of mesenchymal stem cells are widely studied, supporting the use of MSCs as cell-based therapy in immunological diseases. This study aims to generate cell-free MSC extract and improves their immunomodulatory potential. Intracellular extracts were prepared from adipose-derived stem cells (ADSC) spheroid via a freeze-thawing method. The immunomodulatory capacities of ADSC spheroid extracts were investigated in vitro, including lymphocyte proliferation, T regulatory cell expansion, and macrophage assays. A comparative study was conducted with ADSC monolayer extract. The key immunomodulatory mediators presented in ADSC extract were identified. The results revealed that ADSC spheroid extract could suppress lymphocyte activation while enhancing T regulatory cell expansion. Immunomodulatory molecules such as COX-2, TSG-6, and TGF-β1 were upregulated in ADSC priming via spheroid culture. Selective inhibition of COX-2 abrogates the effect of ADSC extract on inducing T regulatory cell expansion. Thus, ADSC spheroid extract gains high efficacy in regulating the immune responses which are associated in part by COX-2 generation. Furthermore, ADSC spheroid extract possessed a potent anti-inflammation by manipulation of TNF-α production from LPS-activated macrophage. Our current study has highlighted the opportunity of using cell-free extracts from adipose tissue-derived mesenchymal stem cells spheroid as novel immunomodulators for the treatment of immunological-associated diseases.
... In addition to synthetic validated tumor antigens (e.g., peptides and mRNAs), the use of tumor cell-derived components (e.g., whole cells, cell lysates, cell-derived vesicles) as antigenic material has also been proposed [73,[87][88][89]. Tumor cell membranes, for example, contain a great variety of antigenic motifs, especially neoantigens and provide an excellent platform for personalized antigen delivery. ...
Article
Full-text available
Intelligent nanomedicine is currently one of the most active frontiers in cancer therapy development. Empowered by the recent progresses of nanobiotechnology, a new generation of multifunctional nanotherapeutics and imaging platforms has remarkably improved our capability to cope with the highly heterogeneous and complicated nature of cancer. With rationally designed multifunctionality and programmable assembly of functional subunits, the in vivo behaviors of intelligent nanosystems have become increasingly tunable, making them more efficient in performing sophisticated actions in physiological and pathological microenvironments. In recent years, intelligent nanomaterial-based theranostic platforms have showed great potential in tumor-targeted delivery, biological barrier circumvention, multi-responsive tumor sensing and drug release, as well as convergence with precise medication approaches such as personalized tumor vaccines. On the other hand, the increasing system complexity of anti-cancer nanomedicines also pose significant challenges in characterization, monitoring and clinical use, requesting a more comprehensive and dynamic understanding of nano-bio interactions. This review aims to briefly summarize the recent progresses achieved by intelligent nanomaterials in tumor-targeted drug delivery, tumor immunotherapy and temporospatially specific tumor imaging, as well as important advances of our knowledge on their interaction with biological systems. In the perspective of clinical translation, we have further discussed the major possibilities provided by disease-oriented development of anti-cancer nanomaterials, highlighting the critical importance clinically-oriented system design.
... In this paper, we describe a novel platform to manufacture tumor-specific T cells for potential use in immunotherapy through the ex vivo-modulated ablation of tumor cells via PBNP-PTT ( Figure 1). Antigen-specific T cells used in ATCT for cancer have traditionally been developed by pulsing autologous DCs with either tumor-associated antigen peptides or tumor cell lysates [38][39][40][41][42]. Here, we applied PBNP-PTT to GBM cells ex vivo to generate tumor cell death and immunogenicity (Figure 2, Supplementary Figure S10), building on our published work in different solid tumor models in vivo [8,13]. ...
Article
Full-text available
Background: Adoptive T cell therapy (ATCT) has been successful in treating hematological malignancies and is currently under investigation for solid-tumor therapy. In contrast to existing chimeric antigen receptor (CAR) T cell and/or antigen-specific T cell approaches, which require known targets, and responsive to the need for targeting a broad repertoire of antigens in solid tumors, we describe the first use of immunostimulatory photothermal nanoparticles to generate tumor-specific T cells. Methods: Specifically, we subject whole tumor cells to Prussian blue nanoparticle-based photothermal therapy (PBNP-PTT) before culturing with dendritic cells (DCs), and subsequent stimulation of T cells. This strategy differs from previous approaches using tumor cell lysates because we use nanoparticles to mediate thermal and immunogenic cell death in tumor cells, rendering them enhanced antigen sources. Results: In proof-of-concept studies using two glioblastoma (GBM) tumor cell lines, we first demonstrated that when PBNP-PTT was administered at a "thermal dose" targeted to induce the immunogenicity of U87 GBM cells, we effectively expanded U87-specific T cells. Further, we found that DCs cultured ex vivo with PBNP-PTT-treated U87 cells enabled 9- to 30-fold expansion of CD4+ and CD8+ T cells. Upon co-culture with target U87 cells, these T cells secreted interferon-ɣ in a tumor-specific and dose-dependent manner (up to 647-fold over controls). Furthermore, T cells manufactured using PBNP-PTT ex vivo expansion elicited specific cytolytic activity against target U87 cells (donor-dependent 32-93% killing at an effector to target cell (E:T) ratio of 20:1) while sparing normal human astrocytes and peripheral blood mononuclear cells from the same donors. In contrast, T cells generated using U87 cell lysates expanded only 6- to 24-fold and killed 2- to 3-fold less U87 target cells at matched E:T ratios compared with T cell products expanded using the PBNP-PTT approach. These results were reproducible even when a different GBM cell line (SNB19) was used, wherein the PBNP-PTT-mediated approach resulted in a 7- to 39-fold expansion of T cells, which elicited 25-66% killing of the SNB19 cells at an E:T ratio of 20:1, depending on the donor. Conclusions: These findings provide proof-of-concept data supporting the use of PBNP-PTT to stimulate and expand tumor-specific T cells ex vivo for potential use as an adoptive T cell therapy approach for the treatment of patients with solid tumors.
... In another study, it has been indicated that human gastric tumor lysates loaded DCs can boost the proliferation of CD3 + T cells (36). In accordance with our results, Schnurr et al. have reported the increased IL-12 secretion by panc-1 tumor cell lysates loaded DCs (37). In addition, in a breast tumor-bearing human-SCID model, suppression of PD-L1 boosted DC maturation, proliferation, and IL-12 secretion, as well as T-cell-mediated responses (38). ...
Article
Full-text available
Dendritic cell (DC)-based immunotherapy has increased interest among anti-cancer immunotherapies. Nevertheless, the immunosuppressive mechanisms in the tumor milieu, e.g., inhibitory immune checkpoint molecules, have been implicated in diminishing the efficacy of DC-mediated anti-tumoral immune responses. Therefore, the main challenge is to overcome inhibitory immune checkpoint molecules and provoke efficient T-cell responses to antigens specifically expressed by cancerous cells. Among the inhibitory immune checkpoints, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) expression on DCs diminishes their maturation and antigen presentation capability. Accordingly, we hypothesized that the expression of CTLA-4 on DCs inhibits the T cell-mediated anti-tumoral responses generated following the presentation of tumor antigens by DCs to T lymphocytes. In this study, we loaded colorectal cancer (CRC) cell lysate on DCs and inhibited the expression of CTLA-4 by small interfering RNA (siRNA) in them to investigate the DCs’ functional and phenotypical features, and T-cell mediated responses following DC/T cell co-culture. Our results demonstrated that blockade of CTLA-4 could promote stimulatory properties of DCs. In addition, CTLA-4 silenced CRC cell lysate-loaded DCs compared to the DCs without CTLA-4 silencing resulted in augmented T cell proliferation and cytokine production, i.e., IFN-γ and IL-4. Taken together, our findings suggest CTLA-4 silenced CRC cell lysate-loaded DCs as a promising therapeutic approach however further studies are needed before this strategy can be used in clinical practice.
... The protein concentration of the lysate was determined by commercial assay (Cat. No. 5000002, BioRad) 28 . ...
... To determine MHC-I restriction of cancer cells lysis, the target cells were preincubated with MHC-I blocking antibody w6/32 (Cat. No. sc-32235, Santa Cruz, 10 μg/mL) for 2 h at 37°C before coculture 28 . The result was further verified by using immunodeficient FO-1 melanoma cell line 10 . ...
Article
Full-text available
Mitochondrial dynamics can regulate Major Histocompatibility Complex (MHC)-I antigen expression by cancer cells and their immunogenicity in mice and in patients with malignancies. A crucial role in the mitochondrial fragmentation connection with immunogenicity is played by the IRE1α-XBP-1s axis. XBP-1s is a transcription factor for aminopeptidase TPP2, which inhibits MHC-I complex cell surface expression likely by degrading tumor antigen peptides. Mitochondrial fission inhibition with Mdivi-1 upregulates MHC-I expression on cancer cells and enhances the efficacy of adoptive T cell therapy in patient-derived tumor models. Therefore mitochondrial fission inhibition might provide an approach to enhance the efficacy of T cell-based immunotherapy. Cancer cells downregulate surface expression of major histocompatibility complex I (MHC-I) for immune evasion. Here, the authors show that rapid mitochondrial fission activates the ER-stress response leading to reduced MHC-I complex formation and cell surface expression in solid cancer cells; moreover inhibition of mitochondrial fission increases the immune-mediated anticancer response in murine models.
... [3][4][5] Immunogenic DCs can inhibit tumor progression via both proliferation and activation of functional tumor-specific T cells, such as type I helper T (Th1) cells and cytotoxic T lymphocytes (CTLs). 6,7 In contrast, tolerogenic DCs can lead to even cancer progression. 8,9 DC-based vaccines have been extensively investigated as a feasible approach for cancer immunotherapy to enhance tumor antigen-specific immune responses. ...
Article
Full-text available
For clinical application by dendritic cell (DC)-based cancer immunotherapy, a proper adjuvant system to elicit a strong anticancer immune response is needed. Here, we investigated the potential of chorismate mutase (TBCM, Rv1885c), a putative Mycobacterium tuberculosis (TB) virulence factor, as an immunoadjuvant in DC-based tumor immunotherapy. First, we found that TBCM functionally activated DCs by upregulating costimulatory molecules, increasing the secretion of proinflammatory cytokines, enhancing migration and inducing the Th1-type immune response in a dose-dependent manner via TLR4-mediated signaling. In addition, subcutaneous injection of TBCM-activated DCs loaded with cell lysates led to reduced tumor mass, enhanced mouse survival and lowered tumor incidence in lung carcinoma (LLC) cell-bearing mice. This is mainly mediated by functional cytotoxic T lymphocyte-mediated oncolytic activity and inhibition of cancer proliferation- and metastasis-related genes. Moreover, TBCM-induced DCs can also generate memory CD4 T cells and exert long-term tumor prevention effects. In conclusion, our findings suggest that TBCM (Rv1885c), a novel TLR4 agonist, could be used as an immunoadjuvant for DC-based cancer immunotherapy.
... CT-26 cell and CCSC lysates were prepared as previously described [21] . Briefly, 3 × 10 6 CT-26 cells and CCSCs in 1 ml PBS were lysed by five cycles of repetitive rapid freezing in liquid nitrogen and thawing in a 37°C water bath. ...
Article
Full-text available
Dendritic cell (DC)-based immunotherapy has been a promising strategy for colon cancer therapy, but the efficacy of dendritic cell vaccines is in part limited by immunogenicity of loaded antigens. In this study, we aimed to identify a putative tumor antigen that can generate or enhance anti-tumor immune responses against colon cancer. CD44+ colon cancer stem cells (CCSCs) were isolated from mouse colorectal carcinoma CT-26 cell cultures and induced to form defective ribosomal products-containing autophagosome-rich blebs (DRibbles) by treatment with rapamycin, bortezomib, and ammonium chloride. DRibbles were characterized by western blot and transmission electron microscopy. DCs generated from the mice bone marrow monocytes were cocultured with DRibbles, then surface markers of DCs were analyzed by flow cytometry. Meanwhile, the efficacy of DRibble-DCs was examined in vivo. Our results showed that CCSC-derived DRibbles upregulated CD80, CD86, major histocompatibility complex (MHC)-I, and MHC-II on DCs and induced proliferation of mouse splenic lymphocytes and CD8+ T cells. In a model of colorectal carcinoma using BALB/c mice with robust tumor growth and mortality, DC vaccine pulsed with CCSC-derived DRibbles suppressed tumor growth and extended survival. A lactate dehydrogenase test indicated a strong cytolytic activity of cytotoxic T-cells derived from mice vaccinated with CCSC-derived DRibbles against CT-26 cells. Furthermore, flow cytometry analyses showed that the percentages of IFN-γ-producing CD8+ T-cells were increased in SD-DC group compare with the other groups. These findings provide a rationale for novel immunotherapeutic anti-tumor approaches based on DRibbles derived from colon cancer stem cells.
... The differentiation step typically relies on the presence of GM-CSF and IL-4 [60], with the latter suppressing monocyte-derived macrophage generation and actively promoting generation of MoDCs via induction of DC-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN; CD209) [61,62]. This combination is considered to be the "gold standard" in GMP facilities for MoDC differentiation, yet concentrations for both cytokines may range between 400 and 1,000 IU/mL [63][64][65][66]. This first step typically requires 5-7 days and transforms monocytes into immature MoDCs. ...
... In a different approach, tumor lysates were tested to increase the availability of immunogenic antigens, comprising lysates from tumor cell lines (e.g., KATO-III [65,93]) or autologous patient tumor lysates [46,47,94]. Using cell lines provides the advantage of preparing readily available lysates in advance; however, again antigen loss of the patient's tumor cannot be addressed. ...
Article
Full-text available
Dendritic cells (DCs) are pivotal regulators of immune responses, specialized in antigen presentation and bridging the gap between the innate and adaptive immune system. Due to these key features, DCs have become a pillar of the continuously growing field of cellular therapies. Here we review recent advances in good manufacturing practice strategies and their individual specificities in relation to DC production for clinical applications. These take into account both small-scale experimental approaches as well as automated systems for patient care.
... This was quanti ed by PI and Annexin V uptake by ow cytometry (Fig. 4F), Th1 cytokines production by speci c ELISAs (Fig. 4G) and residual E/T ratio by ow cytometry (Fig. 4H) compared with control group, respectively. In addition, preincubating cancer cells with neutralizing anti-MHC-I antibody [21] abrogated most of the cytotoxicity on target cancer cells. Similar results can be replicated by using melanoma cell line FO-1, a cultured cell line lost MHC-I expression completely due to a defect in B2M gene expression [22], further con rming the restriction and indispensability of MHC-I in cytotoxic function. ...
... The protein concentration of the lysate was determined by commercial assay (Cat. No. 5000002, BioRad) [21]. ...
... DCs were generated as previously described with slight modi cations [18,21]. In brief, peripheral blood mononuclear cells (PBMCs) were isolated from the peripheral blood of patients with HNSCC or NSCLC by SepMate™ tube for density gradient centrifugation (Cat. ...
Preprint
Full-text available
Hypoexpression of human major histocompatibility complex (MHC) class I is widely known to be an important strategy of immune evasion in most cases of malignancies that lead to poor prognosis. We demonstrated that mitochondrial dynamics can be exploited as an efficient target in regulating MHC-I expression and cancer immunogenicity. Clinically, MHC-I expression and fragmentation of mitochondria are both closely associated to patient survival but are negatively correlated to one another. Mechanistically, it was observed that endoplasmic reticulum (ER) stress, an integrated signal transduction pathway activated in most rapidly proliferating tumor cells, played a crucial role in connecting mitochondrial fragmentation and cancer cell immunogenicity particularly via the IRE1α-XBP-1 s axis. XBP-1 s, which is activated by imbalanced mitochondrial fission and prolonged oxidative stress, served as a potent transcription factor, promoted the expression of aminopeptidase TPP2 and destructed the applicable antigenic peptide to impede MHC-I complex maturation and the activation of adaptive immune system upon cancer antigen. Our findings highlight the importance of mitochondrial dynamics in determining solid tumor immunogenicity and suggest that mediating mitochondrial fragmentation might provide a novel approach in anti-tumor immunotherapy.