- Research
- Open access
- Published:
A fermented Mistletoe (Viscum album L.) extract elicits markers characteristic for immunogenic cell death driven by endoplasmic reticulum stress in vitro
BMC Complementary Medicine and Therapies volume 25, Article number: 175 (2025)
Abstract
Background
Immune evasion is a characteristic hallmark of cancer. Immunotherapies aim to activate and support the body's immune system to recognize and fight tumor cells. Induction of immunogenic cell death (ICD) and the associated activation of danger signaling pathways can increase the immunogenicity of tumor cells. Therapeutic ICD stimuli activate endoplasmic reticulum stress pathways and apoptosis leading to the cellular expression of damage-associated molecular patterns (DAMPs).
The aim of our in vitro study was to investigate whether mistletoe extracts induce characteristics of immunogenic tumor cell death in cancer cell lines.
Methods
Three human breast cancer cell lines and one murine melanoma cell line (SKBR3, MDA-MB-231, MCF-7, and B16F10) were treated with aqueous, fermented Viscum album extract (VAE: Iscador Qu spec.) and taxol or tunicamycin as positive controls, respectively. To investigate whether VAE induces ribotoxic stress, we measured the ER stress regulators p-eIF2a, ATF4, and CHOP by Western blot. Cell surface exposure of DAMPs (calreticulin, heat shock proteins hsp70 and hsp90), apoptosis and induction of mitochondrial reactive oxygen species (ROS) were assessed by flow cytometry. HMGB1 and ATP were quantified by ELISA and chemiluminescence assay, respectively.
Results
Treatment with VAE resulted in phosphorylation of eIF2α in all cancer cell lines tested and increased calreticulin (CRT) exposure on the surface of pre-apoptotic SKBR3 breast cancer and B16F10 mouse melanoma cells. VAE exerted a concentration-dependent effect in all cell lines, resulting in a significantly increased exposure of three DAMPs (CRT, hsp70 and hsp90) on the surface of early apoptotic cells. Furthermore, VAE elevated mitochondrial ROS production and the release of ATP. HMGB1 release was not induced by VAE.
Conclusions
In this in vitro study, we demonstrated for the first time the potential of a mistletoe extract to induce surrogate markers of immunogenic cancer cell death. This is a primary step in investigating the potential of VAEs to contribute to ICD-induced tumor-specific immune activation.
Background
Tumors use various escape mechanisms to prevent the patient's immune system from recognizing and eliminating them [1, 2]. In recent years, the immunological approach to cancer treatment has come to the focus of attention with the aim of breaking the immunological tolerance to tumor cells. Particular attention has been paid to the induction of immunogenic cancer cell death, which is an important step in initiating a specific anti-cancer T cell response. Triggering an immunogenic cancer cell death can make an existing tumor recognizable and attackable again for the immune system [3].
Several factors are responsible for the immunogenicity of cell death. These include the intrinsic antigenicity of the cells, the induction of stress responses prior to cell death, the nature of the cytotoxic reagent, the death pathway involved, and the presence of immune cells capable of responding. An ideal inducer of immunogenic cell death (ICD) activates different pathways such as apoptotic cell death, endoplasmic reticulum (ER) stress response, induction of reactive oxygen species (ROS), autophagy, necroptosis or secondary necrosis. Activation of these pathways leads to the expression of damage-associated molecular patterns (DAMPs) such as phosphatidylserine, chaperones such as calreticulin, heat shock proteins (e.g. hsp70, hsp90), HMGB1, ATP and others on the cell surface of or secreted by dying cells [3,4,5].
When the endoplasmic reticulum is stressed, proteins accumulate due to improper folding, modulation, or trafficking. As a result, three stress sensor proteins (IRE1, PERK, ATF6) initiate the unfolded protein response (UPR) signaling pathway [6]. The protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) arm regulates the translational response of the UPR. PERK is a transmembrane protein with a stress-sensing domain and a protein kinase domain. Upon ER stress, PERK dimerizes and promotes its autophosphorylation and activation. Activated PERK phosphorylates eukaryotic translation initiation factor 2 A (eIF2α), resulting in a transient slowing of the overall rate of translation initiation, the prevention of further protein synthesis [7], and the induction of activating transcription factor 4 (ATF4). ATF4 contributes to the increased folding capacity of ER proteins.
The main goal of UPR is to restore protein homeostasis to ensure cell survival. However, under prolonged and severe ER stress and sustained PERK activation, a signaling switch occurs that leads to apoptosis and cell death. The activated transcription factor C/EBP-homologous protein (CHOP) is involved in the regulation of apoptosis [8].
The induction of ER stress triggers danger signaling pathways leading to the release of danger signals and apoptosis [9, 10]. Ideally, the release of DAMPs, danger signals released by injured cells or tissues, into the tumor microenvironment initiates the so-called cancer-immunity cycle [11]. DAMPs attract and activate dendritic cells (DCs), which in turn engulf and digest tumor cell fragments. Activated DCs transport tumor cell-specific antigens to the lymph nodes, where they encounter and activate naive specific T cells, followed by their clonal expansion and dissemination throughout the body. When the activated tumor-specific cytotoxic T cells encounter the tumor cells, they can attack and eliminate them. In addition, a systemic protective anti-tumor immunity, the so-called abscopal immune response, is possible [12].
The first step in the cancer immunity cycle is the induction of immunogenic tumor cell death, which leads to the release of tumor cell-specific and/or tumor-associated antigens and DAMPs. Known inducers of immunogenic cell death include chemotherapeutics such as anthracyclines, cyclophosphamide, certain endocrine and targeted therapies, photodynamic and radiation therapy [13, 14]. ICD inducers fit well into the concept of intratumoral immunotherapy. Local application of immunostimulatory reagents into the tumor tissue aims to induce a strong antitumor immune response using the tumor as its own vaccine. In combination with systemic immunotherapies, this could lead to an improved response rate and better tolerability of the treatment [15, 16].
European mistletoe (Viscum album L.) belongs to the Santalaceae family and is an evergreen, semi-parasitic plant. Viscum album ssp. album grows on a variety of trees, such as Quercus robur L. or Malus domestica. Mistletoe preparations are prescribed to cancer patients as supportive treatment, either alone or in combination with conventional oncological therapies. Aqueous mistletoe preparations contain various bioactive compounds such as mistletoe lectins (MLs), viscotoxins, and polysaccharides. These components, especially the MLs and viscotoxins, have been chemically and pharmacologically characterized in numerous studies [17,18,19,20,21]. Urech et al. specifically investigated the MLs and viscotoxins in the fermented mistletoe preparation we used [22]. Some of the known antitumor effects of mistletoe preparations are immunomodulatory activities, induction of apoptosis, and cell cycle arrest [23,24,25].
The aim of our study was to investigate whether mistletoe extracts induce cancer cell death with molecular features of immunogenic cell death in vitro. This is a first step in exploring the potential of mistletoe extracts to contribute to the stimulation of an anti-tumor immune response by inducing the release of cell death related immunogenic signals.
Methods
Extracts and reagents
The aqueous, fermented mistletoe preparation Iscador Qu spec. 20 mg (VAE, from the host trees Quercus robur and Q. petraea, lot 9019/70, expiration date 07–2021: total mistletoe lectin concentration 1689 ng/mL and lot 1021/70, expiration date 07–2023: total mistletoe lectin concentration 1592 ng/mL) was obtained from Iscador AG (Arlesheim, Switzerland). To prepare Iscador, mistletoe leaves, stems and berries are harvested in mid-summer and mid-winter. The fresh plant parts are ground and fermented for 3 days after the addition of Lactobacillus plantarum. The solution is separated from the solid material and the aqueous summer and winter extracts obtained are subsequently mixed. Iscador is authorized by Swissmedic, the Swiss regulatory authority for medicines and medical devices. The detailed manufacturing process of the extract is described in the Anthroposophic Pharmaceutical Codex of the International Association of Anthroposophic Pharmacists [26].
Taxol (#T7402), tunicamycin (#T7765) and mitoxantrone dihydrochloride (#M6545) were purchased from Sigma-Aldrich GmbH/Merck & Cie (Buchs, CH).
Cell lines and cell culture
The human breast carcinoma cell lines SKBR3 (RRID:CVCL_0033), MCF-7 (RRID:CVCL_0031), and MDA-MB-231 (RRID:CVCL_0062) (obtained from DSMZ, German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany) and the mouse melanoma cell line B16F10 (RRID:CVCL_0159) (cells kindly provided by Prof. A. Zippelius, University of Basel, Switzerland) were used for the experiments.
The cell lines were grown in the following media:
-
SKBR3 in McCoy’s 5A (Sigma-Aldrich), 10% fetal calf serum, 1% penicillin/streptomycin, 2 mM L-glutamine (Sigma-Aldrich)
-
MDA-MB-231 in DMEM (hg) medium, 10% fetal calf serum and 1% penicillin/streptomycin (Sigma-Aldrich)
-
MCF-7 in DMEM (hg) medium, 10% fetal calf serum, 1% penicillin/streptomycin, 1 mM sodium pyruvate, and 10 μg/mL insulin (Sigma-Aldrich)
-
B16F10 in DMEM (hg) medium, 10% fetal calf serum, 1% penicillin/streptomycin, 1 mM sodium pyruvate
Cells were grown exponentially in a humidified atmosphere of 5% CO2 at 37 °C and harvested from subconfluent monolayers using trypsin–EDTA (Sigma-Aldrich).
Drug concentrations in the assays
For each of the different cell lines, we selected the concentrations of VAE and the reagents used as positive controls at which we could detect concentration-dependent apoptosis as well as the expression of DAMPs. The preliminary dose–response data are provided in Supplementary Material 3.
Western blot analysis
For Western blotting, cells were treated with experimental or control medium for 24 h in 6-well cell culture dishes. Total protein was extracted using RIPA Lysis and Extraction buffer (#89900, Thermo Fisher Scientific) in the presence of phosphatase inhibitors (#78440, Thermo Fisher Scientific). Protein concentration was determined by the Bradford assay using bovine serum albumin (Sigma Aldrich) as a protein standard.
Proteins were separated by SDS–polyacrylamide gel electrophoresis using the BioRad system with 4–20% MP-TGX stain-free polyacrylamide gels (stain-free activation of total protein for 1 min in the UV box of a Vilber Fusion Solo S imaging system) (BioRad). Proteins were transferred to PVDF membranes using the Trans-Blot Turbo system (BioRad) and incubated with primary antibodies (recombinant anti-EIF2S1 (phospho S51) antibody [E90] Abcam Cat# ab32157, RRID: AB_732117); recombinant anti-ATF-4 antibody [EPR18111], (Abcam Cat# ab184909, RRID: AB_2819059); anti-DDIT3 antibody [9 C8], (Abcam Cat# ab11419, RRID: AB_298023) overnight at 4 °C and then with goat-anti-rabbit IgG H&L (HRP) (Abcam Cat# AB205718) for 1 h at room temperature, followed by Clarity Western ECL substrate (Biorad, #1705060) incubation.
Mitochondrial superoxide production assay
Mitochondrial superoxide production was measured using the MitoSOX Red Superoxide Indicator (ThermoFisher Scientific, #M36008). The MitoSOX™ Red Mitochondrial Superoxide Indicator is a fluorogenic dye for the selective detection of superoxide in the mitochondria of living cells.
500′000 cells/well were seeded and grown in 6-well plates in DMEM low glucose medium supplemented with 2% fetal calf serum and 1% penicillin/streptomycin (Sigma-Aldrich). After treatment with VAE for 24 h, cells were trypsinized, transferred to 5 mL Facstubes, washed, and suspended in 0.5 mL prewarmed assay buffer (HBSS with calcium and magnesium, Sigma-Aldrich). Depending on the cell line, 30–60 µM MitoPQ, mitochondria-targeted redox cycler (Abcam plc, Cambridge, UK, Cat# ab146819) was used as a positive control. After the addition of 1 µM MitoSOX, cells were incubated for 30 min in a shaking water bath at 37 °C, protected from light. DMSO was used as a vehicle control. Cells were washed three times and resuspended in 0.5 mL assay buffer. After the addition of DAPI to exclude dead cells, samples were immediately analyzed using a FACSCanto II (BD Biosciences, San Jose, CA). Values are expressed as MFI (geometric mean).
Flow cytometry analysis
DAMP expression on the surface of non-permeabilized cells and apoptosis were determined by flow cytometry. After treatment with VAE or taxol or tunicamycin in 6-well cell culture plates for 24 or 48 h, cells were trypsinized, washed, and suspended in PBS containing 0.5% FCS. Cells were co-stained with either recombinant PE anti-calreticulin antibody [EPR3924]—ER marker (Abcam Cat# ab209577, RRID: AB_2943490) and Alexa Fluor® 488 anti-Hsp70 antibody (BioLegend Cat# 648004, RRID: AB_2119537) or with recombinant Alexa Fluor® 488 anti-Hsp90 antibody [EPR16621-67] (Abcam Cat# ab223467, RRID: AB_2943491) for 30 min on ice in the dark. After washing with Annexin V Binding Buffer (BD Biosciences), cells were incubated with APC Annexin V (#550475, BD Biosciences) or with either 7-AAD or DAPI (#559925/#564907, BD Biosciences) for 15 min at 4 °C in the dark to monitor apoptosis and cell death. Samples were immediately analyzed using a FACSCanto II. Data were analyzed using FlowJo 10.6.2 software (Ashland OR, USA). Early apoptotic cells: Annexin V positive and 7-AAD/DAPI negative. Late apoptotic and/or necrotic cells: Annexin V positive and 7-AAD/DAPI positive. Values are expressed as a percentage of total cell number. Cells were interpreted as pre-apoptotic if they belonged to the population of Annexin V- and 7-AAD/DAPI-negative cells whose mean fluorescence intensity (MFI, geometric mean) for calreticulin was significantly higher than that of untreated control cells.
HMGB1 assay
Cells were seeded, treated with VAE, taxol or mitoxantrone dihydrochloride (Sigma-Aldrich, #M6545) (positive control), and cell culture supernatants were collected at 24 and 48 h and frozen at −80 °C. HMGB1 levels in cell supernatants were quantified using a commercial ELISA kit (Elabscience Biotech, #E-EL-H1554) according to the manufacturer's instructions. The detection limit was 25.31 pg/mL.
ATP assay
Cells were seeded, treated with VAE or taxol, and cell culture supernatants were collected at 6 and 24 h and frozen at −80 °C. The extracellular ATP concentration in cell supernatants was analyzed using an ATP chemiluminescence assay kit (Molecular Probes, # A22066) according to the manufacturer's instructions.
Briefly, 10 µl each of standards or supernatants were added to 90 µl of reaction solution. After incubation for 15 min, luminescence was measured using a Wallac Victor II plate reader.
Data analysis
Data were analyzed by either one-way analysis of variance (ANOVA, uncorrected Fisher's LSD), Friedman test (uncorrected Dunn's test), or Spearman nonparametric correlation analysis using GraphPad Prism, version 9.4.0. The significance level was defined as p ≤ 0.05.
Results
ER stress signaling pathway
To investigate whether the aqueous, fermented mistletoe preparation (Viscum album extract, VAE) induces ribotoxic stress, we analyzed the ER stress regulators p-eIF2α, ATF4, and CHOP by Western blot. Tunicamycin (TM), a compound routinely used to activate an ER stress response including the ER stress markers mentioned above, was used as a positive control [27, 28]. Both VAE and TM significantly increased eIF2α phosphorylation in all cell lines tested. In addition, the ER stress inducer TM significantly stimulated the expression of ATF4 and CHOP in all cell lines, whereas VAE had no stimulating effect on the expression of ATF4 and CHOP under the experimental conditions used, except for a significant increase of CHOP in the triple negative MDA-MB-231 cells. Treatment with VAE in SKBR3 cells resulted in a significant (p < 0.01) reduction of ATF4 (Fig. 1).
The expression levels of p-eIF2α, ATF4 and CHOP after treatment with indicated reagents for 24 h were determined by stain free Western blot analysis. Normalized data are presented as mean ± SE of at least 3 independent experiments with SKBR3, MDA-MB-231, MCF-7 and B16F10 cells. Representative western blot images are shown for all cell lines. The arrow indicates the location of ATF4 protein bands. Significance values are given relative to untreated controls (*p < 0.05, **p < 0.01, ***p < 0.001)
As we will also show for the expression of DAMPs, we observed large sensitivity differences between the cell lines regarding the VAE concentrations necessary to induce phosphorylation of the ER stress regulator eIF2α. In SKBR3, a VAE concentration of 20–30 µg/mL was sufficient, whereas 200–400 µg/mL VAE were required for the induction of p-eIF2α in MCF-7, MDA-MB-231 and B16F10 cells (Fig. 1). Full uncropped blot images are shown in Supplementary Material 2.
Mitochondrial superoxide production
We detected mitochondrial superoxide generation using the MitoSOX red (MSR) superoxide indicator, which rapidly and selectively targets the mitochondria. Positive control MitoPQ is a redox cycler that selectively increases mitochondrial superoxide production [29]. In all cell lines, we detected a concentration-dependent, significantly increased mitochondrial superoxide production after 24 h of treatment with either VAE or the positive control MitoPQ. The human SKBR3 cell line was more sensitive to VAE than the other cell lines, and a significant increase in mitochondrial superoxide production was induced by VAE concentrations of 5 and 10 µg/mL, whereas in the other two breast cancer cell lines, 400 µg/mL and in murine B16F10 cells, 600–800 µg/mL VAE was used to achieve a significant effect. The results are shown in Fig. 2.
Analysis of mitochondrial superoxide production. SKBR3, MDA-MB-231, MCF-7 and B16 F10 cells were treated with either MitoPQ (positive control) or VAE for 24 h, labelled with MitoSOX red Superoxide Indicator and analyzed by flow cytometry. A Representative FACS images of MitoSOX red fluorescence intensity and B Box and whiskers plots represent the MFI data (Min to Max) from at least 3 independent experiments. Significance values are given relative to untreated controls (*p < 0.05, **p < 0.01, ***p < 0.001)
DAMP exposure, apoptosis, and cell death
We determined the exposure of the DAMPs calreticulin (CRT), heat shock protein 70 (hsp70), and hsp90 on the cell surface of DAPI/7-AAD negative cells by flow cytometry. The ICD-inducing chemotherapeutic taxol (paclitaxel) [30] and TM were used as positive controls. Taxol has been shown to induce the release of DAMPs such as calreticulin, ATP and HMGB1 in ovarian cancer cells [31].
Calreticulin exposure on the surface of pre-apoptotic (Annexin-V-negative and 7-AAD/DAPI-negative) cells was highly cell line-dependent and varied significantly (Fig. 3). We observed a significant increase in CRT on the surface of pre-apoptotic SKBR3 cells after treatment with both TM and VAE for 24 h and 48 h. In B16F10 cells, CRT increased significantly after treatment with VAE (24 h and 48 h). TM treatment had no effect. Considering that MDA-MB-231 and MCF-7 cells are much more resistant to treatment with VAE and that in pre-experiments no DAMP exposure was detectable at all after 24 h, we examined these cell lines only after 48 h treatment. In MDA-MB-231 cells, only treatment with TM resulted in significantly increased DAMP exposure, and in MCF-7 cells, none of the treatments induced DAMP exposure on the surface of pre-apoptotic cells.
Exposure of calreticulin on the surface of pre-apoptotic (annexin-V-negative and 7-AAD/DAPI-negative) cells. A Gating strategy of pre-apoptotic cells and representative FACS histogram plot of CRT exposure on untreated control vs. isotype control. Representative FACS histogram plots of CRT-exposure on pre-apoptotic cells after treatment with either B VAE or C TM vs. untreated control. D FACS analysis of the mean fluorescence intensity (MFI, geomean) of CRT exposure on the surface of cells treated with TM or VAE, respectively. Results are presented as mean ± SE from at least 3 independent experiments. Significance values are given relative to untreated controls (*p < 0.05, **p < 0.01)
Because the size of taxol-treated cells increased significantly due to swelling, resulting in a fluorescence shift of the entire cell population, the corresponding MFI data were not included in the analysis.
We also determined the amount of DAMPs on the surface of 7-AAD/DAPI negative cells in relation to the phosphatidylserine expression (Annexin V positive, early apoptotic cells) (Supplementary Fig. 1, Supplementary Fig. 2). Our results indicate a strong variability in the sensitivity of the individual cell lines to VAE, TM, and taxol. All cell lines exposed the investigated DAMPs on the surface of early apoptotic cells after treatment with either VAE or tunicamycin (Figs. 4, 5 and 6).
Cell surface exposure of calreticulin (CRT), hsp70, hsp90 or AV +/DAPI- (early apoptotic) cells after treatment of SKBR3 cells for A. 24 h or B. 48 h with either tunicamycin (TM), taxol (Tax) or viscum album extract (VAE). Results are presented as mean ± SE from at least 3 independent experiments. Significance values are given relative to untreated controls (*p < 0.05, **p < 0.01, ***p < 0.001)
Cell surface exposure of calreticulin (CRT), hsp70, hsp90 or AV +/DAPI- (early apoptotic) cells after treatment of A. MDA-MB-231 or B. MCF-7 cells for 48 h with either tunicamycin (TM), taxol (Tax) or viscum album extract (VAE). Results are presented as mean ± SE from at least 3 independent experiments. Significance values are given relative to untreated controls (*p < 0.05, **p < 0.01, ***p < 0.001)
Cell surface exposure of calreticulin (CRT), hsp70, hsp90 or AV +/DAPI- (early apoptotic) cells after treatment of B16F10 cells for A. 24 h or B. 48 h with either tunicamycin (TM), taxol (Tax) or viscum album extract (VAE). Results are presented as mean ± SE from at least 3 independent experiments. Significance values are given relative to untreated controls (*p < 0.05, **p < 0.01)
In SKBR3 cells, CRT and hsp70 were significantly increased after 24 h treatment with TM and taxol, and hsp90 was significantly increased by taxol. After 48 h treatment with TM and taxol or with 2.5 µg/mL VAE, all DAMPs were significantly increased (Fig. 4). The induction of a significant increase of DAMPs on the cell surface of MDA-MB-231 and MCF 7 cells by taxol and TM was different. VAE induced a significant increase in CRT, hsp70 and hsp90 in both MDA-MB-231 and MCF-7 cells after 48 h of treatment (Fig. 5). In MDA-MB-231 and MCF-7 cell lines, the number of cells with DAMPs translocated to the cell surface and the percentage of early apoptotic cells were considerably lower than in SKBR3 and B16F10, even after 48 h of VAE treatment. A concentration of 500 µg/mL VAE induced the translocation of DAMPs in 7–8% of treated 7-AAD/DAPI negative MDA-MB-231 cells (depending on the DAMP) and the percentage of early apoptotic cells was about 18%; in MCF-7, between 4 and 7% of cells were DAMP-positive and about 11% were early apoptotic. In contrast, 18–51% of viable SKBR3 cells treated with 5 µg/mL were DAMP-positive and 22% were early apoptotic. The B16F10 cell line had between 22 and 56% DAMP-positive cells and 52% early apoptotic cells after 48 h of VAE treatment at 200 µg/mL (Fig. 7). In addition, up to 50-fold higher concentrations of VAE were required to induce the appearance of DAMPs at the cell surface. This suggests that MCF-7 and MDA-MB-231 cells are more resistant to VAE. Nevertheless, we measured slightly but still significantly increased levels of DAMPs in the two cell lines after treatment with TM and VAE, and partially with taxol.
In B16F10 cells after 24 h, only 2.5 µM taxol significantly increased hsp70 and 100 µg/mL VAE significantly increased hsp90 levels (Fig. 6A), but 48 h treatment with taxol significantly increased the amount of CRT and hsp90 and treatment with TM significantly increased the exposure of hsp70 on the surface of early apoptotic cells. 100 µg/mL VAE significantly increased the exposure of all DAMPs at 48 h. (Fig. 6B). The 24 h and 48 h values for the more sensitive SKBR3 and B16F10 cells reflect the kinetics of cell surface DAMP exposure (Figs. 4 and 6).
In SKBR3, B16F10 and MDA-MB-231, the amount of cell surface exposed CRT, hsp70 and hsp90, respectively, correlated significantly (p < 0.05 to p < 0.01, Spearman correlation) with the amount of early apoptosis at both 24 h and 48 h (see Supplementary Table 1 and Supplementary Fig. 3). Only the level of DAMPs on MCF-7 cell surface showed no significant correlation with early apoptosis. (Supplementary Table 1). This cell line showed a concentration-independent characteristically low level (< 10%) of early apoptosis and DAMP exposure, despite a high death rate after VAE, TM, or taxol treatment, respectively. Supplementary Fig. 4 shows representative dot blots of flow cytometric analysis of apoptosis and Fig. 8 shows apoptosis/viability data for all cell lines and treatments. Significance levels shown refer to the sum of all early apoptotic and late apoptotic/necrotic cells.
Cytotoxicity of A SKBR3 cells treated for 24 h, B SKBR3 cells treated for 48 h, C MDA-MB-231 cells treated for 48 h, D MCF-7 cells treated for 48 h, E B16F10 cells treated for 24 h and F B16F10 cells treated for 48 h with indicated concentrations of TM, taxol or VAE, respectively and untreated controls. Results are presented as mean percentage ± SE from at least 3 independent experiments. Significance values are given relative to untreated controls (*p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001). The indicated significance levels refer to the sum of early apoptotic and late apoptotic/necrotic cells
Cellular HMGB1 and ATP release
Under our experimental conditions, no HMGB1 release was detected in the cell culture supernatants of SKBR3, MCF-7 and B16F10 cells (data not shown). Only in MDA-MB-231 cells treatment with the positive control mitoxantrone resulted in a significant increase in extracellular HMGB1 levels. It should be noted that this cell line was the only one that actively released HMGB1 into the supernatant even in the untreated control (see Supplementary Fig. 5).
We measured the release of ATP in the cell culture supernatant after 6 and 24 h of treatment. As shown in Fig. 9, VAE induced a cell line, time- and concentration-dependent release of ATP from the tumor cells. In SKBR3 and MDA-MB-231 cells, no ATP release was observed after 6 h. In MCF-7 cells, ATP levels in the medium increased significantly compared to the untreated control after treatment with 400 and 800 µg/mL VAE for 6 h. In all three breast cancer cell lines, there was a significant up to threefold increase in ATP release after 24 h of VAE treatment. The effect of VAE treatment was most pronounced in the murine melanoma cell line B16F10. Extracellular ATP levels were significantly increased by up to sevenfold after 6 h of treatment with 400 and 800 µg/mL VAE and after 24 h of treatment with all VAE concentrations compared to the untreated control. A slightly increased significant ATP release after taxol treatment was observed only after 24 h of treatment of SKBR3 cells.
ATP release by A SKBR3, B MDA-MB-231, C MCF-7 and D B16F10 cells treated with indicated concentrations of taxol or VAE for 6 h and 24 h, respectively. Results are presented as the average change in extracellular ATP concentration compared to the untreated control (mean ± SE from at least 3 independent experiments). Significance values are expressed relative to untreated controls at 6 h and 24 h, respectively (*p < 0.05, **p < 0.01)
Discussion
For an effective polyclonal adaptive immune response mediated by T and/or B cells against pre-existing tumor-specific or tumor-associated antigens, a sequence of events is essential, which Chen and Mellman have described as the cancer immunity cycle [11]. The first phase necessary to induce a specific antitumor immune response can be initiated by therapies that induce immunogenic cell death. Zhou et al. have reviewed several current and emerging therapeutic approaches to achieve this effect [32].
The aim of our project was to investigate whether treatment of tumor cell lines with an aqueous fermented mistletoe extract leads to cell death with the characteristics of immunogenic cell death. The positive controls were used for verification of the experimental procedures and will not be discussed further.
The induction of immunogenic cell death is mostly initiated by endoplasmic reticulum stress. Phosphorylation of eIF2α (eukaryotic initiation factor 2α) by the kinase PERK is a hallmark of ICD. Phosphorylation of eIF2α is a common stress response in eukaryotes. It leads to inactivation of global protein synthesis and subsequent signaling events such as activation of caspase-8, activation of the pro-apoptotic proteins BAX and BAK, and relocation of CRT to the cell surface [10]. Under our experimental conditions, we observed that treatment of cancer cell lines with VAE resulted in phosphorylation of EIF2α, but not in increased expression of ATF4 or CHOP. This result indicates an incomplete ER stress response, as previously shown by Bezu et al. for various chemotherapeutic and other agents [28]. Further specific stress inducers have been described, such as UV irradiation, which, despite robust phosphorylation of eIF2a, did not lead to an increase in ATF4 expression, but to its transcriptional repression. Tissue-specific regulation of ATF4 under certain stress conditions may also occur [33].
Little information is available on the induction of ER stress by mistletoe extracts. Park et al. showed that treatment of K562 cells with mistletoe extracts resulted in upregulation of GRP78/BiP expression, increased phosphorylation of eIF2α, reduction of p90 ATF6, and time-dependent induction of CHOP mRNA, indicating the induction of ER stress [34]. Maltseva et al. treated Molt4 and CaCo2 cells with mistletoe lectin I (MLI) and analyzed changes in the expression of genes involved in ER stress. Expression profiles were dependent on the cell type, MLI concentration and exposure time. They found increased levels of spliced XBP1 gene mRNA (XBP1 s), chaperone protein gene mRNA (HSPH1, HSPB1, and HSPA5), and transcription factor ATF6 gene mRNA, probably due to activation of IRE1a and ATF6-dependent signaling pathways. Changes in ATF4 and CHOP expression were dependent on MLI concentration [35].
We examined the production of superoxide in mitochondria by flow cytometry using the MitoSOX red assay. As a positive control, we used mito-paraquat (MitoPQ), a mitochondria-targeted agent that acts on mitochondrial complex I and leads to the production of superoxide [36]. ROS play a complex role in the regulation of pathways such as apoptosis, autophagy and necroptosis that are triggered by mitochondrial and ER stress [37, 38]. Both endogenous and exogenous stressors can induce ROS in a cell [39]. The main source of superoxide is leakage from the mitochondrial electron transport chain [40]. Damage to mitochondrial components by ROS can lead to loss of mitochondrial membrane potential, release of cytochrome c, further increase in cellular ROS, and apoptosis. The increased production of ROS is closely related to ER stress. Mitochondria-associated ER stress activates the specific danger signaling required to produce immunogenic DAMPs, and its induction is a powerful strategy for ICD enhancement and cancer immunotherapy [9, 41]. We measured the production of ROS to determine whether ROS may be involved in the VAE-induced processes leading to the expression of surrogate markers of ICD. We showed that VAE treatment significantly increased mitochondrial superoxide production in a concentration-dependent manner in all cell lines tested (Fig. 2). Korean mistletoe lectins induced the production of reactive oxygen species and the loss of mitochondrial membrane potential in hepatocarcinoma and macrophage-like cells [42, 43]. Mistletoe lectins (MLs) are mainly responsible for the cytotoxic and pro-apoptotic effects of VAE [44]. MLs belong to the ribosome-inactivating proteins class II (RIP-II), highly potent cytotoxins that interfere with protein biosynthesis and can induce different cell death mechanisms with different timing and relative potency in different cell lines. MLs are known to induce apoptosis via the intrinsic pathway. Mitochondrial damage leads to loss of mitochondrial membrane potential and activation of caspase-9. The downstream activated caspase-3 is an executor of DNA fragmentation, poly (ADP-ribose) polymerase (PARP) cleavage, and cell death by apoptosis. Bantel et al. showed that MLs can also activate caspase-8 in a death receptor-independent pathway, which may indicate an interplay between different death mechanisms [45, 46].
The major functions of the ER include calcium sequestration and glycoprotein processing. Chaperones are proteins within the ER that help fold newly synthesized proteins. They are involved in protein repair after cells are exposed to physical or chemical stress, during which they are rapidly increased. As a result of a stress response, chaperones may be translatorily upregulated, relocated to the cell surface, or released into the extracellular space. There, they promote the engulfment of dying cells ("eat-me"signals) and serve as a danger signal for the immune system, which can subsequently be activated [5, 47].
We measured the cell surface exposure of the DAMPs calreticulin and the heat shock proteins hsp70 and hsp90 after treatment with VAE or with tunicamycin or taxol. Calreticulin is one of the best-known DAMPs that is a major determinant in the process of immunogenic cell death. Calreticulin is expressed on the surface of most living cells and is involved in the removal of apoptotic cells (efferocytosis) by interacting with the internalization receptor LRP (LDL receptor-related protein, CD91) on the surface of phagocytes [48, 49]. Obeid et al. (2007) described the increased exposure of calreticulin on the cell surface of pre-apoptotic cells as a very early event of immunogenic cell death. Using mass spectrometry analysis, they demonstrated calreticulin exposure on the surface of CT26 mouse colon cancer cells treated with ICD inducers. This active translocation occurred before the first signs of apoptosis [50]. Panaretakis et al. showed that exocytosis of the calreticulin/ERp57 complex is accompanied by sequential activation of an ER stress response and activation of caspases prior to cell apoptosis [10].
In two of the four cell lines studied (SKBR3 and B16F10), we detected increased CRT exposure on the surface of pre-apoptotic cells (Fig. 3). This exposure of CRT prior to the appearance of phosphatidylserine is an important indication that VAE is capable of triggering ICD. As mentioned above, several factors are involved in the triggering of an ICD. Whether the biology of the individual cell lines, the experimental conditions, or other factors are responsible for the different results in the cell lines we studied must be clarified in future studies.
We observed a significant increase of the three DAMPs CRT, hsp70 and hsp90 on the surface of early apoptotic cells in 3 breast cancer and one murine melanoma cell line. For VAE, this effect was time-, concentration- and cell line-dependent. The Her-2-positive breast cancer cell line SKBR3 was the most sensitive, followed by the melanoma cell line B16F10. Significantly more resistant were the estrogen receptor positive cell line MCF-7 and the triple negative cell line MDA-MB-231, in which, despite statistical significance, there was only a small increase in the exposure of DAMPs on the early apoptotic cell surface.
Breast tumors are highly heterogeneous and can be classified based on several characteristics, including histological type, hormone receptor status, morphology and molecular profile. The cell lines we used can be classified into the following subtypes based on their molecular characteristics:
-
SKBR3 (adenocarcinoma) – luminal Her2 +, claudin-low
-
MCF-7 (invasive ductal carcinoma) – luminal A, estrogen receptor +, progesterone receptor +
-
MDA-MB-231 (adenocarcinoma) – basal B, triple negative breast cancer, mesenchymal stem-like [51, 52].
VAE and particularly MLs do not appear to target specific tumor cell types, they bind to certain carbohydrate epitopes via their B-chain. Specifically, they bind to sialic acids, which are the part of glycoproteins present on the cell surface [53, 54]. During cancer progression and metastasis, the glycosylation of cancer cells changes. Sialic acids can be overexpressed compared to normal cells [55]. These changes in sialic acid expression may contribute to variations in the sensitivity of cell lines to VAE. It is known that MLs can influence the expression of genes involved in tumor cell survival, growth and metastasis. Treatment with mistletoe extracts has been shown to elicit specific gene expression profiles, such as immune or stress response genes, which in turn may preferentially trigger certain pathways [56]. Eggenschwiler et al. hypothesized that the higher the number of affected genes, the more sensitive a cell line is to the cytotoxic effect of VAE [57, 58]. Beztsinna et al. analyzed the cell binding, endocytosis, subcellular processing of MLs, and the activation of apoptosis [59]. It is important to note that these processes can vary depending on the specific cell line. Future studies must clarify resistance causes to mistletoe preparations.
The release of HMGB1 into the extracellular space by dying cells is considered one of the hallmarks of immunogenic cell death. HMGB1 can exert an immunostimulatory function via specific receptors such as RAGE and TLR4 [60, 61]. In our study, this molecular signal was not measurable by in vitro treatment of cancer cells with VAE. It is generally believed that HMGB1 is released only during necrotic cell death and not by apoptosis, because under physiological conditions, nuclear components like HMGB1 are retained during apoptosis until they are cleared by phagocytes [62]. Since the lectin-containing VAEs induce apoptosis, this may explain why HMGB1 was not detectable in our cell culture supernatants. Bell et al. though have shown that HMGB1 can also be released during late apoptosis [63]. If this was the case in our experiments, the HMGB1 levels were below the detection limit of the ELISA kit. The pro-inflammatory properties of mistletoe lectins themselves may at least partially compensate for the absence of HMGB1. Kim et al. showed that in vitro treatment of bone marrow-derived dendritic cells with the B chain of KML resulted in their maturation and the expression of co-stimulatory molecules and pro-inflammatory cytokines. These effects were found to be TLR4-dependent. The authors demonstrated the stimulatory effect of KML-B matured DCs on activation and proliferation of specific cytotoxic T cells and their INF-γ secretion. In vivo, they demonstrated the reduction of the tumor size in KML-B treated vaccinated mice [64, 65].
Another key feature of ICD is the release of ATP by dying cells which acts as both a "find me" signal and a potential inflammatory stimulus for DCs [66, 67]. After treatment with VAE, we measured significantly elevated ATP levels in the cell culture supernatants of all cell lines tested whereby the effect of VAE on the murine melanoma cell line was considerably stronger than on the breast cancer cell lines. Martins et al. showed that ATP is secreted via active exocytosis and in an autophagy- and caspase-dependent manner [66]. In the scientific literature we have not found any reports on the effect of mistletoe extracts on autophagy or on the release of ATP by treated cells. For other plant lectins, however, autophagy inducing effects have been demonstrated [68, 69]. The processes of autophagy and apoptosis are closely related [70]. Each can promote or inhibit the other. If the level of autophagy becomes too high, apoptosis will occur. This means that although the commonly known cell death induced by MLs is via the intrinsic apoptotic pathway, autophagy-dependent ATP release is possible at an early stage of VAE treatment. The precise impact of VAE on autophagy remains to be demonstrated in further studies.
The ICD-associated DAMPs promote the recruitment as well as the maturation and activation of DCs. Similar effects have been described in studies of the direct effect of European and Korean mistletoe extracts on DCs in vitro. Treatment of monocyte-derived immature DCs with VAE in vitro resulted in increased expression of HLA-DR, CD40, CD80, CD83, and CD86 and induction of the inflammatory cytokines IL-6, IL-8, IL-12, and TNF-a. In an allogeneic mixed lymphocyte reaction, specific CD8 T cells were activated and IFN-γ secretion was increased [71,72,73].
Furthermore, MLs are immunomodulators that induce the production of cytokines and other mediators. In breast cancer patients, serum IL-12 levels increased after mistletoe therapy, and peripheral blood mononuclear cells showed increased production of IFN-γ and IL-2 [74]. In another study, increased levels of IL-6 and IFN-γ were measured in the serum of mistletoe-treated breast cancer patients [75]. In B16 F10 implanted mice, VAE caused inhibition of tumor growth and upregulation of splenocyte proliferation and IL-12 secretion [76].
Our results, together with the above-mentioned pro-inflammatory properties of VAE led us to hypothesize that local mistletoe treatment could induce an ICD and lead to the stimulation of an antitumor immune response. This hypothesis is supported by the fact that intralesional mistletoe treatment has shown positive effects in clinical use. The (off-label) peri- or intratumoral treatment with mistletoe extracts has already been used in individual cases with promising results. Clinical case reports illustrate the effect of intratumoral mistletoe treatment on patients with cutaneous squamous cell carcinoma, adenoid cystic carcinoma, malignant pleural mesothelioma, diffuse large B-cell lymphoma, cervical carcinoma and carcinoma in situ, skull metastases, and colon carcinoma [77,78,79,80,81,82,83]. Intratumoral mistletoe treatment, sometimes in combination with systemic chemotherapy, has also been used in inoperable pancreatic cancer. No serious side effects were observed, and the treatment was considered feasible and safe [84]. Another clinical case report describes the complete remission of a recurrent duodenal carcinoma after intratumoral mistletoe therapy [85]. The effects and side effects of intraperitoneal, intrapleural, and intratumoral mistletoe therapy were studied and summarized in a multicenter observational study and prospective trials were recommended [86, 87]. To date, there have been no major studies on the use of intratumoral mistletoe therapy. Many studies including meta-analyses for the clinical use of mistletoe therapy can be found on the Mistletoe-therapy online database [88].
Our investigation showed that in vitro treatment of various breast cancer and melanoma cell lines elicited several pathways and features typical of immunogenic cell death.
However, it is important to consider further aspects contributing to a successful induction of an anti-tumor immune response. In this regard, the biology of individual tumors appears to be important and could have a significant impact on a successful ICD induction.
Conclusions
Our study showed that in vitro treatment of various breast cancer and melanoma cell lines induced several stress-related molecular processes characteristic of immunogenic cell death: The induction of p-eIF2α indicated the triggering of an ER stress response and we proved increased mitochondrial ROS production, calreticulin exposure on the surface of pre-apoptotic cells and cell surface expression or release of the DAMPs calreticulin, hsp70, hsp90 and ATP.
Our results encourage further studies to evaluate the effect of mistletoe-induced cancer cell death on the activation of an adaptive anti-tumor immune response.
Data availability
All data generated or analyzed during this study are included in this published article and its supplementary information files. The datasets generated for this study are available on reasonable request from the corresponding author.
Abbreviations
- ICD:
-
Immunogenic cell death
- DAMPs:
-
Damage-associated molecular patterns
- eIF2α/p-eIF2 α:
-
(Phospho)-eukaryotic translation initiating factor 2A
- ATF4:
-
Activating transcription factor 4
- CHOP:
-
C/EBP-homologous protein
- Hsp:
-
Heat shock protein
- ROS:
-
Reactive oxygen species
- CRT:
-
Calreticulin
- VAE:
-
Viscum album extract
- ER:
-
Endoplasmic reticulum
- HMGB1:
-
High-mobility group protein B1
- ATP:
-
Adenosine triphosphate
- IRE1α:
-
Inositol-requiring enzyme 1α
- PERK:
-
Protein kinase R-like endoplasmic reticulum kinase
- ATF6:
-
Activating transcription factor 6
- UPR:
-
Unfolded protein response
- DCs:
-
Dendritic cells
- ML:
-
Mistletoe lectin
- MSR:
-
MitoSOX red
- BAX:
-
Bcl-2–associated X protein
- BAK:
-
Bcl-2 homologous antagonist
- XBP1:
-
X-box binding protein 1
- RIP-II:
-
Ribosome-inactivating protein class II
- PARP:
-
Poly (ADP-ribose) polymerase
- LRP:
-
LDL receptor-related protein
- RAGE:
-
Receptor for advanced glycation endproducts
- TLR4:
-
Toll-like receptor 4
- KML:
-
Korean mistletoe lectin
References
Tang S, Ning Q, Yang L, Mo Z, Tang S. Mechanisms of immune escape in the cancer immune cycle. Int Immunopharmacol. 2020;86:106700.
Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.
Garg AD, Dudek-Peric AM, Romano E, Agostinis P. Immunogenic cell death. Int J Dev Biol. 2015;59(1-2–3):131–40.
Green DR, Ferguson T, Zitvogel L, Kroemer G. Immunogenic and tolerogenic cell death. Nat Rev Immunol. 2009;9(5):353–63.
Wiersma VR, Michalak M, Abdullah TM, Bremer E, Eggleton P. Mechanisms of translocation of ER chaperones to the cell surface and immunomodulatory roles in cancer and autoimmunity. Front Oncol. 2015;5:7.
Clarke R, Cook KL, Hu R, Facey CO, Tavassoly I, Schwartz JL, Baumann WT, Tyson JJ, Xuan J, Wang Y. Endoplasmic reticulum stress, the unfolded protein response, autophagy, and the integrated regulation of breast cancer cell fate. Can Res. 2012;72(6):1321–31.
Harding HP, Zhang Y, Ron D. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature. 1999;397(6716):271–4.
Urra H, Dufey E, Lisbona F, Rojas-Rivera D, Hetz C. When ER stress reaches a dead end. Biochim Biophys Acta. 2013;1833(12):3507–17.
Garg AD, Krysko DV, Vandenabeele P, Agostinis P. The emergence of phox-ER stress induced immunogenic apoptosis. Oncoimmunology. 2012;1(5):786–8.
Panaretakis T, Kepp O, Brockmeier U, Tesniere A, Bjorklund AC, Chapman DC, Durchschlag M, Joza N, Pierron G, Van Endert P. Mechanisms of pre-apoptotic calreticulin exposure in immunogenic cell death. EMBO J. 2009;28(5):578–90.
Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39(1):1–10.
Nelson BE, Adashek JJ, Lin SH, Subbiah V. The abscopal effect in patients with cancer receiving immunotherapy. Med. 2023;4(4):233–44.
Kroemer G, Galassi C, Zitvogel L, Galluzzi L. Immunogenic cell stress and death. Nat Immunol. 2022;23(4):487–500.
Whiteside TL, Demaria S, Rodriguez-Ruiz ME, Zarour HM, Melero I: Emerging opportunities and challenges in cancer immunotherapy. Clin Cancer Res. 2016;22(8):1845–55.
Champiat S, Tselikas L, Farhane S, Raoult T, Texier M, Lanoy E, Massard C, Robert C, Ammari S, De Baere T. Intratumoral immunotherapy: from trial design to clinical practice. Clin Cancer Res. 2021;27(3):665–79.
Marabelle A, Andtbacka R, Harrington K, Melero I, Leidner R, de Baere T, Robert C, Ascierto PA, Baurain JF, Imperiale M, et al. Starting the fight in the tumor: expert recommendations for the development of human intratumoral immunotherapy (HIT-IT). Ann Oncol. 2018;29(11):2163–74.
Urech K, Baumgartner S. Chemical constituents of Viscum album L.: implications for the pharmaceutical preparation of mistletoe. Zaenker, KS, Kaveri, SV, (eds): Mistletoe: From Mythology to Evidence-Based Medicine. Transl Res in Biomed. Basel, Karger. 2015;4:11–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1159/000375422.
Endo Y, Tsurugi K, Franz H. The site of action of the A- chain of mistletoe lectin I on eukaryotic ribosomes. FEBS Lett. 1988;231(2):378–80.
Dietrich JB, RibéreauGayon G, Jung ML, Franz H, Beck JP, Anton R. Identity of the N-terminal sequences of the three A chains of mistletoe (V.ALBUM L.) lectins: homology with ricin-like plant toxins and single-chain ribosome-inhibiting proteins. Anticancer Drugs. 1992;3:507–11.
Jager T, Holandino C, Melo MNO, Penaloza EMC, Oliveira AP, Garrett R, Glauser G, Grazi M, Ramm H, Urech K, et al. Metabolomics by UHPLC-Q-TOF Reveals Host Tree-Dependent Phytochemical Variation in Viscum album L. Plants (Basel). 2021;10(8):1726.
Vanhaverbeke C, Touboul D, Elie N, Prevost M, Meunier C, Michelland S, Cunin V, Ma L, Vermijlen D, Delporte C, et al. Untargeted metabolomics approach to discriminate mistletoe commercial products. Sci Rep. 2021;11(1):14205.
Urech K, Schaller G, Jaggy C. Viscotoxins, mistletoe lectins and their isoforms in mistletoe (Viscum album L.) extracts Iscador. Arzneimittelforschung. 2006;56(6A):428–34.
Klein R. Immunologie von Mistelextrakten. InDie Mistel in der Tumortherapie 4 - Aktueller Stand der Forschung und klinische Anwendung. edn. Edited by Scheer R, Alban S, Becker H, Beer AM, Blaschek W, Kreis W, Matthes H, Schilcher H, Spahn G, Stange R. Essen: KVC Verlag; 2016. pp.113–125.
Oei SL, Thronicke A, Schad F. Mistletoe and immunomodulation: insights and implications for anticancer therapies. Evid Based Complement Alternat Med. 2019;2019:5893017.
Weissenstein U, Kunz M, Urech K, Regueiro U, Baumgartner S. Interaction of a standardized mistletoe (Viscum album) preparation with antitumor effects of Trastuzumab in vitro. BMC Complement Altern Med. 2016;16:271.
Anthroposophic Pharmaceutical Codex (APC) Method 7.2.3 and 7.2.4 Compositions of fermented aqueous extracts and liquid preparations thereof (p.53). In., 5.1 edn; 2024. https://iaap-pharma.org/apc/.
Bassik MC, Kampmann M. Knocking out the door to tunicamycin entry. Proc Natl Acad Sci U S A. 2011;108(29):11731–2.
Bezu L, Sauvat A, Humeau J, Gomes-da-Silva LC, Iribarren K, Forveille S, Garcia P, Zhao L, Liu P, Zitvogel L. eIF2α phosphorylation is pathognomonic for immunogenic cell death. Cell Death Differ. 2018;25(8):1375.
Robb EL, Gawel JM, Aksentijevic D, Cocheme HM, Stewart TS, Shchepinova MM, Qiang H, Prime TA, Bright TP, James AM, et al. Selective superoxide generation within mitochondria by the targeted redox cycler MitoParaquat. Free Radic Biol Med. 2015;89:883–94.
Zhai J, Gu X, Liu Y, Hu Y, Jiang Y, Zhang Z. Chemotherapeutic and targeted drugs-induced immunogenic cell death in cancer models and antitumor therapy: an update review. Front Pharmacol. 2023;14:1152934.
Lau TS, Chan LKY, Man GCW, Wong CH, Lee JHS, Yim SF, Cheung TH, McNeish IA, Kwong J. Paclitaxel induces immunogenic cell death in ovarian cancer via TLR4/IKK2/SNARE-dependent exocytosis. Cancer Immunol Res. 2020;8(8):1099–111.
Zhou J, Wang G, Chen Y, Wang H, Hua Y, Cai Z. Immunogenic cell death in cancer therapy: present and emerging inducers. J Cell Mol Med. 2019;23(8):4854–65.
Dey S, Savant S, Teske BF, Hatzoglou M, Calkhoven CF, Wek RC. Transcriptional repression of ATF4 gene by CCAAT/enhancer-binding protein beta (C/EBPbeta) differentially regulates integrated stress response. J Biol Chem. 2012;287(26):21936–49.
Park Y-K, Do YR, Jang B-C. Apoptosis of K562 leukemia cells by Abnobaviscum F®, a European mistletoe extract. Oncol Rep. 2012;28(6):2227–32.
Maltseva DV, Krainova NA, Khaustova NA, Nikulin SV, Tonevitskaya SA, Poloznikov AA. Biodistribution of viscumin after subcutaneous injection to mice and in vitro modeling of endoplasmic reticulum stress. Bull Exp Biol Med. 2017;163(4):451–5.
Chowdhury AR, Zielonka J, Kalyanaraman B, Hartley RC, Murphy MP, Avadhani NG. Mitochondria-targeted paraquat and metformin mediate ROS production to induce multiple pathways of retrograde signaling: a dose-dependent phenomenon. Redox Biol. 2020;36:101606.
Redza-Dutordoir M, Averill-Bates DA. Activation of apoptosis signalling pathways by reactive oxygen species. Biochem Biophys Acta. 2016;1863(12):2977–92.
Villalpando-Rodriguez GE, Gibson SB. Reactive Oxygen Species (ROS) Regulates different types of cell death by acting as a rheostat. Oxid Med Cell Longev. 2021;2021:9912436.
Rahal A, Kumar A, Singh V, Yadav B, Tiwari R, Chakraborty S, Dhama K. Oxidative stress, prooxidants, and antioxidants: the interplay. Biomed Res Int. 2014;2014:761264.
Dickinson BC, Chang CJ. Chemistry and biology of reactive oxygen species in signaling or stress responses. Nat Chem Biol. 2011;7(8):504–11.
Garg AD, Nowis D, Golab J, Agostinis P. Photodynamic therapy: illuminating the road from cell death towards anti-tumour immunity. Apoptosis. 2010;15:1050–71.
Kim W-H, Park WB, Gao B, Jung MH. Critical role of reactive oxygen species and mitochondrial membrane potential in Korean mistletoe lectin-induced apoptosis in human hepatocarcinoma cells. Mol Pharmacol. 2004;66(6):1383–96.
Byeon SE, Lee J, Yu T, Kwon M, Hong S, Cho JY. Extracellular signal-regulated kinase is a major enzyme in Korean mistletoe lectin-mediated regulation of macrophage functions. Biomolecules und Therapeutics. 2009;17(3):293–8.
Büssing A. Induction of apoptosis by the mistletoe lectins: a review on the mechanisms of cytotoxicity mediated by VISCUM ALBUM L. Apoptosis. 1996;1(1):25–32.
Bantel H, Engels IH, Voelter W, Schulze-Osthoff K, Wesselborg S. Mistletoe lectin activates caspase-8/FLICE independently of death receptor signaling and enhances anticancer drug-induced apoptosis. Cancer Res. 1999;59(9):2083–90.
Orning P, Lien E. Multiple roles of caspase-8 in cell death, inflammation, and innate immunity. J Leucocyte Biol. 2021;109(1):121–41.
Jethmalani SM, Henle KJ, Gazitt Y, Walker PD, Wang SY. Intracellular distribution of heat-induced stress glycoproteins. J Cell Biochem. 1997;66(1):98–111.
White TK, Zhu Q, Tanzer ML. Cell surface calreticulin is a putative mannoside lectin which triggers mouse melanoma cell spreading (∗). J Biol Chem. 1995;270(27):15926–9.
Ogden CA, deCathelineau A, Hoffmann PR, Bratton D, Ghebrehiwet B, Fadok VA, Henson PM. C1q and mannose binding lectin engagement of cell surface calreticulin and CD91 initiates macropinocytosis and uptake of apoptotic cells. J Exp Med. 2001;194(6):781–96.
Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini J-L, Castedo M, Mignot G, Panaretakis T, Casares N. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13(1):54.
Holliday DL, Speirs V. Choosing the right cell line for breast cancer research. Breast Cancer Res. 2011;13:1–7.
Dai X, Cheng H, Bai Z, Li J. Breast cancer cell line classification and its relevance with breast tumor subtyping. J Cancer. 2017;8(16):3131.
Hoessli DC, Ahmad I. Mistletoe lectins: carbohydrate-specific apoptosis inducers and immunomodulators. Curr Organic Chem. 2008;12(11):918–25.
Muthing J, Meisen I, Bulau P, Langer M, Witthohn K, Lentzen H, Neumann U, Peter-Katalinic J. Mistletoe lectin I is a sialic acid-specific lectin with strict preference to gangliosides and glycoproteins with terminal Neu5Ac alpha 2–6Gal beta 1–4GlcNAc residues. Biochemistry. 2004;43(11):2996–3007.
Singh K, Agrawal L, Gupta R, Singh D, Kathpalia M, Kaur N. Lectins as a promising therapeutic agent for breast cancer: a review. Breast Dis. 2024;43(1):193–211.
Majeed M, Hakeem KR, Rehman RU. Mistletoe lectins: from interconnecting proteins to potential tumour inhibiting agents. Phytomedicine Plus. 2021;1(3):100039.
Eggenschwiler J, Patrignani A, Wagner U, Rehrauer H, Schlapbach R, Rist L, Ramos M, Viviani A. Gene expression profiles of different breast cancer cells compared with their responsiveness to fermented mistletoe (Viscum album L.) extracts Iscador from oak (Quercus), pine (Pinus), white fir (Abies), and apple tree (Malus) in vitro. Arzneimittel-Forschung/Drug Res. 2006;56(6a):483–96.
Wagschal I, Eggenschwiler J, von Balthazar L, Patrignani A, Rehrauer H, Schlapbach R, Ramos FMH, Viviani A. Gene expression signatures of pathway alterations in tumor cells caused by plant extracts. MEDICINA-BUENOS AIRES-. 2007;67:97.
Beztsinna N, de Matos M, Walther J, Heyder C, Hildebrandt E, Leneweit G, Mastrobattista E, Kok R. Quantitative analysis of receptor-mediated uptake and pro-apoptotic activity of mistletoe lectin-1 by high content imaging. Sci Rep. 2018;8(1):1–10.
Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, Mignot G, Maiuri MC, Ullrich E, Saulnier P, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13(9):1050–9.
Kang R, Tang D, Schapiro NE, Loux T, Livesey KM, Billiar TR, Wang H, Van Houten B, Lotze MT, Zeh HJ. The HMGB1/RAGE inflammatory pathway promotes pancreatic tumor growth by regulating mitochondrial bioenergetics. Oncogene. 2014;33(5):567–77.
Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418(6894):191–5.
Bell CW, Jiang W, Reich CF 3rd, Pisetsky DS. The extracellular release of HMGB1 during apoptotic cell death. Am J Physiol Cell Physiol. 2006;291(6):C1318-1325.
Kim JJ, Hwang YH, Kang KY, Kim I, Kim JB, Park JH, Yoo YC, Yee ST. Enhanced dendritic cell maturation by the B-chain of Korean mistletoe lectin (KML-B), a novel TLR4 agonist. Int Immunopharmacol. 2014;21:309–19.
Kim JJ, Hwang YH, Kang KY, Lee SJ, Kim JB, Choi J, Yee ST. Antitumor effect of KML-B-treated dendritic cells via induction of lymphocyte activation. J Immunol Res. 2017;2017:2471627.
Martins I, Wang Y, Michaud M, Ma Y, Sukkurwala AQ, Shen S, Kepp O, Metivier D, Galluzzi L, Perfettini JL, et al. Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ. 2014;21(1):79–91.
Medina CB, Ravichandran KS. Do not let death do us part: “find-me” signals in communication between dying cells and the phagocytes. Cell Death Differ. 2016;23(6):979–89.
Fu LL, Zhou CC, Yao S, Yu JY, Liu B, Bao JK. Plant lectins: targeting programmed cell death pathways as antitumor agents. Int J Biochem Cell Biol. 2011;43(10):1442–9.
Bhutia SK, Panda PK, Sinha N, Praharaj PP, Bhol CS, Panigrahi DP, Mahapatra KK, Saha S, Patra S, Mishra SR, et al. Plant lectins in cancer therapeutics: Targeting apoptosis and autophagy-dependent cell death. Pharmacol Res. 2019;144:8–18.
Xi H, Wang S, Wang B, Hong X, Liu X, Li M, Shen R, Dong Q. The role of interaction between autophagy and apoptosis in tumorigenesis (Review). Oncol Rep. 2022;48(6):208.
Elluru SR. Duong van Huyen JP, Delignat S, Kazatchkine MD, Friboulet A, Kaveri SV, Bayry J: Induction of maturation and activation of human dendritic cells: a mechanism underlying the beneficial effect of Viscum album as complimentary therapy in cancer. BMC Cancer. 2008;8:161.
Saha C, Das M, Stephen-Victor E, Friboulet A, Bayry J, Kaveri SV. Differential effects of Viscum album preparations on the maturation and activation of human dendritic cells and CD4+ T cell responses. Molecules. 2016;21(7):912.
Steinborn C, Klemd AM, Sanchez-Campillo AS, Rieger S, Scheffen M, Sauer B, Garcia-Kaufer M, Urech K, Follo M, Ucker A, et al. Viscum album neutralizes tumor-induced immunosuppression in a human in vitro cell model. PLoS One. 2017;12(7):e0181553.
Kovacs E. Serum levels of IL- 12 and the production of IFN- gamma, IL- 2 and IL- 4 by peripheral blood mononuclear cells (PBMC) in cancer patients treated with VISCUM ALBUM extract. Biomed Pharmacother. 2000;54(6):305–10.
Son GS, Ryu WS, Kim HY, Woo SU, Park KH, Bae JW. Immunologic response to mistletoe extract (Viscum album L.) after conventional treatment in patients with operable breast cancer. J Breast Cancer. 2010;13(1):14–8.
Van Duong Huyen JP, Delignat S, Bayry J, Kazatchkine MD, Bruneval P, Nicoletti A, Kaveri SV. Interleukin-12 is associated with the in vivo anti-tumor effect of mistletoe extracts in B16 mouse melanoma. Cancer letters. 2006;243(1):32–7.
Werthmann PG, Strater G, Friesland H, Kienle GS. Durable response of cutaneous squamous cell carcinoma following high-dose peri-lesional injections of Viscum album extracts–a case report. Phytomedicine. 2013;20(3–4):324–7.
Werthmann PG, Helling D, Heusser P, Kienle GS: Tumour response following high-dose intratumoral application of VISCUM ALBUM on a patient with adenoid cystic carcinoma. Case Reports. 2014;2014:bcr2013203180.
Werthmann PG, Saltzwedel G, Kienle GS. Minor regression and long-time survival (56 months) in a patient with malignant pleural mesothelioma under Viscum album and Helleborus niger extracts-a case report. J Thorac Dis. 2017;9(12):E1064–70.
Gutsch J, Werthmann PG, Rosenwald A, Kienle GS. Complete remission and long-term survival of a patient with a diffuse large B-cell lymphoma under Viscum album extracts after resistance to R-CHOP: a case report. Anticancer Res. 2018;38(9):5363–9.
Reynel M, Villegas Y, Kiene H, Werthmann PG, Kienle GS. Intralesional and subcutaneous application of Viscum album L. (European mistletoe) extract in cervical carcinoma in situ: a CARE compliant case report. Medicine (Baltimore). 2018;97:e13420.
Werthmann PG, Huber R, Kienle GS. Durable clinical remission of a skull metastasis under intralesional Viscum album extract therapy: Case report. Head Neck. 2018;40(7):E77–81.
Matthes B, Fritz P, Mürdter TE, Kröz M, von Laue HB, Matthes H: Untersuchungen zu Immunreaktion und klinischem Outcome nach intraläsionaler VISCUM-Applikation bei Kolonkarzinom. In: Fortschritte in der Misteltherapie, Aktueller Stand der Forschung und klinische Anwendung. edn. Edited by Scheer R, Bauer R, Becker H, Fintelmann V, Kemper F, Schilcher H. Essen: KVC Verlag; 2005. pp.491–498.
Schad F, Atxner J, Buchwald D, Happe A, Popp S, Kröz M, Matthes H. Intratumoral mistletoe (VISCUM ALBUM L) therapy in patients with unresectable pancreas carcinoma: A retrospective analysis. Integr Cancer Ther. 2014;13(4):332–40.
Nabrotzki M, Scheffler A: Komplette Remission nach intratumoraler Misteltherapie eines Duodenum- Karzinom- Rezidivs. In: Die Mistel in der Tumortherapie, Grundlagenforschung und Klinik. edn. Edited by Scheer R, Bauer R, Becker H, Fintelmann V. Essen: KVC Verlag; 2001. pp.413–422.
Wiebelitz KR, Beer A-M: Misteltherapie intraperitoneal, intrapleural und intratumoral: Wirkungen und unerwünschte Reaktionen. In: Die Mistel in der Tumortherapie 5 - Aktueller Stand der Forschung und klinische Anwendung. edn. Edited by Scheer R, Alban S, Becker H, Beer A-M, Blaschek W, Klein R, Kreis W, Leneweit G, Matthes H, Spahn G et al. Essen: KVC; 2020. pp.377–398.
Steele ML, Axtner J, Happe A, Kröz M, Matthes H, Schad F. Use and safety of intratumoral application of European mistletoe (Viscum album L.) preparations in oncology. Integr Cancer Ther. 2015;14(2):140–8.
Mistletoe-therapy. Medical Section of the School of Spiritual Science, Dornach, Switzerland. https://www.mistletoe-therapy.org/. Accessed 13 March 2025
Acknowledgements
We acknowledge Dr. João V.C. Batista for proofreading this manuscript.
Clinical trial number
Not applicable.
Funding
Not applicable.
Author information
Authors and Affiliations
Contributions
UW conceived and designed the study, UW, BL and ST performed experiments and analyses, UW and SB performed statistical analysis. UW prepared a manuscript draft, which was critically revised by BL, ST and SB. All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable. This study did not involve the use of animal or human subjects.
Consent for publication
Not applicable.
Competing interests
The Society for Cancer Research holds a minority share in stocks of the manufacturer (Iscador AG, Arlesheim, Switzerland) of the mistletoe extracts investigated.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Weissenstein, U., Tschumi, S., Leonhard, B. et al. A fermented Mistletoe (Viscum album L.) extract elicits markers characteristic for immunogenic cell death driven by endoplasmic reticulum stress in vitro. BMC Complement Med Ther 25, 175 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12906-025-04909-8
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12906-025-04909-8