Waiting
登录处理中...

Trial ends in Request Full Access Tell Your Colleague About Jove

Cancer Research

In Vivo Immunogenicity Screening of Tumor-Derived Extracellular Vesicles by Flow Cytometry of Splenic T Cells

Published: September 23, 2021 doi: 10.3791/62811

Summary

This manuscript describes how to assess in vivo immunogenicity of tumor cell-derived extracellular vesicles (EVs) using flow cytometry. EVs derived from tumors undergoing treatment-induced immunogenic cell death seem particularly relevant in tumor immunosurveillance. This protocol exemplifies the assessment of oxaliplatin-induced immunostimulatory tumor EVs but can be adapted to various settings.

Abstract

Immunogenic cell death of tumors, caused by chemotherapy or irradiation, can trigger tumor-specific T cell responses by releasing danger-associated molecular patterns and inducing the production of type I interferon. Immunotherapies, including checkpoint inhibition, primarily rely on preexisting tumor-specific T cells to unfold a therapeutic effect. Thus, synergistic therapeutic approaches that exploit immunogenic cell death as an intrinsic anti-cancer vaccine may improve their responsiveness. However, the spectrum of immunogenic factors released by cells under therapy-induced stress remains incompletely characterized, especially regarding extracellular vesicles (EVs). EVs, nano-scale membranous particles emitted from virtually all cells, are considered to facilitate intercellular communication and, in cancer, have been shown to mediate cross-priming against tumor antigens. To assess the immunogenic effect of EVs derived from tumors under various conditions, adaptable, scalable, and valid methods are sought-for. Therefore, herein a relatively easy and robust approach is presented to assess EVs' in vivo immunogenicity. The protocol is based on flow cytometry analysis of splenic T cells after in vivo immunization of mice with EVs, isolated by precipitation-based assays from tumor cell cultures under therapy or steady-state conditions. For example, this work shows that oxaliplatin exposure of B16-OVA murine melanoma cells resulted in the release of immunogenic EVs that can mediate the activation of tumor-reactive cytotoxic T cells. Hence, screening of EVs via in vivo immunization and flow cytometry identifies conditions under which immunogenic EVs can emerge. Identifying conditions of immunogenic EV release provides an essential prerequisite to testing EVs' therapeutic efficacy against cancer and exploring the underlying molecular mechanisms to ultimately unveil new insights into EVs' role in cancer immunology.

Introduction

The immune system plays a pivotal role in the fight against cancer, both when incited by immune checkpoint inhibition and for the efficacy of conventional cancer therapies. Tumor cells succumbing to genotoxic therapies such as the chemotherapeutic agents oxaliplatin and doxorubicin, or ionizing radiation treatment can release antigens and danger-associated molecular patterns (DAMPs) that potentially initiate an adaptive anti-tumor immune response1. The most prominent DAMPs, in the context of immunogenic cell death, include find-me signals such as chemotactic ATP, eat-me signals such as the exposure of calreticulin, that promotes tumor cell uptake by antigen-presenting cells, and the release of HMGB1, that activates pattern recognition receptors, thereby enhancing the cross-presentation of tumor antigens2. Furthermore, type I interferons (IFN-I), induced via tumor-derived immunogenic nucleic acids or other stimuli, are sensed by dendritic cells, enabling them to effectively prime tumor-specific cytotoxic T cells3,4. Clinically, activated and proliferating CD8+ T cells infiltrating the tumor provide an independent prognostic factor for prolonged survival in many cancer patients. Released from such activated T cells, IFN-γ mediates direct antiproliferative effects on cancer cells and drives Th1 polarization and cytotoxic T cell differentiation, thereby contributing to effective immunosurveillance against cancer5,6. Oxaliplatin is a bona fide immunogenic cell death inducer, mediating such adaptive immune response against cancer7. However, the plethora of initial immunogenic signals released by tumor cells under therapy-induced stress remains to be fully unveiled. Despite significant advances in cancer immunotherapy, expanding its benefits to a larger portion of patients remains a challenge. A more detailed understanding of immunogenic signals that initiate T cell activation may guide the development of novel therapies.

A heterogeneous group of membrane-enclosed structures, known as extracellular vesicles (EVs), seem to serve as intercellular communication devices. Emitted by virtually all cell types, EVs carry functional proteins, RNA, DNA, and other molecules to a recipient cell or may alter a cell's functional state just by binding to receptors on the cell surface. Their biologically active cargo varies significantly by the type and functional state of the generating cell8. In cancer immunology, EVs released from tumor cells have been predominantly regarded as adversarial to immunotherapy because they eventually promote invasive growth, preform metastatic niches9, and suppress the immune response10. In contrast, some studies have shown that EVs can transfer tumor antigens to dendritic cells for effective cross-presentation11,12. EVs may provide immunostimulatory nucleic acids if they emerge under therapy-induced stress, facilitating an anti-tumor immune response13,14. Sensing of such RNA and DNA innate immune ligands in the tumor microenvironment has recently been shown to modulate responsiveness to checkpoint blockade significantly15,16,17. Hence, the immunogenic role of EVs released by tumor cells under different therapy-induced stress needs to be further elucidated. Since EVs constitute a young yet growing field of research, standardization of methods is still ongoing. Therefore, sharing knowledge is essential to improve the research reproducibility on interactions between EVs and cancer immunology. With this in mind, this manuscript describes a simple protocol to assess the immunogenic effect of tumor-derived EVs in vivo.

This assessment is performed by generating tumor-derived EVs, immunizing recipient mice with those EVs, and analyzing splenic T cells via flow cytometry. EV generation is ideally performed by seeding murine tumor cells in an EV-free cell culture medium for a high degree of purity. Cells are treated with a specific cell stress stimulus, such as chemotherapy, to compare the effect of therapy-induced EVs against baseline immunogenicity of respective tumor-derived EVs. The isolation of EVs may well be performed by various techniques that should be selected according to in vivo applicability and local availability. The following protocol describes a precipitation-based assay with a commercial kit for EV purification. Mice are immunized twice with those EVs. Fourteen days after the first injection, T cells are extracted from the spleen and analyzed for IFN-γ production via flow cytometry to evaluate a systemic immune response. With this, the potential of tumor-derived EVs, emerging under different therapeutic regimens, to induce anti-tumor T cell responses is assessed relatively easily, quickly, and with high validity13. Therefore, this method is suitable for an immunological screening of EVs derived from cancer cells under various conditions.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

At the onset of experiments, mice were at least 6 weeks of age and were maintained under specific pathogen-free conditions. The present protocol complies with the Institutional ethical standards and prevailing local regulations. Animal studies were approved by the local regulatory agency (Regierung von Oberbayern, Munich, Germany). Possible sex-related biases were not investigated in these studies.

1. Generation and isolation of EVs derived from tumor cells after chemotherapy exposure

  1. Culture murine B16 melanoma cells expressing ovalbumin (B16-OVA) in DMEM (containing 4 mM L-glutamine and 4.5 g/L D-glucose) supplemented with FCS (10% v/v), penicillin (100 Units/mL), and streptomycin (100 μg/mL) at 37 °C, until cells grow steadily and are approximately 90% confluent.
    NOTE: Perform this section of the protocol entirely under sterile conditions using a cell culture hood. For analysis of other cancer entities, cell lines expressing a potent antigen are preferable to assess antigen-specific T cell responses, as they allow for specific ex vivo antigen restimulation.
  2. To prepare the cell culture media required for EV generation, deplete bovine EVs within FCS by ultracentrifugation at 100,000 x g for 24 h at 4 °C, and then discard the pellet. Alternatively, choose a commercial preparation low in animal EVs beforehand.
  3. Harvest B16-OVA cells and wash them twice in PBS, and then seed at a concentration of 400,000 cells/mL in EV-depleted media.
    NOTE: Adapt the cell concentration to the growth dynamic of the cell line under investigation so that cells do not overgrow.
  4. Treat B16-OVA cells by adding 30 μg of oxaliplatin per mL and incubate for 24 h at 37 °C. Leave control conditions untreated. At the first use of a genotoxic substance, titrate the desired cytotoxic efficacy using cell-viability assays such as trypan blue exclusion18.
    NOTE: This assay may also evaluate EVs generated in cell cultures treated with other immune-modulating substances or ionizing irradiation besides chemotherapeutics.
    CAUTION: Oxaliplatin causes skin and severe eye irritation and may cause an allergic skin reaction and respiratory irritation. Oxaliplatin is suspected of causing cancer. As a precaution, use personal protective equipment, including adequate gloves, goggles, masks, and clothing, cleaned before reuse. Avoid inhalation and wash hands thoroughly after handling. Avoid release into the environment and dispose oxaliplatin according to prevailing regulations. Obtain detailed information from the safety data sheet.
  5. Collect cell culture supernatant. Centrifuge first at 400 x g for 5 min at 4 °C, and then at 2,000 x g for 30 min at 4 °C, each time discarding the pellet. Finally, filter through a 220 nm PVDF membrane. Use a fresh tube for each step to remove any cell debris.
    NOTE: At this stage, the EV-containing supernatant may be stored at 4 °C for a day before resuming the protocol. However, it is strongly recommended to adhere to the described schedule with immediate EV purification.
  6. Mix 1 mL of supernatant with 0.5 mL of a specific commercially available exosome isolation reagent (see Table of Materials) in a V-shaped 1.5 mL tube. Thoroughly pipette up and down or vortex to create a homogenous solution. Incubate overnight at 4 °C.
  7. Centrifuge at 10,000 x g for 60 min at 4 °C. Carefully discard the supernatant. Remove the remaining drops by tapping the 1.5 mL tube upside down on a paper towel and by aspiration through a pipette with a fine tip without touching the EV-pellet at the bottom.
    1. Thoroughly remove all fluids to prevent uncontrolled dilution of the EV-pellet. Also, execute these tasks quickly to prevent the pellet from drying out.
  8. Resuspend the EVs in cold PBS by pipetting up and down without scratching the pellet from the tube's wall with the tip. Now, transfer the suspension step by step from the first to the last tube to pool the EVs.
    NOTE: Use a volume of PBS that equals 5 μL multiplied by the number of tubes. 5 μL of the final suspension contains the isolated EVs released from 400,000 cells under chemotherapy or at a steady state.
  9. Preferably, use EVs directly. If this is not possible, store EV suspensions at -80 °C in siliconized vessels for up to 28 days until application.
    NOTE: The EVs described here and in several other publications do not lose their respective biological function when stored at -80 °C for that time period19.
  10. Quantify and characterize EV isolates according to the MISEV2018 guidelines20.
    NOTE: Possible methods for quantification include nanoparticle tracking analysis (NTA)21 and the detection of EV's membrane-bound proteins22. Possible approaches to further characterize EVs include electron microscopy23 and western blot20.

2. Immunization of mice with EVs

  1. Plan the in vivo experiment with C57BL/6 mice (or other syngeneic mice corresponding to the tumor cell line), including treatment groups receiving EVs derived from treated cells, untreated cells, and PBS (vehicle), respectively.
    NOTE: Preferably, use mice at the age of 6-8 weeks to prevent physiological senescence from diminishing the immune response24.
  2. Mix 5 μL of EV-suspension with 55 μL cold PBS for each mouse within the respective treatment group to immunize it with EVs isolated from 4.0 x 105 B16-OVA cells.
    NOTE: This amount of EVs corresponds to approximately 2 x 109 particles measured by nanoparticle tracking analysis (data not shown). OVA protein mixed with an adjuvant (e.g., LPS) can be applied as a potent vaccine positive control.
    1. Fill syringes (needle size 26-30 G) with 60 μL of the diluted EVs or PBS, respectively and put immediately on ice.
      NOTE: In the protocol, the amount of injected EVs is normalized to the number of EV-releasing tumor cells to experimentally consider both qualitative and quantitative effects of oxaliplatin on tumor cell EV biogenesis. For some readers, normalization to a specific concentration of produced EVs may better fit their experimental setup depending on their scientific question.
  3. Inoculate EVs or PBS subcutaneously into the medial aspect of the mice's thigh and repeat the immunization after 7 days. Fourteen days after the first treatment, sacrifice mice, e.g., by cervical dislocation to analyze the immune response.
    ​NOTE: Alternative subcutaneous injection routes may be used, according to the local standards.

3. Flow cytometry analysis of splenic T cells

  1. Prepare and cool complete RPMI (cRPMI), supplementing RPMI-1640 with FCS (10% v/v), penicillin (100 Units/mL), streptomycin (100 μg/mL), L-glutamine (2 mM), and β-mercaptoethanol (50 μM).
  2. Resect the spleen from the opened abdominal cavity. Mash the spleen with a moistened 100 μm cell strainer and the plastic plunger of a syringe and flush the splenic cells into a 50 mL tube with 5-10 mL of cRPMI. Centrifuge at 400 x g for 5 min at 4 °C and discard the supernatant.
    NOTE: Keep cells on ice whenever possible. To analyze the local rather than the splenic immune response, resect the draining popliteal and inguinal lymph nodes, following the same protocol. In this case, skip the next step for the lysis of erythrocytes.
  3. To remove erythrocytes from the cell suspension, resuspend the pellet with 2 mL of red blood cell lysis buffer (see Table of Materials) and incubate for 5 min at room temperature. Then, stop the reaction by adding cRPMI. Centrifuge at 400 x g for 5 min at 4 °C and discard the supernatant.
  4. For seeding, resuspend the cell pellet in cRPMI and count the cells to place triplicates of 200,000 cells with 200 μL cRPMI into each well, using a 96-well plate with a U-shaped bottom. Incubate for 48 h at 37 °C.
    NOTE: To address the antigen-specificity of activated T cells, add 1 μg/mL soluble ovalbumin (or another tumor antigen corresponding to cell line under investigation) or leave without additional stimulus, respectively. Adding the immune-dominant peptide epitope SIINFEKL, instead of full-length ovalbumin, allows for a shorter incubation period. Besides flow cytometry, the mice's serum and the cell culture supernatant after 48 h of incubation can be analyzed for various cytokines.
  5. After 48 h, to enhance intracellular IFN-γ staining by, inter alia, blocking the Golgi-mediated secretion of proteins, add Brefeldin A (5 ng/mL), PMA (20 ng/mL), and Ionomycin (1 µg/mL) to the cell culture. Incubate for 4 h at 37 °C.
  6. Before staining surface biomarkers, transfer splenocytes to a 96-well plate with a V-shaped bottom and wash twice with PBS. Then, add fluorescent antibodies, compatible with the locally available flow cytometer, directed against surface biomarkers, CD3, CD8, and CD4 (see Table of Materials), diluted 1:400, plus a fixable viability dye, diluted 1:1,000 in PBS. Resuspend pelleted splenocytes in the staining solution and incubate for 30 min at 4 °C, protected from light.
  7. For fixation and permeabilization of splenocytes, wash twice in FACS-buffer (PBS plus 3% v/v FCS), and then resuspend in 100 µL of fixation/permeabilization buffer (see Table of Materials) per well. Incubate for 30 min at 4 °C, protected from light.
  8. For staining of intracellular IFN-γ, wash splenocytes in fixation/permeabilization buffer and resuspend with fluorescent antibodies against IFN-γ, diluted 1:200 in the buffer. Incubate for at least 1 h (up to a maximum of 12 h) at 4 °C, protected from light.
  9. Before measuring the samples by flow cytometry, wash splenocytes twice in fixation/permeabilization buffer and resuspend in FACS-buffer. Analyze the activation of cytotoxic T cells according to the gating strategy displayed in Figure 2.
    1. First, to detect single cells, blot FSC-H against FSC-A. Then, to detect lymphoid cells, blot SSC against FSC-A. Subsequently, select living CD3+, CD4-, CD8+ cells and determine their IFN-γ-producing subset to quantify the activation of cytotoxic T cells in the spleen.
      NOTE: Include a Fluorescence-minus-one (FMO) stain with all fluorochromes except the fluorochrome targeted against IFN-γ as negative technical control.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

This protocol is intended to facilitate the straightforward and easily reproducible assessment of the immunogenicity of tumor-derived EVs. Hereby, mice are inoculated with EVs derived from in vitro cultures of tumor cells expressing the model antigen chicken ovalbumin (OVA). The subsequent immune response is analyzed in splenic T cells via flow cytometry.

Figure 1 gives an overview of the practical steps of the entire protocol. Since the work focuses on immunogenic cell death, cross-presentation, and EV-induced anti-tumor immunity, this protocol is restricted to the function of CD8+ cytotoxic T cells. As displayed in Figure 2, cells were gated as single cells, lymphocyte subset (by size and granularity), viable cells (excluding a life/dead marker), and CD3+ CD4- CD8+ cytotoxic T cells. Intracellular accumulation of IFN-γ was assessed as a surrogate marker for activation. Possible additional markers regarding T cell differentiation and exhaustion are discussed below.

Using the method described here, mice were immunized with EVs derived from OVA-expressing tumor cells cultured either under steady-state (untreated) or genotoxic stress conditions (oxaliplatin-treated). Only mice injected with EVs derived from tumor cells under genotoxic stress conditions induced potent activation of splenic cytotoxic T cells in recipient animals (Figure 3A). Injection of EVs derived from tumors under steady-state conditions resulted in some T cell activation, but that was not significantly different from T cell activation in mice injected with the PBS vehicle. These data show that under genotoxic stress, tumor cells can release potently immunogenic EVs. The production of IFN-γ was particularly increased when splenocytes of tumor EV-treated animals were ex vivo restimulated with the model tumor antigen OVA before analysis (Figure 3B). These data suggest that tumor-derived EVs can induce tumor antigen-specific immune responses. Interestingly, IFN-γ-production - even though to a much lesser extent - is also detected in the absence of antigen-specific restimulation. Possibly, other melanoma-associated antigens, such as the differentiation antigen TRP225, may be targeted by some part of the EV-induced T cell response.

Figure 1
Figure 1: Pictographic overview of the protocol. (A) Isolation procedure of EVs generated in tumor cell cultures resembling chemotherapy. (B) Schedule for the immunization of mice with EVs. (C) Staining protocol for flow cytometry analysis of cytotoxic T cells. Please click here to view a larger version of this figure.

Figure 2
Figure 2: Flow cytometry gating strategy to analyze cytotoxic T cell activation in the spleen. The numbers represent the percentage of its respective parent population. FSC-A: forward scatter area; FSC-H: forward scatter height; SSC: sideward scatter; live/dead: cell death marker. Please click here to view a larger version of this figure.

Figure 3
Figure 3: EVs derived from tumor cells under genotoxic stress can induce antigen-specific T cell responses in recipient animals. (A) Mice were immunized with EVs derived from tumor cells cultured either under steady-state (untreated) or genotoxic stress conditions (oxaliplatin-treated). Vehicle (PBS) injections were used as a negative control. IFN-γ production by cytotoxic T cells in the spleen upon EV immunization was determined. With this, splenic cell suspensions were restimulated with ovalbumin ex vivo before analysis. (B) Mice were treated with EVs derived from tumor cells under genotoxic stress conditions as described above. Splenic T cell activation was determined after ex vivo restimulation either in the presence or absence of ovalbumin. Bars depict the mean per group and whiskers its standard error. The one-way analysis of variance (ANOVA) test with Bonferroni posttest was used for multiple statistical comparisons of a dataset. The significance level was set at P < 0.05, P < 0.01, and P < 0.001 and is indicated here with asterisks (*, **, and ***). Please click here to view a larger version of this figure.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

This protocol provides an immunological in vivo assessment of EVs derived from melanoma cells under chemotherapy-induced stress while adapting to EVs emitted from various cancers under various treatments. Immunizing mice with EVs derived from oxaliplatin-treated B16-OVA cells, for instance, expands IFN-γ-producing CD8+ T cells in the spleen, which are further stimulated by ex vivo incubation with ovalbumin, indicating a tumor-specific immune response. Thus, detection of immunogenic EVs by screening through this protocol facilitates a more comprehensive understanding of conventional cancer therapies and enables a focused investigation into the role of EVs in cancer immunology.

Of note, experiments with EVs require some special considerations. In this protocol, EVs are semi-quantitatively normalized to the number of tumor cells released within 24 h. This approach reflects the aim to identify enhanced immunogenicity, regardless of whether it emerges from alterations in quality or quantity of released EVs. Therefore, reproducible in vivo results rely on the constant isolation efficacy of EVs. To this end, ensuring that EV pellets are resuspended entirely and promptly to avoid desiccation is a critical step.

Additionally, EVs must be quantified and characterized, e.g., by nanoparticle tracking analysis and western blot of canonical transmembrane, luminal, and at least one negative EV-marker20. Quantifying and characterizing EVs controls for inconstant isolation and addresses quantitative differences in EVs or EV subsets. This type of EV characterization constitutes a part of the minimum information that needs to be reported in studies about extracellular vesicles, according to the International Society of Extracellular Vesicles (ISEV) guidelines. However, various characterization methods are equally legitimate and should be selected concerning local availability and the individual research question. Notably, defining dosage of a substance of interest or ionizing irradiation constitutes another critical step of the protocol, which may require experimental validation to achieve an adequate level of cell death.

In general, potential contamination of isolated EVs with the treatment substance, soluble proteins, and lipoproteins must be considered. One strategy in this regard consists in reproducing the experiment with complementary EV-isolation techniques that the same type of contamination20 may not compromise. Immunoaffinity, for instance, isolates EVs with a lower yield but higher specificity than purely precipitation-based methods and may provide an appropriate control in this regard26. An alternative approach is to compare wild-type cell-derived EVs with isolations from genetically engineered cells with a specific deletion of genes involved in EV biogenesis or deploy substances that reduce EVs emission20.

Results obtained from this protocol should be complemented by a more comprehensive characterization of the EV-mediated immune response. Especially the classification of CD8+ T cells into effector and effector memory cells, through CD4427, as well as antigen-naïve and central memory cells, through CD62L28, may convey deeper insight. Furthermore, the analysis of T helper cells, regulatory T cells, and NK cells may be of interest. For testing the anti-tumor efficacy of EVs, mice may be challenged with the corresponding cancer cells after receiving EV immunization or treated with EVs against a preestablished cancer, thereby adapting the guidelines for detection of immunogenic cell death2,29 to this cell-free tumor derivate. However, conclusions from all these experimental setups are limited by the fact that cancer cells in a Petri dish potentially generate functionally different EVs than cancer cells embedded in a dynamic tumor microenvironment30 that often suppresses anti-cancer immunity. Thus, assessing EVs derived from tumor/fibroblast cocultures or ex vivo tumor tissue may better reflect the actual situation. In a next translational step toward clinical reality, EVs from patient material may be analyzed for immunogenicity before and during therapy to assess their usability as biomarkers.

As cancer-derived EVs were recently found to - under certain circumstances - modulate the immune system, the journey of exploring their clinical potential has only just begun31. For an in-depth analysis of the immune mechanisms co-opted by immunogenic EVs, useful tools comprise fluorescence microscopy visualizing the EV uptake by specific cells, the deployment of mice with genetic deficiencies for specific immune pathways, and screening methods for molecular alterations in the EV content. Ultimately, identifying immunogenic tumor-derived EVs, with screening methods described herein, will enable a better understanding of the underlying mechanisms behind EV-mediated immunity and therefore constitutes a crucial step toward harnessing their potential against cancer.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors declare that there is no conflict of interest.

Acknowledgments

This study was supported by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) - Projektnummer 360372040 - SFB 1335 and Projektnummer 395357507 - SFB 1371 (to H.P.), a Mechtild Harf Research Grant from the DKMS Foundation for Giving Life (to H.P.) a Young Investigator Award by the Melanoma Research Alliance (to S.H.), a scholarship by the Else-Kröner-Fresenius-Stiftung (to F.S.), a Seed Fund by the Technical University Munich (to S.H.) and a research grant by the Wilhelm Sander Foundation (2021.041.1, to S.H.). H. P. is supported by the EMBO Young Investigator Program.

AUTHOR CONTRIBUTIONS:

F.S., H.P., and S.H. designed the research, analyzed, and interpreted the results. F.S. and S.H. wrote the manuscript. H.P. and S.H. guided the study.

Materials

Name Company Catalog Number Comments
Anti-CD3 FITC Biolegend 100204 Clone 17A2
Anti-CD4 PacBlue Biolegend 100428 Clone GK1.5
Anti-CD8 APC Biolegend 100712 Clone 53-6.7
Anti-IFNγ PE eBioscience RM90022 Clone XMG1.2
Brefeldin A Biolegend 420601 Brefeldin A Solution (1,000x)
Cell Strainer, 100 µm Greiner 542000 EASYstrainer 100 µm
DMEM Sigma-Aldrich D6429 Dulbecco's Modified Eagle's Medium with D-glucose (4.5 g/L) and L-glutamine (4 mM)
FBS Good Forte PAN BIOTECH P40-47500 Fetal Calf Serum (FCS)
Fixable Viability Dye eFluor 506 eBioscience, division of Thermo Fischer Scientific 65-0866-14
Fixation/Permeabilization Concentrate eBioscience 00-5123-43 Fixation/Permeabilization Concentrate (10x)
Fixation/Permeabilization Diluent eBioscience 00-5223-56
Ionomycin Sigma-Aldrich 407952 From Streptomyces conglobatus - CAS 56092-82-1, ≥ 97% (HPLC)
L-Glutamine Gibco 25030-032 L-Glutamine (200 mM)
Ovalbumin InvivoGen vac-pova Ovalbumine with < 1 EU/mg endotoxin - CAS 9006-59-1
Oxaliplatin Pharmacy of MRI hospital
PBS Sigma-Aldrich D8537 Phosphate Buffered Saline without calcium chloride and magnesium chloride
Penicillin-Streptomycin Gibco 1514-122 Mixture of penicillin (10,000 U/mL) and streptomycin (10,000 ug/mL)
PMA Sigma-Aldrich P1585 Phorbol 12-myristate 13-acetate, ≥ 99% (HPLC)
PVDF filter, 0,22 µm, for syringes Merck Millipore SLGV033RS Millex-GV Filter Unit 0.22 µm Durapore PVDF Membrane
Red Blood Cell Lysis Buffer Invitrogen 00-4333-57
RPMI 1640 Thermo Fischer Scientific 11875 Roswell Park Memorial Institute 1640 Medium with D-glucose (2.00 g/L) and L-glutamine (300 mg/L), without HEPES
Syringe, 26 G BD Biosciences 305501 1 mL Sub-Q Syringes with needle (0.45 mm x 12.7 mm)
Total Exosome Isolation Reagent Invitrogen 4478359 For isolation from cell culture media
β-Mercaptoethanol Thermo Fischer Scientific 31350 β-Mercaptoethanol (50 mM)

DOWNLOAD MATERIALS LIST

References

  1. Kroemer, G., Galluzzi, L., Kepp, O., Zitvogel, L. Immunogenic cell death in cancer therapy. Annual Review of Immunology. 31, 51-72 (2013).
  2. Kepp, O., et al. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology. 3 (9), 955691 (2014).
  3. Fuertes, M. B., et al. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8{alpha}+ dendritic cells. The Journal of Experimental Medicine. 208 (10), 2005-2016 (2011).
  4. Sistigu, A., et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nature Medicine. 20 (11), 1301-1309 (2014).
  5. Fridman, W. H., Pages, F., Sautes-Fridman, C., Galon, J. The immune contexture in human tumours: impact on clinical outcome. Nature reviews. Cancer. 12 (4), 298-306 (2012).
  6. Shankaran, V., et al. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature. 410 (6832), 1107-1111 (2001).
  7. Tesniere, A., et al. Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene. 29 (4), 482-491 (2010).
  8. van Niel, G., D'Angelo, G., Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nature reviews. Molecular Cell Biology. 19 (4), 213-228 (2018).
  9. Zomer, A., van Rheenen, J. Implications of extracellular vesicle transfer on cellular heterogeneity in cancer: What are the potential clinical ramifications. Cancer Research. 76 (8), 2071-2075 (2016).
  10. Whiteside, T. L. Exosomes and tumor-mediated immune suppression. The Journal of Clinical Investigation. 126 (4), 1216-1223 (2016).
  11. Wolfers, J., et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nature Medicine. 7 (3), 297-303 (2001).
  12. Zeelenberg, I. S., et al. Targeting tumor antigens to secreted membrane vesicles in vivo induces efficient antitumor immune responses. Cancer Research. 68 (4), 1228-1235 (2008).
  13. Diamond, J. M., et al. Exosomes Shuttle TREX1-Sensitive IFN-Stimulatory dsDNA from Irradiated Cancer Cells to DCs. Cancer Immunology Research. 6 (8), 910-920 (2018).
  14. Kitai, Y., et al. DNA-containing exosomes derived from cancer cells treated with topotecan activate a STING-dependent pathway and reinforce antitumor immunity. Journal of Immunology. 198 (4), 1649-1659 (2017).
  15. Heidegger, S., et al. RIG-I activation is critical for responsiveness to checkpoint blockade. Science Immunology. 4 (39), 8943 (2019).
  16. Schadt, L., et al. Cancer-cell-intrinsic cGAS expression mediates tumor immunogenicity. Cell Reports. 29 (5), 1236-1248 (2019).
  17. Cheng, Y., et al. In situ immunization of a TLR9 agonist virus-like particle enhances anti-PD1 therapy. Journal for Immunotherapy of Cancer. 8 (2), (2020).
  18. Strober, W. Trypan blue exclusion test of cell viability. Current Protocols in Immunology. 111, 1-3 (2015).
  19. Jeyaram, A., Jay, S. M. Preservation and storage stability of extracellular vesicles for therapeutic applications. The AAPS Journal. 20 (1), 1 (2017).
  20. Thery, C., et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. Journal of Extracellular Vesicles. 7 (1), 1535750 (2018).
  21. Vestad, B., et al. Size and concentration analyses of extracellular vesicles by nanoparticle tracking analysis: a variation study. Journal of Extracellular Vesicles. 6 (1), 1344087 (2017).
  22. Suarez, H., et al. A bead-assisted flow cytometry method for the semi-quantitative analysis of Extracellular Vesicles. Scientific Reports. 7 (1), 11271 (2017).
  23. Yuana, Y., et al. Cryo-electron microscopy of extracellular vesicles in fresh plasma. Journal of Extracellular Vesicles. 2, (2013).
  24. Kapasi, Z. F., Murali-Krishna, K., McRae, M. L., Ahmed, R. Defective generation but normal maintenance of memory T cells in old mice. European Journal of Immunology. 32 (6), 1567-1573 (2002).
  25. Bloom, M. B., et al. Identification of tyrosinase-related protein 2 as a tumor rejection antigen for the B16 melanoma. The Journal of Experimental Medicine. 185 (3), 453-459 (1997).
  26. Patel, G. K., et al. Comparative analysis of exosome isolation methods using culture supernatant for optimum yield, purity and downstream applications. Scientific Reports. 9 (1), 5335 (2019).
  27. DeGrendele, H. C., Kosfiszer, M., Estess, P., Siegelman, M. H. CD44 activation and associated primary adhesion is inducible via T cell receptor stimulation. The Journal of Immunology. 159 (6), 2549-2553 (1997).
  28. Yang, S., Liu, F., Wang, Q. J., Rosenberg, S. A., Morgan, R. A. The shedding of CD62L (L-selectin) regulates the acquisition of lytic activity in human tumor reactive T lymphocytes. PLoS One. 6 (7), 22530 (2011).
  29. Bek, S., et al. Targeting intrinsic RIG-I signaling turns melanoma cells into type I interferon-releasing cellular antitumor vaccines. Oncoimmunology. 8 (4), 1570779 (2019).
  30. Nabet, B. Y., et al. Exosome RNA unshielding couples stromal activation to pattern recognition receptor signaling in cancer. Cell. 170 (2), 352-366 (2017).
  31. Pitt, J. M., Kroemer, G., Zitvogel, L. Extracellular vesicles: masters of intercellular communication and potential clinical interventions. The Journal of Clinical Investigation. 126 (4), 1139-1143 (2016).

Tags

In Vivo Immunogenicity Screening Tumor-derived Extracellular Vesicles Flow Cytometry Splenic T Cells Cancer Immunotherapy Personalized Anti-cancer Treatment Autologous Extracellular Vesicles Cell Culture Media Ultracentrifugation B16 Melanoma Cells Ovalbumin Expression Oxaliplatin Treatment
<em>In Vivo</em> Immunogenicity Screening of Tumor-Derived Extracellular Vesicles by Flow Cytometry of Splenic T Cells
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Stritzke, F., Poeck, H., Heidegger,More

Stritzke, F., Poeck, H., Heidegger, S. In Vivo Immunogenicity Screening of Tumor-Derived Extracellular Vesicles by Flow Cytometry of Splenic T Cells. J. Vis. Exp. (175), e62811, doi:10.3791/62811 (2021).

Less
Copy Citation Download Citation Reprints and Permissions
View Video

Get cutting-edge science videos from JoVE sent straight to your inbox every month.

Waiting X
Simple Hit Counter