Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Cancer Research

Experimental Melanoma Immunotherapy Model Using Tumor Vaccination with a Hematopoietic Cytokine

Published: February 24, 2023 doi: 10.3791/64082

Summary

The protocol presents a cancer immunotherapy model using cell-based tumor vaccination with Flt3L-expressing B16-F10 melanoma. This protocol demonstrates the procedures, including preparation of cultured tumor cells, tumor implantation, cell irradiation, measurement of tumor growth, isolation of intratumoral immune cells, and flow cytometry analysis.

Abstract

Fms-like tyrosine kinase 3 ligand (Flt3L) is a hematopoietic cytokine that promotes the survival and differentiation of dendritic cells (DCs). It has been used in tumor vaccines to activate innate immunity and enhance antitumor responses. This protocol demonstrates a therapeutic model using cell-based tumor vaccine consisting of Flt3L-expressing B16-F10 melanoma cells along with phenotypic and functional analysis of immune cells in the tumor microenvironment (TME). Procedures for cultured tumor cell preparation, tumor implantation, cell irradiation, tumor size measurement, intratumoral immune cell isolation, and flow cytometry analysis are described. The overall goal of this protocol is to provide a preclinical solid tumor immunotherapy model, and a research platform to study the relationship between tumor cells and infiltrating immune cells. The immunotherapy protocol described here can be combined with other therapeutic modalities, such as immune checkpoint blockade (anti-CTLA-4, anti-PD-1, anti-PD-L1 antibodies) or chemotherapy in order to improve the cancer therapeutic effect of melanoma.

Introduction

Cancer immunotherapy has been recognized as a promising therapeutic strategy based on its less toxic side effects and more durable responses. Several types of immunotherapies have been developed, including oncolytic virus therapies, cancer vaccines, cytokine therapies, monoclonal antibodies, adoptive cell transfer (CAR-T cells or CAR-NK), and immune checkpoint blockade1.

For cancer vaccines, there are different forms of therapeutic vaccines, such as whole cell-based vaccines, protein or peptide vaccines, and RNA or DNA vaccines. Vaccination relies on the ability of antigen-presenting cells (APCs) to process tumor antigens, including tumor-specific antigens, and present them in an immunogenic form to T cells. Dendritic cells (DCs) have been known to be the most potent APCs and are believed to play an important role in antitumor immunity2,3. These cells take up and process tumor antigens, and then migrate to the draining lymph nodes (dLN) to prime and activate tumor-specific T effector (Teff) cells through engagement of the T-cell receptor (TCR) and costimulatory molecules. This results in differentiation and expansion of tumor-specific cytotoxic T cells (CTL), which infiltrate the tumor and kill tumor cells4. Consequently, activation and maturation of DCs represent attractive strategies to stimulate immunity against tumor antigens.

Flt3L is known to promote the maturation and expansion of functionally mature DCs that express MHC class II, CD11c, DEC205, and CD86 proteins5. Intratumoral, but not intravenous, administration of an adenovirus vector incorporating the Flt3L gene (Adv-Flt3L) has been shown to promote immune therapeutic activity against orthrotopic tumors6. Flt3L has also been used in tumor cell-based vaccines consisting of irradiated B16-F10 cells stably expressing retrovirally transduced Flt3L as a way of enhancing the cross-presentation of tumor antigens by DCs and, thus, increasing antitumor responses. The protocol of B16-Flt3L tumor vaccination described here is based on a study published by Dr. James Allison's group7. In this paper, they reported that a B16-Flt3L vaccine combined with CTLA-4 blockade synergistically induced the rejection of established melanoma, resulting in increased survival.

The goal of this protocol is to provide a preclinical immunotherapy model for melanoma. Here, detailed procedures of how to prepare and implant tumor vaccines, and how to analyze the composition and function of intratumoral immune cells from solid tumor are described.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

All mice used in the study were maintained and housed in the vivarium of the La Jolla Institute for Immunology (LJI) under specific pathogen-free conditions with controlled temperature and humidity. Animal experiments were performed with 8-14 weeks old female C57BL/6 mice according to guidelines and protocols approved by the LJI Animal Care Committee.

1. Preparation of cultured tumor cells for implantation

  1. Culture B16-F10 melanoma cells in Iscove's Modified Dulbecco's medium (IMDM) containing 10% heat-inactivated FBS, 2 mM glutamine, 1 mM sodium pyruvate, 1 mM MEM nonessential amino acids, and 100 U/mL each of penicillin and streptomycin. Maintain the cell line at 37 °C under 5% CO2.
  2. Seed 1.5-2 x 106 B16-F10 cells in a 175T flask and culture for 2 days. Harvest cells when they are 75%-80% confluent.
  3. Remove the culture medium and wash the flask once with PBS. Aspirate PBS and add 5 mL of 0.25% trypsin-EDTA followed by harshly tapping the rim of the culture flask.
  4. Add 15 mL of culture medium to neutralize the trypsin-EDTA and pour the contents of the flask into a 50 mL centrifuge tube. Wash the dish surface with 10 mL of PBS and pour into the same 50 mL tube.
  5. Centrifuge the cells for 5 min at 200 x g. Discard the supernatant and break the cell pellet by finger tapping the bottom of the tube.
  6. Add cold 10 mL of PBS and gently pipet the cell suspension; then, manually count cells using hemocytometer. Keep the cells on ice before injection.

2. Tumor implantation

  1. Gas anesthetize mice with 5% isoflurane at gas flow rate of 1.0 L per min in the fume hood. Change the flow rate to the maintenance dose of 2% isoflurane once the mice are fully anesthetized. For this protocol, anesthetization was performed by the veterinarian following the institutional animal care and use guidelines.
  2. Shave the hair on the left flank of the mice and sterilize the injection site using alcohol wipes. Intradermally (i.d.) implant B16-F10 tumor cells at 5 x 105 cells in 50 µL of cold PBS in the left flank using a 30 G needle.
    NOTE: The dose of implanted B16-F10 tumor cells may need to be adjusted in the range of 0.5-5 x 105 cells for successful tumor development.
  3. After implantation, measure the tumor length and width three times a week using an electronic digital caliper. Calculate the tumor volume (mm3) using the formula:
    Tumor volume (mm3) = width2 × length × 0.5
    Treat mice with tumor vaccine when tumors have reached a size of ≥2 mm.
    ​NOTE: Tumors usually can be measured on day 3 after implantation of 5 x 105 tumor cells. Faster B16-F10 tumor growth rate was observed in male C57BL/6, Rag1-/-, or Rag2-/-γc-/-mice. A similar observation was described in other studies8. Keeping the gender of the mice consistent is recommended. However, note that the NIH places emphasis on sex as an important biological variable in biomedical research.

3. Vaccine preparation and injection of Flt3L-expressing B16-F10 (B16-Flt3L) cells

  1. Maintain the B16-Flt3L cells in DMEM containing 8% heat-inactivated FBS, 2 mM glutamine, and 100 U/mL each of penicillin and streptomycin at 37 °C under 5% CO2.
  2. Seed 1 x 106 B16-Flt3L cells in a 175T flask and culture for 2 days. Harvest cells when they are 75%-80% confluent as described in steps 1.3 to 1.6 and suspend in 1 mL of cold PBS.
  3. Irradiate cells at 150 Gy dose of gamma rays using X-ray Irradiator with 160 kV and 25 mA parameter setting. Count and check the cell viability by trypan blue staining before injection.
  4. Gas anesthetize mice as described earlier and sterilize the injection site using alcohol wipes. Intradermally inject the mice with 1 x 106 irradiated B16-Flt3L cells in 50 µL of cold PBS on the same flank as the original tumor implantation, ~1 cm away from the site of the primary tumor on days 3, 6, and 9 after the initial cell implantation.
  5. Mark the vaccine injection sites with a colored pen to distinguish it from the primary tumor.
    ​NOTE: If 0.5 x 105 B16-F10 cells are initially implanted, it is recommended to perform vaccine treatment on days 8, 11, and 14.

4. Intratumoral immune cell isolation

  1. Sacrifice mice using CO2 and cervical dislocation in the fume hood at the end of the experiment (on day 15 after tumor implantation; Figure 1).
  2. Surgically remove the tumor with the skin from each mouse and put it into 24-well plate with 1 mL 10% FBS/RPMI-1640 medium. Dry the tumors using a paper towel before weighing it.
  3. Cut the tumors into small pieces. Add 2 mL of digestion buffer (100 µg/mL TL Liberase and 200 µg/mL DNase I in RPMI-1640 medium) and incubate for 25 min at 37 °C.
  4. Add 10 mL of 10% FBS/RPMI-1640 medium to stop the digestion. Transfer the cells using a 25 mL serological pipette and use the plunger of a 1 mL syringe to grind tissue on a 40 µm cell strainer.
  5. Centrifuge the cells at 500 x g for 5 min at 4 °C. Resuspend the pellet in 5 mL of 40% density gradient specific medium in PBS, diluted to 1x concentration).
  6. Add the cell suspension slowly on top of 5 mL of 80% density-gradient-specific medium containing PBS. Centrifuge the cells at 325 x g with low brake setting for 23 min at room temperature (RT).
  7. After centrifugation, carefully collect the leukocytes layer at the interface between 40% and 80% density-gradient-specific medium and pass it through a 40 µm cell strainer. Centrifuge the cells at 500 x g for 5 min at 4 °C.
  8. Incubate the pellet in 2 mL of red blood cell (RBC) lysis buffer for 5 min at RT. After incubation, add 10 mL of 10% FBS/RPMI-1640 medium to quench the RBC lysis buffer.
  9. Centrifuge the cells at 500 x g for 5 min at 4 °C. Resuspend the cells in 0.5 mL of 10% FBS/RPMI-1640 medium and count the total number of cells before use for further analysis.
    ​NOTE: Collect the spleen or dLN as controls for gating strategy of immune cell subsets by flow cytometry analysis. Follow the cell isolation method as described above with the following changes: Use the plunger of a 1 mL syringe to grind tissue on a 70 µm mesh filter. Wash the tissue with 10% FBS/RPMI-1640 medium to get single-cell suspensions. Lyse the RBC as described.

5. Flow cytometry analysis

NOTE: Cells collected from the leukocytes layer contain immune cells and tumor cells. Two independent staining panels are recommended.

  1. Surface staining
    1. Transfer the cells into a 96-well V-shape-bottom plate. Wash the cells with PBS and stain them with cell viability dye (50-100 µL/well) for 15 min at RT.
    2. Centrifuge the cells at 500 x g for 5 min at 4 °C. Stain the surface markers with mixed antibodies (detailed dilution is provided in Table of Materials) in FACS buffer (1% FBS and 0.05% NaN3 in PBS) for 30 min on ice (50-100 µL/ well).
    3. Centrifuge the cells at 500 x g for 5 min and wash them with FACS buffer twice.
    4. Fix the cells with cell fixation buffer (50-100 µL/ well) for 35 min on ice. Centrifuge the cells at 500 x g for 5 min and wash them twice with FACS buffer.
    5. Store the samples in FACS buffer (150-200 µL/tube) at 4 °C and protect from light. Acquire the samples on a flow cytometer. For DCs and T cells population, follow the gating strategies provided in Figure 2B and Figure 3A.
      NOTE: For the staining of myeloid DCs, incubating FC-blocker (Rat anti-mouse CD16/CD32) for 15 min on ice before surface antibodies incubation (step 5.1.2) is recommended.
  2. Cytokine analysis upon ex vivo re-stimulation
    1. Plate the cells in complete medium (RPMI-1640 supplemented with 10% FBS, 10 mM HEPES, pH 7.2-7.6, 0.1 mM non-essential amino acid, 1 mM sodium pyruvate, 100 U/mL each penicillin and streptomycin, 50 μM 2-mercaptoethanol, and 2 mM L-glutamine) and stimulate with 50 ng/mL of PMA plus 1 µM ionomycin in the presence of protein transport inhibitor for 4 h at 37 °C under 5% CO2.
    2. Perform surface staining for surface markers as described above in step 5.1.
    3. Add the permeabilization solution (50-100 µL/well) and incubate for 5 min at RT for intracellular staining. Incubate the cells with antibodies specific for cytokines or nuclear proteins in permeabilization solution (50-100 µL/well) for 60 min on ice or overnight at 4 °C.
    4. Centrifuge the cells at 500 x g for 5 min and wash them with FACS buffer twice. Store the samples at 4 °C and protect from light. Acquire the samples on a flow cytometer.
      NOTE: Antibody dilution may have to be adjusted as necessary to accomplish optimal staining.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

A visible black dot of the implanted B16-F10 cells is usually observed on the skin surface ~3 days after tumor implantation. Mice are treated with the tumor vaccine 3, 6, and 9 days after the tumor nodule has reached a size of ≥2 mm. We observed a significant reduction in tumor growth in vaccinated mice group ~2 weeks after tumor implantation (Figure 1). At the end of the experiment, we isolated the intratumoral immune cells and analyzed their number and cell surface marker expression, as well as cytokine production after a short in vitro stimulation as described above. Cells collected from the leukocytes layer still contain many tumor cells, making it somewhat difficult to readily define the lymphocyte population. Therefore, using in parallel splenocytes is recommended for proper gating of intratumoral immune cell subsets in flow cytometry analysis (Figure 2A). Here, gating strategies of CD103+CD11c+DC, CD8+, CD4+, and Treg are shown (Figure 2B and Figure 3A) along with the compensation matrix (Table 1 and Table 2). Representative data of acquired counts and frequency of each population are also provided in Figure 2C and Figure 3B.

Intratumoral CD103+CD11c+DCs, which represent the most potent tumor antigen-processing and presenting cells9,10, from vaccinated mice displayed a significantly elevated expression of the costimulatory ligand CD86 (Figure 4). Vaccinated mice also displayed an increase in tumor-infiltrating CD8+ and CD4+Foxp3 T cells (Figure 5A), as well as in CD8+GzmB+ and IFN-γ+ CTLs (Figure 5B). These results suggest that this tumor vaccination promotes DC maturation and induces stronger antitumor immunity.

Figure 1
Figure 1: Analysis of tumor growth in a therapeutic model of cell-based tumor vaccine using Flt3L-expressing B16-F10 melanoma cells. Female C57BL/6 mice were implanted i.d. with B16-F10 cells (5 x 105) and injected with irradiated (150 Gy) B16-Flt3L cells (1 x 106) in an adjacent site on the same flank. The arrowheads indicate the time points of vaccination. Cumulative data of four experiments are shown (Control, n = 12; B16-Flt3L, n = 10). Data are presented as the mean ± SEM. Statistical analysis by two-way repeated measures ANOVA test with Bonferroni post-test. *P < 0.05; ***P < 0.001. This figure has been modified from11. Please click here to view a larger version of this figure.

Figure 2
Figure 2: Gating strategies of lymphocytes and CD103+CD11c+DCs. (A) Gating strategies of lymphocytes in spleen and tumor samples. (B) Gating strategies of intratumoral CD103+CD11c+DCs in tumor sample. (C) Acquired counts and frequency of each population from a representative unvaccinated control mouse are shown. Please click here to view a larger version of this figure.

Figure 3
Figure 3: Gating strategies of CD8+, CD4+, and Treg. (A) Gating strategies of T cells in tumor sample. (B) Acquired counts and frequency of each population from a representative unvaccinated control mouse are shown. Please click here to view a larger version of this figure.

Figure 4
Figure 4: CD86 surface expression on CD103+ intratumoral DCs. Expression is reported as median fluorescence intensity (MFI) normalized to average MFI in the control group (= 1). Control, n = 8; B16-Flt3L, n = 10. Data are presented as the mean ± SEM. Statistical analysis by unpaired Student's t-test. **P < 0.01. This figure has been modified from11. Please click here to view a larger version of this figure.

Figure 5
Figure 5: Analysis of intratumoral T cells in control and vaccinated mice. (A) Enumeration of tumor infiltrating CD8+ (left) and non-Treg CD4+ (right) per gram of tumor tissue. (B) Enumeration of intratumoral GzmB+ (left) and IFNγ+ (right) CD8+ T cells. Control, n = 11; B16-Flt3L, n = 10. Data are presented as the mean ± SEM. Statistical analysis by unpaired Student's t-test. *P < 0.05; **P < 0.01. This figure has been modified from11. Please click here to view a larger version of this figure.

Table 1: Compensation matrix of Figure 2. Please click here to download this Table.

Table 2: Compensation matrix of Figure 3. Please click here to download this Table.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

The protocol described here is based on the study by Allison's group. They demonstrated that combination of B16-Flt3L vaccine with CTLA-4 blockade showed a synergistic effect on survival rate and tumor growth, whereas no reduction of tumor growth was seen in mice receiving the B16-Flt3L vaccine or anti-CTLA-4 antibody treatment alone7. Recent studies have revealed a novel Treg-intrinsic CTLA4-PKCη signaling pathway that plays an important obligatory role in regulating the contact-dependent suppressive activity of Treg11. Both B16-Flt3L vaccine treatment alone or vaccine combination with Treg-specific PKCη deletion significantly reduced the growth of tumor when higher number (0.5-5 x 105) of B16-F10 melanoma cells than in the Allison's study (1 x 104) cells were implanted. Subtle differences in the source of melanoma cells or mice may account for this difference. Therefore, titrating the number of implanted tumor cells is recommended. Increased infiltration of CD8+ T cells in the primary tumor was similarly observed in the previous study7. In addition, we observed increased CD86 expression on intratumoral CD103+CD11c+DCs, which likely accounts for a stronger activation and expansion of tumor infiltrating CD8+ CTLs.

This protocol, using a cell-based tumor vaccine expressing Flt3L, is convenient and straightforward to use, and serves as a reliable model to study intratumoral immune cell infiltrates, including DCs. For example, PKCη signaling is required for the contact-dependent suppressive activity of Treg. Thus, Prkch−/− Treg displayed higher conjugation efficiency with APCs in a Treg -DC in vitro coculture system, indicating a defect in the ability of Prkch−/− Treg to break contact and disengage from attached DCs12. B16-Flt3L vaccination likely recruits more mature DCs that present tumor-specific antigens more efficiently and, thus, it is likely to facilitate observation of the interaction between tumor-infiltrating Treg and DC by live cell imaging.

Keeping a sufficient distance between the primary tumor and the tumor vaccine is an important feature of this protocol. This physical separation is critical in order to allow room for growth of the primary tumor and avoid potential fusion between the two tumor implants. Alternatively, the tumor vaccine can be implanted in the opposite flank relative to the primary tumor, as this was also reported to inhibit tumor growth7. One limitation of the study is the lack of a commercial source of B16-Flt3L cell line, but the same concept can be applied to other tumor types, for example, TRAMP-C2 prostate adenocarcinomas7.

Although the protocol described here uses B16-F10 melanoma cells as a tumor model, the underlying principles can be adapted and modified as necessary to establish immunotherapeutic models for other solid tumors. Furthermore, the vaccination protocol we used can be readily combined with other therapeutic modalities, e.g., CTLA-4- or PD-1- based checkpoint blockade, in order to achieve additive or synergistic effects that can potentially result in more potent antitumor immunity and increased survival.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors have nothing to disclose.

Acknowledgments

We thank Dr. Stephen Schoenberger for providing B16-Flt3L cells and the staff of the LJI animal and flow cytometry facilities for excellent support.

Materials

Name Company Catalog Number Comments
0.25% trypsin-EDTA  Gibco 25200-056
10% heat-inactivated FBS Omega Scientific FB-02  Lot# 209018
30G needle BD Biosciences 305106
96 well V-shape-bottom plate SARSTEDT 83.3926.500
B16 cell line expressing Fms-like tyrosine kinase 3 ligand (B16-Flt3L) Gift of Dr. Stephen Schoenberger, LJI  Flt3L cDNAs were cloned into the pMG-Lyt2 retroviral vector, as in refernce 5, Supplemental Figure 1
B16-F10 cell lines ATCC CRL-6475
Centrifuge 5810R Eppendorf
Cytofix fixation buffer  BD Biosciences BDB554655 Cell fixation buffer (4.2% PFA) 
Cytofix/Cytoperm kit  BD Biosciences 554714 Fixation/Permeabilization Solution Kit
DNase I Sigma 11284932001
Dulbecco's Modified Eagle Medium  (DMEM)  Corning 10013CV
Electronic digital caliper Fisherbrand 14-648-17
FlowJo software  Tree Star Flow cytometer data analysis
GolgiStop (protein transport inhibitor) BD Biosciences 554724 1:1500 dilution
HEPES (1M) Gibco 15630-080
Ionomycin Sigma I0634
Iscove’s modified Dulbecco’s medium (IMDM) Thermo Fisher 12440053
LSR-II cytometers  BD Biosciences Flow cytometer
MEM nonessential amino acids Gibco 11140-050
penicillin and streptomycin  Gibco 15140-122
Percoll  GE Healthcare Life Sciences GE17-0891-02 density gradient specific medium
PMA Sigma P1585
Red Blood Cell Lysing Buffer Hybri-Max liquid Sigma R7757-100ML
RPMI 1640 medium Corning 10-040-CV
RS2000 X-ray Irradiator Rad Source Technologies
sodium pyruvate Gibco 11360-070
Sterile cell strainer 40 μm Fisherbrand 22-363-547
Sterile cell strainer 70 μm Fisherbrand 22-363-548
TL Liberase Roche 477530
Zombie Aqua fixable viability kit BioLegend 423101
Antibodies
Anti-mCD45 BioLegend 103135 Clone: 30-F11
Fluorophore: BV570
Dilution: 1:200
Anti-mCD3ε BioLegend 100327 Clone: 145-2C11
Fluorophore: PerCP-Cy5.5
Dilution: 1:200
Anti-mCD8 BioLegend 100730
100724
Clone: 53-6.7
Fluorophore: Alexa Fluor 700, Alexa Fluor 647
Dilution: 1:200
Anti-mCD4 BioLegend 100414 Clone: GK1.5
Fluorophore: APC-Cy7
Dilution: 1:200
Anti-mFoxp3 Thermo Fisher Scientific 11577382 Clone: FJK-16s
Fluorophore: FITC
Dilution: 1:100
Anti-m/hGzmB BioLegend 372208 Clone: QA16A02
Fluorophore: PE
Dilution: 1:100
Anti-mIFNg BioLegend 505826 Clone: XMG1.2
Fluorophore: PE-Cy7
Dilution: 1:100
Anti-mCD19 BioLegend 115543 Clone: 6D5
Fluorophore: BV785
Dilution: 1:100
Anti-mGr1 BioLegend 108423 Clone: RB6-8C5
Fluorophore: APC/Cy7
Dilution: 1:200
Anti-mCD11b BioLegend 101223 Clone: M1/70
Fluorophore: Pacific blue
Dilution: 1:100
Anti-mF4/80 BioLegend 123114 Clone: BM8
Fluorophore: PECy7
Dilution: 1:100
Anti-mCD11c BioLegend 117328 Clone: N418
Fluorophore: PerCP Cy5.5
Dilution: 1:100
Anti-mMHCII BioLegend 107622 Clone: M5/114.15.2
Fluorophore: AF700
Dilution: 1:400
Anti-mCD103 BioLegend 121410 Clone: 2E7
Fluorophore: Alexa Fluor 647
Dilution: 1:200
Anti-mCD86 BioLegend 105007 Clone: GL-1
Fluorophore: PE
Dilution: 1:200
FC-blocker (Rat anti-mouse CD16/CD32) BD Biosciences 553141 Clone: 2.4G2
Dilution: 1:200

DOWNLOAD MATERIALS LIST

References

  1. Zhang, Y., Zhang, Z. The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell & Molecular Immunology. 17 (8), 807-821 (2020).
  2. Banchereau, J., Steinman, R. M. Dendritic cells and the control of immunity. Nature. 392 (6673), 245-252 (1998).
  3. Banchereau, J., et al. Immunobiology of dendritic cells. Annual Review of Immunology. 18, 767-811 (2000).
  4. Martinez-Lostao, L., Anel, A., Pardo, J. How do cytotoxic lymphocytes kill cancer cells. Clinical Cancer Research. 21 (22), 5047-5056 (2015).
  5. Maraskovsky, E., et al. Dramatic increase in the numbers of functionally mature dendritic cells in Flt3 ligand-treated mice: multiple dendritic cell subpopulations identified. Journal of Experimental Medicine. 184 (5), 1953-1962 (1996).
  6. Talmadge, J. E., et al. Intratumoral, injection of adenoviral Flt3 ligand has therapeutic activity in association with increased intratumoral levels of T cells but not dendritic cells. Blood. 104 (11), 5280 (2004).
  7. Curran, M. A., Allison, J. P. Tumor vaccines expressing flt3 ligand synergize with ctla-4 blockade to reject preimplanted tumors. American Association for Cancer Research. 69 (19), 7747-7755 (2009).
  8. Simon, S. R., Ershler, W. B. Hormonal influences on growth of B16 murine melanoma. Journal of the National Cancer Institute. 74 (5), 1085-1088 (1985).
  9. Broz, M. L., et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell. 26 (6), 938 (2014).
  10. Salmon, H., et al. Expansion and activation of CD103(+) dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity. 44 (4), 924-938 (2016).
  11. Liu, H. Y., et al. Leveraging the Treg-intrinsic CTLA4-PKCeta signaling pathway for cancer immunotherapy. Journal for Immunotherapy Cancer. 9 (9), 002792 (2021).
  12. Kong, K. F., et al. Protein kinase C-eta controls CTLA-4-mediated regulatory T cell function. Nature Immunology. 15 (5), 465-472 (2014).

Tags

Melanoma Immunotherapy Tumor Vaccination Hematopoietic Cytokine Solid Tumor Immunotherapy Research Platform Tumor Vaccine Checkpoint Blockade Additive Effect Synergistic Effect Tumor Immunity B16-F10 Melanoma Cells IMDM FBS Glutamine Sodium Pyruvate MEM Non-essential Amino Acids Penicillin Streptomycin Trypsin-EDTA
Experimental Melanoma Immunotherapy Model Using Tumor Vaccination with a Hematopoietic Cytokine
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Liu, H. Y., Altman, A.,More

Liu, H. Y., Altman, A., Canonigo-Balancio, A. J., Croft, M. Experimental Melanoma Immunotherapy Model Using Tumor Vaccination with a Hematopoietic Cytokine. J. Vis. Exp. (192), e64082, doi:10.3791/64082 (2023).

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