Jennifer Rowley [1]
[1] Harvard Medical School, Boston, Massachusetts 02115
Correspondance: Jennifer_Rowley@hms.harvard.edu
ABSTRACT
Ovarian cancer is the most lethal gynecological malignancy, largely driven by high rates of relapse and chemoresistance. Ovarian cancer is thought to be immunogenic, making it amenable to immunotherapy. However, immunotherapies such as PD-L1 inhibitors and T cell transfers have produced modest, if any, survival benefit. One particular immunotherapy of interest is the dendritic cell vaccine, which delivers mature dendritic cells loaded with tumor antigens with the goal of mounting a T cell response against tumor cells. This review will focus on the role that dendritic cells play in the ovarian tumor microenvironment, general approaches to engineering dendritic cell vaccines and assessing their efficacy alone and in combination with other immunotherapies and systemic chemotherapies. Finally, we will discuss important areas of ongoing research in the field, including the development of personalized neoantigen-targeting DC vaccines.
INTRODUCTION
Ovarian cancer is the most lethal gynecological cancer, with an overall 5-year survival of 48% in 2020. Of those that present at an advanced stage, the 5-year survival is just 29% (1). The current standard treatment of epithelial ovarian cancer (EOC) is debulking surgery followed by platinum-based chemotherapy. Despite good initial responses in most patients, chemoresistance and relapse are common (2, 3). For patients with resistance to platinum-based therapies, their treatment options remain limited.
Ovarian cancer is thought to be immunogenic as it expresses multiple well-known tumor-associated antigens (TAA). Some tumors are infiltrated by lymphocytes, which correlates positively with progression-free survival and overall survival (4, 5). These data suggest that ovarian cancer could be amenable to immunotherapy targeting. However, to date, immunotherapies have shown modest, if any, benefit. For example, PD-1 inhibitors have a response rate of 11.5% in advanced metastatic disease, which is thought in part due to poor T cell infiltration as well as poor antigen presenting function of antigen-presenting cells (APCs) (6, 7).
DENDRITIC CELLS IN OVARIAN CANCER
Dendritic cells in the tumor microenvironment take up and process tumor-associated antigens and present them on MHC I/II molecules to activate CD8+ and CD4+ cells, respectively. In comparison to other APCs such as B cells, mononuclear cells and macrophages, DCs are regarded as the most powerful cell type in its ability to capture, process and present antigens (8). In general, different DCs subtypes can play a multitude of roles in the tumor microenvironment. Conventional DCs (cDC) are the main subtype tasked with activating CD8+ T cells (particularly cDC type 1) and differentiation of CD4+ T cells (cDC type 2) through cytokine production (9). Conversely, plasmacytoid DCs (pDC), which are the main subtype of DCs in ovarian cancer, can exert both anti-tumor and immunosuppressive effects. Whether pDCs skew towards being tumor-protective or tumor-suppressive is largely determined by the signals they receive from their tumor microenvironment (9, 10).
Investigations into ovarian cancer have revealed that dendritic cells (DCs) make essential contributions to the depressed immune function observed in the ovarian tumor microenvironment. While ovarian cancer lesions have a high degree of DC infiltration, these DCs can have low efficacy of antigen presentation due to DC tolerance, which is characterized by downregulated expression of costimulatory molecules on the surface of DC cells and weaker antigen-presenting ability (11). Further, DCs can support the immunosuppressive milieu through their interactions with Tregs. For example, DC expression of indoleamine 2,3-deoxygenase, an essential enzyme in amino acid metabolism, can reduce the amount of tryptophan near Tregs and as a result maintain Tregs in an immunosuppressive state through mTORC-Akt signaling (12). DCs have also been shown to activate immunosuppressive Tregs by expressing ICOS ligand, leading to tumor progression (13).
Vaccines of functional DCs loaded with tumor-associated antigens have held promise for expanding tumor-specific T cell populations by restoring antigen presentation to T cells and bypassing the dysregulated milieu of the tumor microenvironment. There are different types of cancer vaccines, including cell-based vaccines, peptide/protein vaccines, epigenetic vaccines and genetic vaccines (14). This review will focus on the two most common DC vaccine types: cell-based and peptide/protein-based vaccines, which are designed to present T cells with tumor-associated antigens. Specifically, we will review the general approaches to engineering these vaccines as well as assessing their efficacy alone and in combination with other immunotherapies and systemic chemotherapies. Finally, we will discuss important areas of ongoing research in the field, including the development on personalized neoantigen-targeting DC vaccines.
ENGINEERING DENDRITIC CELL VACCINES
Dendritic cell vaccines have long been an immunotherapy of interest for ovarian cancer, particularly given the demonstrated dysfunction of DCs surrounding ovarian tumors. Further, these vaccines are generally well-tolerated by patients and can induce long-term immunologic memory (15).
Currently, vaccines targeting DCs ex vivo are produced using three general steps. First, apheresis is performed to obtain either immune cells that have the potential to become DCs, such as monocytes, or immature DCs from peripheral blood. Among all cell types, monocyte-derived DCs (MoDCs) are most often used as immature DCs are typically not found in sufficient quantity in peripheral blood to produce a vaccine. Monocytes are subsequently cultured in vitro with a cytokine cocktail of GM-CSF and IL-4 that induces differentiation into immature DCs (16). However, from a functional standpoint, MoDCs have been shown to be inferior to cDCs in inducing long-lasting immune responses through T-cell activation, raising questions of their appropriateness as the DC subtype used in many EOC vaccines (17).
Second, once immature DCs have been obtained, they are loaded with tumor-associated antigens, ranging from specific peptides to proteins to multiple antigens from whole tumor lysates. To date, the most common approach has been to load DCs with one or several peptides known to be expressed on ovarian cancer cells. One example includes Wilms tumor 1 (WT-1), which is overexpressed in ovarian cancer along with many other solid tumors and can be targeted by cytotoxic T cells (CTLs). One group incubated DCs with an MHCI-restricted WT-1 peptide and a streptococcal primer and showed that these DCs elicited a CTL effect (18). This has been repeated with other peptides expressed by ovarian cancer cells, such as Her-2/neu, epithelial mucin 1, and p53. These vaccines reproducibly generated antigen-specific IFN-y secreting T cells (19, 20). However, the success of these single peptide/protein vaccines has been limited, resulting in short-term disease stabilization that ultimately gives way to progression after several months. One possible theory for this is that when a vaccine target is a non-mutated self-antigen or shared antigen that is overexpressed in the tumor, vaccine efficacy can be low because T cell recognition of self-antigens will be limited by central tolerance (21).
More recently, whole tumor cell lysates have been investigated as an antigen source for DC vaccines. In this scheme, DCs are pulsed with lysed ovarian tumor cells. These cells can be derived from ovarian cancer cell lines or even from a patient’s own tissue sample (22). This has the benefit over single peptide vaccines in that lysates can elicit responses to more than one neoantigen, thus reducing avenues for tumor escape. Further, in the case of an autologous tumor cell lysate, the patient can produce a more “personalized” tumor-specific T cell pool by targeting their own unique set of tumor-associated antigens (23, 24). Previous studies with DC vaccines loaded with whole tumor lysate have demonstrated clinical benefit for patients with non-Hodgkin’s lymphoma and melanoma (25, 26). There are multiple approaches to stimulating cell death to induce antigen release, including repetitive freeze-thaw cycles or exposing cells to hypochlorous acid (HOCl). Chiang et al. found that autologous ovarian tumor cells killed with oxidation and lysed with freeze-thaw cycles were superior to cells killed with irradiation or freeze-thaw lysis in priming T cell responses in vitro (27).
Finally, regardless of the antigen, immature antigen-presenting DCs are then matured in the presence of immunogenic substances like LPS and IFN-y to trigger expression of co-stimulatory molecules on the DC surface. These co-stimulatory molecules are essential for T cell activation upon antigen presentation in the lymph node. Once this step is complete, DCs are typically fractionated into multiple doses to be used as serial vaccines over a defined treatment period. Typically, vaccines are given intranodally, but can also be given with intramuscular or subcutaneous injection (16).
PERSONALIZED NEOANTIGEN-TARGETING VACCINES – THE NEXT BIG LEAP?
One key area of interest for DC vaccines is the development of neoantigen-targeted vaccines. Neoantigens are proteins expressed by tumors that differ from other tumor-associated antigens in several keys. First, neoantigens arise from DNA mutations within the tumor and thus produce peptides that are specific to tumor cells. Because these antigens are not expressed on other cell types in the body, they are often highly immunogenic in comparison to other tumor-associated antigens, which are typically non-mutated self-antigens that are simply overexpressed on cancer cells. This increased immunogenicity is in part driven by increased affinity for major histocompatibility complexes (MHCs) (31). Further, because neoantigens are only expressed by tumor cells, this limits potential off-target effects. Thus, neoantigens are ideal targets for an anti-tumor T cell response (31). While ovarian cancers have lower mutational burdens than most other cancer types, recent analyses have shown that some patients can express moderate to high levels of neoantigens (32). In other cancers, neoantigen-loaded DC vaccines have shown promising results in small phase I trials in melanoma patients and non-small cell lung cancers (33, 34).
Neoantigens can be classified as being shared or personalized. Shared neoantigens are common in some tumor types and can be used to broadly treat patients who have the same tumor type. However, not all patients express these shared neoantigens, and even if they are expressed, different patients may mount different immune responses to them (35). Thus, personalized neoantigens, which are specific to individual patients and tumors, have become a point of interest for DC vaccine design across all cancer types (22). However, identifying a patient’s neoantigen repertoire has only recently become possible with the development of next-generation sequencing (NGS) such as whole exome sequencing, mass spectrometry analysis of the immunopeptidome (i.e., peptides associated with HLAs), as well as highly predictive bioinformatics tools (36-38).
Initial evidence suggests that recognition and targeting of tumor-specific neoantigens improves the effectiveness of dendritic cell vaccines in ovarian cancers. For example, patients with tumor lysate-pulsed DCs were found to have activated high-avidity CD8+ T cell clonal expansion specific for de novo neoantigens, which improved progression-free survival compared to patients without neoantigen-specific T cell responses (29).
However, multiple research groups are still investigating the true benefit of this approach compared to using overexpressed self-antigens or whole tumor lysates, particularly because the process of identifying a patient’s neoantigens remains costly and resource-intensive (22). Currently, one trial is underway to assess a personalized neoantigen-pulsed DC vaccine in ovarian cancer patients. This trial will investigate the feasibility and safety of a personalized neoantigen-loaded DC vaccine in patients with ovarian cancer (ClinicalTrials.gov NCT04024878).
CONCLUSIONS
Dendritic cell vaccines have been shown to be an effective immunotherapy for ovarian cancer, however it remains an active area of innovation in the wake of new technological advances in the realms of NGS and bioinformatics. Further, combinations of various immunomodulatory treatments with DC vaccines will likely be required to capitalize on the immunogenicity of ovarian malignancies and will be a crucial area of clinical investigation going forward.
DISCLOSURES
Funding: Not applicable.
Conflicts of interest: None.
Availability of data and materials: Not applicable.
Code availability: Not applicable.
Ethics approval: Not applicable.
Consent to participate: Not applicable.
REFERENCES
1. Siegel, R.L., Miller, K.D., Jemal, A. (2020). Cancer statistics. CA: A Cancer Journal for Clinicians, 70(1), 7-30. doi:10.3322/CAAC.21590
2. Gadducci, A., Cosio, S., Conte, P.F., Genazzani, A.R. (2005). Consolidation and maintenance treatments for patients with advanced epithelial ovarian cancer in complete response after first-line chemotherapy: a review of the literature. Critical reviews in oncology/hematology, 55(2), 153-166. doi:10.1016/J.CRITREVONC.2005.03.003
3. Stuart, G.C.E. (2003). First-line treatment regimens and the role of consolidation therapy in advanced ovarian cancer. Gynecologic oncology, 90(3 Pt 2), S8. doi:10.1016/S0090-8258(03)00472-4
4. Vonderheide, R.H., Hahn, W.C., Schultze, J.L., Nadler, L.M. (1999). The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes. Immunity, 10(6), 673-679. doi: 10.1016/s1074-7613(00)80066-7
5. Sato, E., Olson, S.H., Ahn, J., et al. (2005). Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proceedings of the National Academy of Sciences of the United States of America, 102(51), 18538-43. doi: 10.1073/pnas.0509182102
6. Kandalaft, L.E., Odunsi, K., Coukos, G. (2019). Immunotherapy in ovarian cancer: Are we there yet? Journal of Clinical Oncology, 37(27), 2460-2471. doi:10.1200/JCO.19.00508
7. Varga, A., Piha-Paul, S., Ott, P.A., et al. (2019). Pembrolizumab in patients with programmed death ligand 1–positive advanced ovarian cancer: Analysis of KEYNOTE-028. Gynecologic Oncology, 152(2), 243-250. doi:10.1016/J.YGYNO.2018.11.017
8. Wculek, S.K., Cueto, F.J., Mujal, A.M., Melero, I., Krummel, M.F., Sancho, D. (2020). Dendritic cells in cancer immunology and immunotherapy. Nature Reviews Immunology, 20(1), 7-24. doi:10.1038/S41577-019-0210-Z
9. Labidi-Galy, S.I., Sisirak, V., Meeus, P., et al. (2011). Quantitative and functional alterations of plasmacytoid dendritic cells contribute to immune tolerance in ovarian cancer. Cancer Research, 71(16), 5423-5434. doi:10.1158/0008-5472.CAN-11-0367
10. Labidi-Galy, S.I., Treilleux, I., Goddard-Leon, S., et al. (2012). Plasmacytoid dendritic cells infiltrating ovarian cancer are associated with poor prognosis. OncoImmunology, 1(3), 380-382. doi:10.4161/ONCI.18801
11. Harimoto, H., Shimizu, M., Nakagawa, Y., et al. (2013). Inactivation of tumor-specific CD8+CTLs by tumor-infiltrating tolerogenic dendritic cells. Immunology and Cell Biology, 91(9), 545-555. doi:10.1038/ICB.2013.38
12. Munn, D.H. and Mellor, A.L. (2016). IDO in the Tumor Microenvironment: Inflammation, Counter-Regulation, and Tolerance. Trends in Immunology, 37(3), 193-207. doi:10.1016/J.IT.2016.01.002
13. Conrad, C., Gregorio, J., Wang, Y.H., et al. (2012). Plasmacytoid dendritic cells promote immunosuppression in ovarian cancer via ICOS costimulation of Foxp3+ T-regulatory cells. Cancer Research, 72(20), 5240-5249. doi:10.1158/0008-5472.CAN-12-2271
14. Lluesma, S.M., Wolfer, A., Harari, A., Kandalaft, L.E. (2016). Cancer Vaccines in Ovarian Cancer: How Can We Improve? Biomedicines, 4(2). doi:10.3390/BIOMEDICINES4020010
15. Bol, K.F., Schreibelt, G., Gerritsen, W.R., de Vries, I.J.M., Figdor, C.G. (2016). Dendritic cell-based immunotherapy: state of the art and beyond. Clin Cancer Res, 22(8), 1897-1906. doi:10.1158/1078-0432.ccr-15-1399
16. Zhang, X., He, T., Li, Y., et al. (2021). Dendritic Cell Vaccines in Ovarian Cancer. Frontiers in Immunology, 11, 613773. doi:10.3389/FIMMU.2020.613773/BIBTEX
17. Zhou, Y., Slone, N., Chrisikos, T.T., et al. (2020). Vaccine efficacy against primary and metastatic cancer with in vitro-generated CD103 + conventional dendritic cells. Journal for ImmunoTherapy of Cancer, 8(1). doi:10.1136/JITC-2019-000474
18. Zhang, W., Lu, X., Cui, P., et al. (2019). Phase I/II clinical trial of a Wilms’ tumor 1-targeted dendritic cell vaccination-based immunotherapy in patients with advanced cancer. Cancer Immunology, Immunotherapy, 68(1), 121-130. doi:10.1007/S00262-018-2257-2
19. Chu, C.S., Boyer, J., Schullery, D.S., et al. (2012). Phase I/II randomized trial of dendritic cell vaccination with or without cyclophosphamide for consolidation therapy of advanced ovarian cancer in first or second remission. Cancer Immunology, Immunotherapy, 61(5), 629-641. doi:10.1007/S00262-011-1081-8
20. Rahma, O.E., Ashtar, E., Czystowska, M., et al. (2012). A gynecologic oncology group phase II trial of two p53 peptide vaccine approaches: Subcutaneous injection and intravenous pulsed dendritic cells in high recurrence risk ovarian cancer patients. Cancer Immunology, Immunotherapy, 61(3), 373-384. doi:10.1007/S00262-011-1100-9
21. Schietinger, A., Philip, M., Schreiber, H. (2008). Specificity in cancer immunotherapy. Seminars in Immunology, 20(5), 276-285. doi:10.1016/j.smim.2008.07.001
22. Tang, L., Zhang, R., Zhang, X., Yang, L. (2021). Personalized Neoantigen-Pulsed DC Vaccines: Advances in Clinical Applications. Frontiers in Oncology, 11, 701777. doi:10.3389/FONC.2021.701777/BIBTEX
23. Hatfield, P., Merrick, A.E., West, E., et al. (2008). Optimization of dendritic cell loading with tumor cell lysates for cancer immunotherapy. Journal of Immunotherapy, 31(7), 620-32. doi: 10.1097/CJI.0b013e31818213df
24. Cho, D., Yang, W., Lee, H., et al. (2012). Adjuvant immunotherapy with whole-cell lysate dendritic cells vaccine for glioblastoma multiforme: a phase II clinical trial. World Neurosurgery, 77(5-6), 736-44. doi: 10.1016/j.wneu.2011.08.020
25. Di Nicola, M., Zappasodi, R., Carmelo, C.S., et al. (2009). Vaccination with autologous tumor-loaded dendritic cells induces clinical and immunologic responses in indolent B-cell lymphoma patients with relapsed and measurable disease: a pilot study. Blood, 113(1), 18-27. doi:10.1182/blood-2008-06-165654
26. Palucka, A.K., Ueno, H., Connolly, J., et al. (2006). Dendritic cells loaded with killed allogeneic melanoma cells can induce objective clinical responses and MART-1 specific CD8 + T-cell immunity. J Immunother, 29(5), 545-557. doi:10.1097/01.cji.0000211309.90621.8b
27. Chiang, C.L.L., Kandalaft, L.E., Tanyi, J., et al. (2013). A dendritic cell vaccine pulsed with autologous hypochlorous acid-oxidized ovarian cancer lysate primes effective broad antitumor immunity: from bench to bedside. Clinical cancer research : an official journal of the American Association for Cancer Research, 19(17), 4801-4815. doi:10.1158/1078-0432.CCR-13-1185
28. Kobayashi, M., Chiba, A., Izawa, H., et al. (2014). The feasibility and clinical effects of dendritic cell-based immunotherapy targeting synthesized peptides for recurrent ovarian cancer. Journal of ovarian research, 7(1). doi:10.1186/1757-2215-7-48
29. Tanyi, J.L., Bobisse, S., Ophir, E., et al. (2018). Personalized cancer vaccine effectively mobilizes antitumor T cell immunity in ovarian cancer. Science translational medicine, 10(436). doi:10.1126/SCITRANSLMED.AAO5931
30. Kandalaft, L.E., Powell, D.J., Chiang, C.L., et al. (2013). Autologous lysate-pulsed dendritic cell vaccination followed by adoptive transfer of vaccine-primed ex vivo co-stimulated t cells in recurrent ovarian cancer. OncoImmunology, 2(1). doi:10.4161/ONCI.22664
31. Lawrence, M.S., Stojanov, P., Polak, P., et al. (2013). Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature, 499(7457), 214-218. doi:10.1038/nature12213
32. O’Donnell, T., Christie, E.L., Ahuja, A., et al. (2018). Chemotherapy weakly contributes to predicted neoantigen expression in ovarian cancer. BMC Cancer, 18(1), 87. doi:10.1186/s12885-017-3825-0
33. Ding, Z., Li, Q., Zhang, R., et al. (2021). Personalized neoantigen pulsed dendritic cell vaccine for advanced lung cancer. Signal Transduction and Targeted Therapy, 6(1). doi:10.1038/S41392-020-00448-5
34. Carreno, B.M., Magrini, V., Becker-Hapak, M., et al. (2015). A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science, 348(6236), 803-808. doi:10.1126/SCIENCE.AAA3828
35. Mullard, A. (2016). The cancer vaccine resurgence. Nature Reviews Drug Discovery, 15(10), 663-665. doi:10.1038/NRD.2016.201
36. Boisguérin, V., Castle, J.C., Loewer, M., et al. (2014). Translation of genomics-guided RNA-based personalised cancer vaccines: towards the bedside. British Journal of Cancer, 111(8), 1469-1475. doi:10.1038/bjc.2013.820
37. Bassani-Sternberg, M., Gfeller, D. (2016). Unsupervised HLA peptidome deconvolution improves ligand prediction accuracy and predicts cooperative effects in peptide-HLA interactions. Journal of Immunology, 197(6), 2492-2499. doi:10.4049/jimmunol.1600808
38. Gfeller, D., Guillaume, P., Michaux, J., et al. (2018). The length distribution and multiple specificity of naturally presented HLA-I ligands. Journal of Immunology, 201(12), 3705-3716. doi:10.4049/jimmunol.1800914