Tumor Vaccines: Unleashing the Power of the Immune System to Fight Cancer
Abstract
:1. Introduction
2. Tumor Microenvironment
3. Oncology Vaccines
3.1. Cancer Immunology
3.2. Mechanism of Action
3.2.1. Cellular Immunity
3.2.2. Humoral Immunity
- Antibody-dependent cell-mediated cytotoxicity (ADCC) (Figure 3A(1–3)): At the first stage, effector cell recruitment: Fc-receptor-bearing immune cells, like natural killer (NK) cells, macrophages, and neutrophils, are drawn to the tumor site via interactions between their Fc receptors and the Fc segment of the antibody bound to the malignant cell [86,87]. At the next stage, formation of immunological synapses, degranulation, and release of cytotoxic molecules: Immunological synapses are formed when Fc receptors on effector cells interact with the Fc portion of cancer-bound antibodies, enabling the targeted release of cytotoxic molecules [88]. Effector cell degranulation ensues, culminating in cancer cell elimination via perforin and granzymes [89]. Finally, phagocytosis: Post-apoptosis, macrophages engulf the cancerous debris, facilitating clearance and preventing the dissemination of harmful cellular components [90].
- Complement-dependent cytotoxicity (CDC) (Figure 3B(1–4)): At the first stage, complement activation and cascade: Bound antibodies’ Fc portions engage C1q (complement component 1, q subcomponent), instigating the classical complement pathway and the formation of the C3 convertase enzyme complex [91,92]. At the next stage, membrane attack complex (MAC) assembly: Complement protein cleavage and activation yield the MAC, comprising C5b, C6, C7, C8, and C9 [93]. After that, cell lysis: MACs perforate cancer cell membranes, causing uncontrolled ion and water movement, cellular lysis, and death [93]. Finally, opsonization and phagocytosis: Cancer cells are targeted for destruction by phagocytes when complement activation promotes opsonization [92].
- Neutralization of growth factors and inhibition of signaling pathways: Antibodies obstruct tumor cell proliferation-promoting growth factors and impede signaling pathways that are crucial for cancer cell survival and invasion [15].
4. Types and Characteristics of Cancer Vaccines
4.1. Peptide Vaccines
4.2. DNA/RNA-Based Vaccines
4.3. Viral-Vector-Based Vaccines
4.4. Dendritic-Cell-Based Vaccines
4.5. Whole-Cell-Based Vaccines
Types of Tumor Vaccines | Strengths | Weaknesses | Examples | Mechanisms of Action | Effects | Limitations | References |
---|---|---|---|---|---|---|---|
Peptide vaccines |
|
| Nelipepimut-S (NeuVax) | HER2-derived peptide vaccine | Activation of T-cell response | Limited overall survival improvement | [166] |
CIMAvax-EGF | EGF-based peptide vaccine | Inhibition of EGF signaling | No direct tumor targeting | [103] | |||
MUC1-based peptide vaccine | Targeting MUC1 tumor-associated antigens | Enhanced immune response | Heterogeneous patient response | [167] | |||
DNA/RNA-based vaccines |
|
| CV9104 (CureVac) | Uses mRNA to encode six antigens overexpressed in prostate cancer | Induced antigen-specific immune responses in early clinical trials | Efficacy in late-stage trials yet to be established; possibility of inducing autoimmune responses | [168] |
Viral-vector-based vaccines |
|
| Adenovirus-based vaccines (OncoVEXGM-CSF, CG0070) | Adenoviruses are modified to express a tumor-specific antigen or an immunomodulatory molecule; these stimulate an immune response against the tumor | Effective in stimulating an immune response against the tumor | Immune response to the viral vector can limit repeat dosing | [169] |
Lentivirus-based vaccines (LV305) | Lentiviruses are engineered to deliver tumor-specific antigens to dendritic cells to stimulate a T-cell response | Successful in initiating T-cell responses | Safety concerns over integration into the host genome | [23] | |||
Vaccinia-virus-based vaccines (JX-594) | Vaccinia viruses are genetically engineered to express a tumor antigen and/or immunostimulatory molecule; they can directly lyse cancer cells | Showed antitumor activity and were well tolerated in clinical trials | Immune response to the viral vector can limit its effectiveness | [170] | |||
Dendritic-cell-based vaccines |
|
| Provenge (Sipuleucel-T) | The patient’s own dendritic cells are exposed to a fusion protein (prostatic acid phosphatase linked to an immune cell stimulating factor) | Extended overall survival in metastatic castration-resistant prostate cancer | Limited clinical benefits, high cost, and complex manufacturing process | [5] |
DCVax-L | Autologous dendritic cells are pulsed with tumor lysate | Prolonged progression-free survival in glioblastoma multiforme (GBM) patients | Not FDA-approved; requires personalized manufacturing | [171] | |||
Whole-cell-based vaccines |
|
| GVAX | Utilizes autologous/allogeneic tumor cells that have been genetically modified to secrete the immune-stimulating cytokine GM-CSF | Demonstrated a significant immune response against cancer, studied in various types of cancer, including pancreatic and prostate cancers | Production can be labor-intensive and personalized; often requires co-administration with adjuvants or other immunomodulatory agents to enhance their immunogenicity | [159,172] |
Canvaxin | Allogeneic melanoma cells mixed with Bacillus Calmette–Guérin (BCG) to stimulate immune response | Intended for melanoma treatment, but development discontinued due to insufficient effectiveness | Limited efficacy; potential for BCG-related side effects | [173] | |||
Oncophage (Vitespen) | Uses heat shock proteins (gp96) derived from the patient’s tumor as an autologous vaccine | Showed efficacy in extending disease-free survival in certain patients with kidney cancer and melanoma | Not universally effective; personalized manufacturing can be labor-intensive | [174,175] |
4.6. Another Cancer Vaccine Therapy: In Situ Cancer Vaccines
4.7. Influencing Factors of Tumor Vaccines
5. Combination Therapies
5.1. Cancer Vaccine + Immune Checkpoint Inhibitors
5.2. Cancer Vaccine + Chemotherapy
5.3. Cancer Vaccine + Radiotherapy
5.4. Cancer Vaccine + Targeted Therapy
5.5. Cancer Vaccine + Oncolytic Virotherapy
6. Personalized Cancer Vaccines
6.1. Tumor Sample Collection
6.2. Sequencing and Analysis
6.3. Neoantigen Selection
6.4. Vaccine Design
6.5. Vaccine Production
6.6. Vaccine Administration
6.7. Immune Response Activation
6.8. Monitoring and Evaluation
6.9. Follow-Up, Maintenance, and Patient Education and Support
6.10. Data Collection and Analysis
6.11. Integration with Other Therapies
6.12. Expanding Applications
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
ADCC | Antibody-dependent cell-mediated cytotoxicity |
AML | Acute myeloid leukemia |
APC | Antigen-presenting cell |
BCRs | B-cell receptors |
CAR | Chimeric antigen receptor |
CAR-Ts | Chimeric antigen receptor T cells |
CDC | Complement-dependent cytotoxicity |
CTLA-4 | Cytotoxic T-lymphocyte-associated protein 4 |
CTLs | Cytotoxic T cells |
DAMPs | Damage-associated molecular patterns |
DCs | Dendritic cells |
ECM | Extracellular matrix |
EGF | Epidermal growth factor |
FAK | Focal adhesion kinase |
FDA | Food and Drug Administration |
GBM | Glioblastoma multiforme |
ICIs | Immune checkpoint inhibitors |
IFN-γ | Interferon-γ |
IL | Interleukin |
LNPs | Lipid nanoparticles |
LOX | Lysyl oxidase |
M1 | Classically activated macrophages |
M2 | Alternatively activated macrophages |
MAC | Membrane attack complex |
mCRPC | Metastatic castration-resistant prostate cancer |
M-CSF | Macrophage colony-stimulating factor |
MDSCs | Myeloid-derived suppressor cells |
MHC | Major histocompatibility complex |
MM | Multiple myeloma |
MMPs | Matrix metalloproteinases |
NGS | Next-generation sequencing |
NK | Natural killer cells |
NSCLC | Non-small-cell lung cancer |
PAMPs | Pathogen-associated molecular patterns |
PBMCs | Peripheral blood mononuclear cells |
PD-1 | Programmed cell death protein 1 |
PD-L1 | Programmed cell death ligand 1 |
TAAs | Tumor-associated antigens |
TAMs | Tumor-associated macrophages |
TCR | T-cell receptor |
TGF-β | Transforming growth factor-β |
Th | Helper T cells |
TLR | Toll-like receptor |
TME | Tumor microenvironment |
TNF-α | Tumor necrosis factor-α |
Tregs | Regulatory T cells |
TSAs | Tumor-specific antigens |
VEGF | Vascular endothelial growth factor |
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Mellman, I.; Coukos, G.; Dranoff, G. Cancer immunotherapy comes of age. Nature 2011, 480, 480–489. [Google Scholar] [CrossRef] [PubMed]
- Wiemann, B.; Starnes, C.O. Coley’s toxins, tumor necrosis factor and cancer research: A historical perspective. Pharmacol. Ther. 1994, 64, 529–564. [Google Scholar] [CrossRef] [PubMed]
- Burnet, M. Cancer-A Biological Approach: III Viruses Associated with Neoplastic Conditions. Br. Med. J. 1957, 1, 841. [Google Scholar] [CrossRef]
- Kantoff, P.W.; Higano, C.S.; Shore, N.; Berger, E.; Small, E.J. Sipuleucel T immunotherapy for CRPC. N. Engl. J. 2010, 363, 411–422. [Google Scholar]
- Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef]
- Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef]
- Sahin, U.; Türeci, Ö. Personalized vaccines for cancer immunotherapy. Science 2018, 359, 1355–1360. [Google Scholar] [CrossRef]
- Malonis, R.J.; Lai, J.R.; Vergnolle, O. Peptide-Based Vaccines: Current Progress and Future Challenges. Chem. Rev. 2020, 120, 3210–3229. [Google Scholar] [CrossRef]
- Saade, F.; Petrovsky, N. Technologies for enhanced efficacy of DNA vaccines. Expert Rev. Vaccines 2012, 11, 189–209. [Google Scholar] [CrossRef]
- Pardi, N.; Hogan, M.J.; Porter, F.W.; Weissman, D. mRNA vaccines-a new era in vaccinology. Nat. Rev. Drug Discov. 2018, 17, 261–279. [Google Scholar] [CrossRef] [PubMed]
- Cecilia Larocca, B.S.; Schlom, J. Viral Vector Y Based Therapeutic Cancer Vaccines. Cancer J. 2011, 17, 359–371. [Google Scholar] [CrossRef] [PubMed]
- Bubenik, J. Dendritic cell-based cancer vaccines. Folia Biol. 1999, 45, 71–74. [Google Scholar]
- De Gruijl, T.D.; Van Den Eertwegh, A.J.M.; Pinedo, H.M.; Scheper, R.J. Whole-cell cancer vaccination: From autologous to allogeneic tumor- and dendritic cell-based vaccines. Cancer Immunol. Immunother. 2008, 57, 1569–1577. [Google Scholar] [CrossRef]
- Melero, I.; Gaudernack, G.; Gerritsen, W.; Huber, C.; Parmiani, G.; Scholl, S.; Thatcher, N.; Wagstaff, J.; Zielinski, C.; Faulkner, I.; et al. Therapeutic vaccines for cancer: An overview of clinical trials. Nat. Rev. Clin. Oncol. 2014, 11, 509–524. [Google Scholar] [CrossRef]
- Galluzzi, L.; Buqué, A.; Kepp, O.; Zitvogel, L.; Kroemer, G. Immunological Effects of Conventional Chemotherapy and Targeted Anticancer Agents. Cancer Cell 2015, 28, 690–714. [Google Scholar] [CrossRef]
- Sharabi, A.B.; Lim, M.; DeWeese, T.L.; Drake, C.G. Radiation and checkpoint blockade immunotherapy: Radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 2015, 16, e498–e509. [Google Scholar] [CrossRef]
- Palucka, K.; Banchereau, J. Cancer immunotherapy via dendritic cells. Nat. Rev. Cancer 2012, 12, 265–277. [Google Scholar] [CrossRef]
- Russell, S.J.; Peng, K.W.; Bell, J.C. Oncolytic virotherapy. Nat. Biotechnol. 2012, 30, 658–670. [Google Scholar] [CrossRef]
- Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74. [Google Scholar] [CrossRef]
- Ott, P.A.; Hu, Z.; Keskin, D.B.; Shukla, S.A.; Sun, J.; Bozym, D.J.; Zhang, W.; Luoma, A.; Giobbie-Hurder, A.; Peter, L.; et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017, 547, 217–221. [Google Scholar] [CrossRef] [PubMed]
- Keskin, D.B.; Anandappa, A.J.; Sun, J.; Tirosh, I.; Mathewson, N.D.; Li, S.; Oliveira, G.; Giobbie-Hurder, A.; Felt, K.; Gjini, E.; et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature 2019, 565, 234–239. [Google Scholar] [CrossRef] [PubMed]
- Kranz, L.M.; Diken, M.; Haas, H.; Kreiter, S.; Loquai, C.; Reuter, K.C.; Meng, M.; Fritz, D.; Vascotto, F.; Hefesha, H.; et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 2016, 534, 396–401. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
- Balkwill, F.R.; Capasso, M.; Hagemann, T. The tumor microenvironment at a glance. J. Cell Sci. 2012, 125, 5591–5596. [Google Scholar] [CrossRef] [PubMed]
- Naci, D.; El Azreq, M.A.; Chetoui, N.; Lauden, L.; Sigaux, F.; Charron, D.; Al-Daccak, R.; Aoudjit, F. α2β1 integrin promotes chemoresistance against doxorubicin in cancer cells through extracellular signal-regulated kinase (ERK). J. Biol. Chem. 2012, 287, 17065–17076. [Google Scholar] [CrossRef]
- Joyce, J.A.; Pollard, J.W. Microenvironmental regulation of metastasis. Nat. Rev. Cancer 2009, 9, 239–252. [Google Scholar] [CrossRef]
- Laczko, R.; Szauter, K.M.; Jansen, M.K.; Hollosi, P.; Muranyi, M.; Molnar, J.; Fong, K.S.K.; Hinek, A.; Csiszar, K. Active lysyl oxidase (LOX) correlates with focal adhesion kinase (FAK)/paxillin activation and migration in invasive astrocytes. Neuropathol. Appl. Neurobiol. 2007, 33, 631–643. [Google Scholar] [CrossRef]
- Caon, I.; Bartolini, B.; Parnigoni, A.; Caravà, E.; Moretto, P.; Viola, M.; Karousou, E.; Vigetti, D.; Passi, A. Revisiting the hallmarks of cancer: The role of hyaluronan. Semin. Cancer Biol. 2019, 62, 9–16. [Google Scholar] [CrossRef]
- Jabłońska-Trypuć, A.; Matejczyk, M.; Rosochacki, S. Matrix metalloproteinases (MMPs), the main extracellular matrix (ECM) enzymes in collagen degradation, as a target for anticancer drugs. J. Enzym. Inhib. Med. Chem. 2016, 31, 177–183. [Google Scholar] [CrossRef]
- Niland, S.; Riscanevo, A.X.; Eble, J.A. Matrix metalloproteinases shape the tumor microenvironment in cancer progression. Int. J. Mol. Sci. 2022, 23, 146. [Google Scholar] [CrossRef] [PubMed]
- Lepucki, A.; Orlińska, K.; Mielczarek-Palacz, A.; Kabut, J.; Olczyk, P.; Komosińska-Vassev, K. The Role of Extracellular Matrix Proteins in Breast Cancer. J. Clin. Med. 2022, 11, 1250. [Google Scholar] [CrossRef]
- Azimzade, Y.; Saberi, A.A.; Sahimi, M. Regulation of migration of chemotactic tumor cells by the spatial distribution of collagen fiber orientation. Phys. Rev. E 2019, 99, 062414. [Google Scholar] [CrossRef]
- Carmeliet, P.; Jain, R.K. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011, 473, 298–307. [Google Scholar] [CrossRef] [PubMed]
- Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Associated with anti-PD-1 Response. Nature 2019, 560, 382–386. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Yu, R.; Cai, T.; Chen, Z.; Lan, M.; Zou, T.; Wang, B.; Wang, Q.; Zhao, Y.; Cai, Y. Effects of immune cells and cytokines on inflammation and immunosuppression in the tumor microenvironment. Int. Immunopharmacol. 2020, 88, 106939. [Google Scholar] [CrossRef]
- Multhoff, G.; Vaupel, P. Hypoxia compromises anti-cancer immune responses. Adv. Exp. Med. Biol. 2020, 1232, 131–143. [Google Scholar] [PubMed]
- Boedtkjer, E.; Pedersen, S.F. The Acidic Tumor Microenvironment as a Driver of Cancer. Annu. Rev. Physiol. 2020, 82, 103–126. [Google Scholar] [CrossRef]
- Roman-Trufero, M.; Auner, H.W.; Edwards, C.M. Multiple myeloma metabolism—A treasure trove of therapeutic targets? Front. Immunol. 2022, 13, 897862. [Google Scholar] [CrossRef]
- Gopalakrishnan, V.; Helmink, B.A.; Spencer, C.N.; Reuben, A.; Wargo, J.A. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell. 2018, 33, 570–580. [Google Scholar] [CrossRef]
- Li, W.; Deng, Y.; Chu, Q.; Zhang, P. Gut microbiome and cancer immunotherapy. Cancer Lett. 2019, 447, 41–47. [Google Scholar] [CrossRef] [PubMed]
- Gajewski, T.F.; Schreiber, H.; Fu, Y.X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef] [PubMed]
- Fridman, W.H.; Pagès, F.; Saut̀s-Fridman, C.; Galon, J. The immune contexture in human tumours: Impact on clinical outcome. Nat. Rev. Cancer 2012, 12, 298–306. [Google Scholar] [CrossRef]
- Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef]
- Mantovani, A.; Sozzani, S.; Locati, M.; Allavena, P.; Sica, A. Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002, 23, 549–555. [Google Scholar] [CrossRef] [PubMed]
- Koelwyn, G.J.; Quail, D.F.; Zhang, X.; White, R.M.; Jones, L.W. Exercise-dependent regulation of the tumour microenvironment. Nat. Rev. Cancer 2017, 17, 620–632. [Google Scholar] [CrossRef]
- Gordon, S. Alternative activation of macrophages. Nat. Rev. Immunol. 2003, 3, 23–35. [Google Scholar] [CrossRef]
- Pickup, M.; Novitskiy, S.; Moses, H.L. The roles of TGFβ in the tumour microenvironment. Nat. Rev. Cancer 2013, 13, 788–799. [Google Scholar] [CrossRef]
- Schroder, K.; Hertzog, P.J.; Ravasi, T.; Hume, D.A. Interferon-γ: An overview of signals, mechanisms and functions. J. Leukoc Biol. 2004, 75, 163–189. [Google Scholar] [CrossRef]
- Rosenberg, S.A. IL-2: The First Effective Immunotherapy for Human Cancer. J. Immunol. 2014, 192, 5451–5458. [Google Scholar] [CrossRef]
- Trinchieri, G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat. Rev. Immunol. 2003, 3, 133–146. [Google Scholar] [CrossRef]
- Kim, H.J.; Cantor, H. CD4 T-cell subsets and tumor immunity: The helpful and the not-so-helpful. Cancer Immunol. Res. 2014, 2, 91–98. [Google Scholar] [CrossRef]
- David, C.J.; Massagué, J. Contextual determinants of TGFβ action in development, immunity and cancer. Nat. Rev. Mol. Cell Biol. 2018, 19, 419–435. [Google Scholar] [CrossRef]
- Johnson, D.E.; O’Keefe, R.A.; Grandis, J.R. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat. Rev. Clin. Oncol. 2018, 15, 234–248. [Google Scholar] [CrossRef]
- De Streel, G.; Lucas, S. Targeting immunosuppression by TGF-β1 for cancer immunotherapy. Biochem. Pharmacol. 2021, 192, 114697. [Google Scholar] [CrossRef] [PubMed]
- Cattin, S.; Fellay, B.; Pradervand, S.; Trojan, A.; Ruhstaller, T.; Rüegg, C.; Fürstenberger, G. Bevacizumab specifically decreases elevated levels of circulating KIT+CD11b+ cells and IL-10 in metastatic breast cancer patients. Oncotarget 2016, 7, 11137–11150. [Google Scholar] [CrossRef] [PubMed]
- Bolandi, S.M.; Abdolmaleki, Z.; Assarehzadegan, M.A. Bevacizumab regulates inflammatory cytokines and inhibits VEGFR2 signaling pathway in an ovalbumin-induced rat model of airway hypersensitivity. Inflammopharmacology 2021, 29, 683–694. [Google Scholar] [CrossRef] [PubMed]
- Arenas-Ramirez, N.; Woytschak, J.; Boyman, O. Interleukin-2: Biology, Design and Application. Trends. Immunol. 2015, 36, 763–777. [Google Scholar] [CrossRef]
- Prochazkova, J.; Pokorna, K.; Holan, V. IL-12 inhibits the TGF-β-dependent T cell developmental programs and skews the TGF-β-induced differentiation into a Th1-like direction. Immunobiology 2012, 217, 74–82. [Google Scholar] [CrossRef]
- Kwok, G.; Yau, T.C.C.; Chiu, J.W.; Tse, E.; Kwong, Y.L. Pembrolizumab (Keytruda). Hum. Vaccines Immunother. 2016, 12, 2777–2789. [Google Scholar] [CrossRef]
- Lei, X.; Lei, Y.; Li, J.-K.; Du, W.-X.; Li, R.-G.; Yang, J.; Li, J.; Li, F.; Tan, H.-B. Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy. Cancer Lett. 2020, 470, 126–133. [Google Scholar] [CrossRef] [PubMed]
- Jou, J.; Harrington, K.J.; Zocca, M.B.; Ehrnrooth, E.; Cohen, E.E.W. The changing landscape of therapeutic cancer vaccines-novel platforms and neoantigen identification. Clin. Cancer Res. 2021, 27, 689–703. [Google Scholar] [CrossRef] [PubMed]
- Saxena, M.; van der Burg, S.H.; Melief, C.J.M.; Bhardwaj, N. Therapeutic cancer vaccines. Nat. Rev. Cancer 2021, 21, 360–378. [Google Scholar] [CrossRef]
- Thomas, S.; Prendergast, G.C. Cancer vaccines: A brief overview. Methods Mol. Biol. 2016, 1403, 755–761. [Google Scholar] [PubMed]
- Jiang, W.; He, Y.; He, W.; Wu, G.; Zhou, X.; Sheng, Q.; Zhong, W.; Lu, Y.; Ding, Y.; Lu, Q.; et al. Exhausted CD8+ T Cells in the Tumor Immune Microenvironment: New Pathways to Therapy. Front. Immunol. 2021, 11, 622509. [Google Scholar] [CrossRef] [PubMed]
- Swain, S.L.; McKinstry, K.K.; Strutt, T.M. Expanding roles for CD4+ T cells in immunity to viruses. Nat. Rev. Immunol. 2012, 12, 136–148. [Google Scholar] [CrossRef]
- Sallusto, F.; Lanzavecchia, A.; Araki, K.; Ahmed, R. From vaccines to memory and back. Immunity 2010, 33, 451–463. [Google Scholar] [CrossRef]
- Irvine, D.J.; Swartz, M.A.; Szeto, G.L. Engineering synthetic vaccines using cues from natural immunity. Nat. Mater. 2013, 12, 978–990. [Google Scholar] [CrossRef]
- Pulendran, B.; Ahmed, R. Immunological mechanisms of vaccination. Nat. Immunol. 2011, 12, 509–517. [Google Scholar] [CrossRef]
- Weiner, L.M.; Surana, R.; Wang, S. Monoclonal antibodies: Versatile platforms for cancer immunotherapy. Nat. Rev. Immunol. 2010, 10, 317–327. [Google Scholar] [CrossRef]
- Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef] [PubMed]
- Mittal, D.; Gubin, M.M.; Schreiber, R.D.; Smyth, M.J. New insights into cancer immunoediting and its three component phases-elimination, equilibrium and escape. Curr. Opin. Immunol. 2014, 27, 16–25. [Google Scholar] [CrossRef] [PubMed]
- Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.M.C.S.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef] [PubMed]
- Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef]
- Chen, D.S.; Mellman, I. Elements of cancer immunity and the cancer-immune set point. Nature 2017, 541, 321–330. [Google Scholar] [CrossRef]
- Galon, J.; Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 2019, 18, 197–218. [Google Scholar] [CrossRef]
- Galon, J.; Mlecnik, B.; Bindea, G.; Angell, H.K.; Berger, A.; Lagorce, C.; Lugli, A.; Zlobec, I.; Hartmann, A.; Bifulco, C.; et al. Towards the introduction of the “Immunoscore” in the classification of malignant tumours. J. Pathol. 2014, 232, 199–209. [Google Scholar] [CrossRef]
- Topalian, S.L.; Taube, J.M.; Pardoll, D.M. Neoadjuvant checkpoint blockade for cancer immunotherapy. Science 2020, 367, 6477. [Google Scholar] [CrossRef]
- Finn, O.J. Cancer vaccines: Between the idea and the reality. Nat. Rev. Immunol. 2003, 3, 630–641. [Google Scholar] [CrossRef]
- Cella, M.; Engering, A.; Pinet, V.; Pieters, J.; Lanzavecchia, A. Inflammatory stimuli induce accumulation of MHC class II complexes on dendritic cells. Nature 1997, 388, 782–787. [Google Scholar] [CrossRef]
- Chen, L.; Flies, D.B. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat. Rev. Immunol. 2013, 13, 227–242. [Google Scholar] [CrossRef] [PubMed]
- Rosenberg, S.A.; Yang, J.C.; Restifo, N.P. Cancer immunotherapy: Moving beyond current vaccines. Nat. Med. 2004, 10, 909–915. [Google Scholar] [CrossRef]
- Scott, A.M.; Wolchok, J.D.; Old, L.J. Antibody therapy of cancer. Nat. Rev. Cancer 2012, 12, 278–287. [Google Scholar] [CrossRef] [PubMed]
- Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef]
- Schroeder, H.W.; Cavacini, L. Structure and function of immunoglobulins. J. Allergy Clin. Immunol. 2010, 125 (Suppl. 2), S41–S52. [Google Scholar] [CrossRef]
- Golay, J.; Introna, M. Mechanism of action of therapeutic monoclonal antibodies: Promises and pitfalls of in vitro and in vivo assays. Arch. Biochem. Biophys. 2012, 526, 146–153. [Google Scholar] [CrossRef] [PubMed]
- Nimmerjahn, F.; Ravetch, J.V. Fcγ receptors as regulators of immune responses. Nat. Rev. Immunol. 2008, 8, 34–47. [Google Scholar] [CrossRef]
- Orange, J.S. Formation and function of the lytic NK-cell immunological synapse. Nat. Rev. Immunol. 2008, 8, 713–725. [Google Scholar] [CrossRef]
- Bryceson, Y.T.; March, M.E.; Ljunggren, H.G.; Long, E.O. Activation, coactivation, and costimulation of resting human natural killer cells. Immunol. Rev. 2006, 214, 73–91. [Google Scholar] [CrossRef]
- Greenberg, S.; Grinstein, S. Phagocytosis and innate immunity Greenberg and Grinstein 137. Sci. Rep. 2002, 14, 136–145. [Google Scholar]
- Dunkelberger, J.R.; Song, W.C. Complement and its role in innate and adaptive immune responses. Cell Res. 2010, 20, 34–50. [Google Scholar] [CrossRef] [PubMed]
- Ricklin, D.; Hajishengallis, G.; Yang, K.; Lambris, J.D. Complement: A key system for immune surveillance and homeostasis. Nat. Immunol. 2010, 11, 785–797. [Google Scholar] [CrossRef] [PubMed]
- Morgan, B.P. The membrane attack complex as an inflammatory trigger. Immunobiology 2016, 221, 747–751. [Google Scholar] [CrossRef] [PubMed]
- Schumacher, T.; Bunse, L.; Pusch, S.; Sahm, F.; Wiestler, B.; Quandt, J.; Menn, O.; Osswald, M.; Oezen, I.; Ott, M.; et al. A vaccine targeting mutant IDH1 induces antitumour immunity. Nature 2014, 512, 324–327. [Google Scholar] [CrossRef] [PubMed]
- Coulie, P.G.; Van Den Eynde, B.J.; Van Der Bruggen, P.; Boon, T. Tumour antigens recognized by T lymphocytes: At the core of cancer immunotherapy. Nat. Rev. Cancer 2014, 14, 135–146. [Google Scholar] [CrossRef] [PubMed]
- Melief, C.J.M.; Van Hall, T.; Arens, R.; Ossendorp, F.; Van Der Burg, S.H. Therapeutic cancer vaccines. J. Clin. Investig. 2015, 125, 3401–3412. [Google Scholar] [CrossRef]
- Melief, C.J.M.; Van der Burg, S.H.; Toes, R.E.M.; Ossendorp, F.; Offringa, R. Effective therapeutic anticancer vaccines based on precision guiding of cytolytic T lymphocytes. Immunol. Rev. 2002, 188, 177–182. [Google Scholar] [CrossRef]
- Jiang, S.; Gong, M.; Xu, X.N. Editorial: Research, Development and Clinical Trials for Peptide-Based Vaccines. Front. Immunol. 2022, 13, 894989. [Google Scholar] [CrossRef]
- Schneble, E.J.; Berry, J.S.; Trappey, F.A.; Clifton, G.T.; Ponniah, S.; Mittendorf, E.; Peoples, G.E. The HER2 peptide nelipepimut-S (E75) vaccine (NeuVaxTM) in breast cancer patients at risk for recurrence: Correlation of immunologic data with clinical response. Immunotherapy 2014, 6, 519–531. [Google Scholar] [CrossRef]
- Chick, R.C.; Clifton, G.T.; Hale, D.F.; Vreeland, T.J.; Hickerson, A.T.; Bohan, P.M.K.; McCarthy, P.M.; Litton, J.K.; Alatrash, G.; Murthy, R.K.; et al. Subgroup analysis of nelipepimut-S plus GM-CSF combined with trastuzumab versus trastuzumab alone to prevent recurrences in patients with high-risk, HER2 low-expressing breast cancer. Clin. Immunol. 2021, 225, 108679. [Google Scholar] [CrossRef]
- Clifton, G.T.; Peoples, G.E.; Mittendorf, E.A. The development and use of the E75 (HER2 369-377) peptide vaccine. Future Oncol. 2016, 12, 1321–1329. [Google Scholar] [CrossRef] [PubMed]
- Mittendorf, E.A.; Lu, B.; Melisko, M.; Hiller, J.P.; Bondarenko, I.; Brunt, A.M.; Sergii, G.; Petrakova, K.; Peoples, G.E. Efficacy and safety analysis of nelipepimut-S vaccine to prevent breast cancer recurrence: A randomized, multicenter, phase III clinical trial. Clin. Cancer Res. 2019, 25, 4248–4254. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez, P.C.; Popa, X.; Martínez, O.; Mendoza, S.; Santiesteban, E.; Crespo, T.; Amador, R.M.; Fleytas, R.; Acosta, S.C.; Otero, Y.; et al. A Phase III Clinical Trial of the Epidermal Growth Factor Vaccine CIMAvax-EGF as Switch Maintenance Therapy in Advanced Non-Small Cell Lung Cancer Patients. Clin. Cancer Res. 2016, 22, 3782–3790. [Google Scholar] [CrossRef] [PubMed]
- Tagliamento, M.; Rijavec, E.; Barletta, G.; Biello, F.; Rossi, G.; Grossi, F.; Genova, C. CIMAvax-EGF, a therapeutic non-small cell lung cancer vaccine. Expert Opin. Biol. Ther. 2018, 18, 829–835. [Google Scholar] [CrossRef] [PubMed]
- Hanisch, F.G. Design of a MUC1-based cancer vaccine. Biochem. Soc. Trans. 2005, 33, 705–708. [Google Scholar] [CrossRef] [PubMed]
- Gao, T.; Cen, Q.; Lei, H. A review on development of MUC1-based cancer vaccine. Biomed. Pharmacother. 2020, 132, 110888. [Google Scholar] [CrossRef] [PubMed]
- Taylor-Papadimitriou, J.; Burchell, J.M.; Graham, R.; Beatson, R. Latest developments in MUC1 immunotherapy. Biochem. Soc. Trans. 2018, 46, 659–668. [Google Scholar] [CrossRef]
- Zahm, C.D.; Colluru, V.T.; McNeel, D.G. DNA vaccines for prostate cancer. Pharmacol. Ther. 2017, 174, 27–42. [Google Scholar] [CrossRef]
- Sahin, U.; Derhovanessian, E.; Miller, M.; Kloke, B.-P.; Simon, P.; Löwer, M.; Bukur, V.; Tadmor, A.D.; Luxemburger, U.; Schrörs, B.; et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017, 547, 222–226. [Google Scholar] [CrossRef]
- Kreiter, S.; Vormehr, M.; van de Roemer, N.; Diken, M.; Löwer, M.; Diekmann, J.; Boegel, S.; Schrörs, B.; Vascotto, F.; Castle, J.C.; et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 2015, 520, 692–696. [Google Scholar] [CrossRef]
- Mei, Y.; Wang, X. RNA modification in mRNA cancer vaccines. Clin. Exp. Med. 2023, 14, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Kuai, R.; Ochyl, L.J.; Bahjat, K.S.; Schwendeman, A.; Moon, J.J. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nat. Mater. 2017, 16, 489–498. [Google Scholar] [CrossRef]
- Su, C.; Peng, L.; Sham, J.; Wang, X.; Zhang, Q.; Chua, D.; Liu, C.; Cui, Z.; Xue, H.; Wu, H.; et al. Immune Gene-Viral Therapy with Triplex Efficacy Mediated by Oncolytic Adenovirus Carrying an Interferon-γ Gene Yields Efficient Antitumor Activity in Immunodeficient and Immunocompetent Mice. Mol. Ther. 2006, 13, 918–927. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.A. Transgene FDNA vaccines: A review. J. Intern. Med. 2003, 253, 402–410. [Google Scholar] [CrossRef] [PubMed]
- Blass, E.; Ott, P.A. Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat. Rev. Clin. Oncol. 2021, 18, 215–229. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Zhang, L.; Xu, Z.; Miao, L.; Huang, L. mRNA Vaccine with Antigen-Specific Checkpoint Blockade Induces an Enhanced Immune Response against Established Melanoma. Mol. Ther. 2018, 26, 420–434. [Google Scholar] [CrossRef]
- Schoenmaker, L.; Witzigmann, D.; Kulkarni, J.A.; Verbeke, R.; Kersten, G.; Jiskoot, W.; Crommelin, D.J. mRNA-lipid nanoparticle COVID-19 vaccines: Structure and stability. Int. J. Pharm. 2021, 601, 120586. [Google Scholar] [CrossRef]
- Miao, L.; Zhang, Y.; Huang, L. mRNA vaccine for cancer immunotherapy. Mol. Cancer 2021, 20, 41. [Google Scholar] [CrossRef]
- Cu, Y.; Broderick, K.E.; Banerjee, K.; Hickman, J.; Otten, G.; Barnett, S.; Kichaev, G.; Sardesai, N.Y.; Ulmer, J.B.; Geall, A. Enhanced Delivery and Potency of Self-Amplifying mRNA Vaccines by Electroporation in Situ. Vaccines 2013, 1, 367–383. [Google Scholar] [CrossRef]
- Ulmer, J.B.; Geall, A.J. Recent innovations in mRNA vaccines. Curr. Opin. Immunol. 2016, 41, 18–22. [Google Scholar] [CrossRef]
- Ho, W.; Gao, M.; Li, F.; Li, Z.; Zhang, X.Q.; Xu, X. Next-Generation Vaccines: Nanoparticle-Mediated DNA and mRNA Delivery. Adv. Healthc. Mater. 2021, 10, 2001812. [Google Scholar] [CrossRef] [PubMed]
- Huang, X.; Zhang, G.; Tang, T.Y.; Gao, X.; Liang, T.B. Personalized pancreatic cancer therapy: From the perspective of mRNA vaccine. Mil. Med. Res. 2022, 9, 53. [Google Scholar] [CrossRef] [PubMed]
- Brisse, M.; Vrba, S.M.; Kirk, N.; Liang, Y.; Ly, H. Emerging Concepts and Technologies in Vaccine Development. Front. Immunol. 2020, 11, 583077. [Google Scholar] [CrossRef] [PubMed]
- Rausch, S.; Schwentner, C.; Stenzl, A.; Bedke, J. mRNA vaccine CV9103 and CV9104 for the treatment of prostate cancer. Hum. Vaccines Immunother. 2014, 10, 3146–3152. [Google Scholar] [CrossRef] [PubMed]
- Rosenberg, S.A.; Restifo, N.P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015, 348, 62–68. [Google Scholar] [CrossRef]
- Finn, O.J. The dawn of vaccines for cancer prevention. Nat. Rev. Immunol. 2018, 18, 183–194. [Google Scholar] [CrossRef]
- Lichty, B.D.; Breitbach, C.J.; Stojdl, D.F.; Bell, J.C. Going viral with cancer immunotherapy. Nat. Rev. Cancer 2014, 14, 559–567. [Google Scholar] [CrossRef]
- O’Bryan, S.M.; Mathis, J.M. Oncolytic Virotherapy for Breast Cancer Treatment. Curr. Gene Ther. 2018, 18, 192–205. [Google Scholar] [CrossRef]
- Drake, C.G. Prostate cancer as a model for tumour immunotherapy. Nat. Rev. Immunol. 2010, 10, 580–593. [Google Scholar] [CrossRef]
- Gubin, M.M.; Artyomov, M.N.; Mardis, E.R.; Schreiber, R.D. Tumor neoantigens: Building a framework for personalized cancer immunotherapy. J. Clin. Investig. 2015, 125, 3413–3421. [Google Scholar]
- Russell, S.J.; Peng, K.W. Viruses as anticancer drugs. Trends Pharmacol. Sci. 2007, 28, 326–333. [Google Scholar] [CrossRef]
- Kaufman, H.L.; Kohlhapp, F.J.; Zloza, A. Oncolytic viruses: A new class of immunotherapy drugs. Nat. Rev. Drug Discov. 2015, 14, 642–662. [Google Scholar] [CrossRef] [PubMed]
- Rollier, C.S.; Reyes-Sandoval, A.; Cottingham, M.G.; Ewer, K.; Hill, A.V.S. Viral vectors as vaccine platforms: Deployment in sight. Curr. Opin. Immunol. 2011, 23, 377–382. [Google Scholar] [CrossRef] [PubMed]
- Ura, T.; Okuda, K.; Shimada, M. Developments in viral vector-based vaccines. Vaccines 2014, 2, 624–641. [Google Scholar] [CrossRef] [PubMed]
- Pol, J.; Bloy, N.; Obrist, F.; Eggermont, A.; Galon, J.; Cremer, I.; Erbs, P.; Limacher, J.M.; Preville, X.; Zitvogel, L.; et al. Trial watch: Oncolytic viruses for cancer therapy. Oncoimmunology 2014, 3, 37–41. [Google Scholar] [CrossRef] [PubMed]
- Prestwich, R.J.D.; Harrington, K.J.; Pandha, H.S.; Vile, R.G.; Melcher, A.A.; Errington, F. Oncolytic viruses: A novel form of immunotherapy. Expert Rev. Anticancer Ther. 2008, 8, 1581–1588. [Google Scholar] [CrossRef]
- Anderson, R.J.; Schneider, J. Plasmid DNA and viral vector-based vaccines for the treatment of cancer. Vaccine 2007, 25 (Suppl. 2), 24–34. [Google Scholar] [CrossRef]
- Vanneman, M.; Dranoff, G. Combining immunotherapy and targeted therapies in cancer treatment. Nat. Rev. Cancer 2012, 12, 237–251. [Google Scholar] [CrossRef]
- Galluzzi, L.; Buqué, A.; Kepp, O.; Zitvogel, L.; Kroemer, G. Immunogenic cell death in cancer and infectious disease. Nat. Rev. Immunol. 2017, 17, 97–111. [Google Scholar] [CrossRef]
- Watanabe, D.; Goshima, F. Oncolytic virotherapy by HSV. Adv. Exp. Med. Biol. 2018, 1045, 63–84. [Google Scholar]
- Ramesh, N.; Ge, Y.; Ennist, D.L.; Zhu, M.; Mina, M.; Ganesh, S.; Reddy, P.S.; Yu, D.C. CG0070, a conditionally replicating granulocyte macrophage colony-stimulating factor—Armed oncolytic adenovirus for the treatment of bladder cancer. Clin. Cancer Res. 2006, 12, 305–313. [Google Scholar] [CrossRef] [PubMed]
- Albershardt, T.C.; Campbell, D.J.; Parsons, A.J.; Slough, M.M.; Ter Meulen, J.; Berglund, P. LV305, a dendritic cell-targeting integration-deficient ZVex TM-based lentiviral vector encoding NY-ESO-1, induces potent anti-tumor immune response. Mol. Ther. Oncolytics 2016, 3, 305–313. [Google Scholar] [CrossRef] [PubMed]
- Heo, J.; Reid, T.; Ruo, L.; Breitbach, C.J.; Rose, S.; Bloomston, M.; Cho, M.; Lim, H.Y.; Chung, H.C.; Kim, C.W.; et al. Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer. Nat. Med. 2013, 19, 329–336. [Google Scholar] [CrossRef]
- Breitbach, C.; Bell, J.C.; Hwang, T.-H.; Kirn, D.; Burke, J. The emerging therapeutic potential of the oncolytic immunotherapeutic Pexa-Vec (JX-594). Oncolytic Virother. 2015, 4, 25–31. [Google Scholar] [CrossRef] [PubMed]
- Parato, K.A.; Breitbach, C.J.; Le Boeuf, F.; Wang, J.; Storbeck, C.; Ilkow, C.; Diallo, J.-S.; Falls, T.; Burns, J.; Garcia, V.; et al. The oncolytic poxvirus JX-594 selectively replicates in and destroys cancer cells driven by genetic pathways commonly activated in cancers. Mol. Ther. 2012, 20, 749–758. [Google Scholar] [CrossRef] [PubMed]
- Banchereau, J.; Steinman, R.M. Dendritic cells and the control of immunity. Nature 1998, 392, 245–252. [Google Scholar] [CrossRef] [PubMed]
- Rosenberg, S.A.; Yang, J.C.; Schwartzentruber, D.J.; Hwu, P.; Marincola, F.M.; Topalian, S.L.; Restifo, N.P.; Sznol, M.; Schwarz, S.L.; Spiess, P.J.; et al. Impact of Cytokine Administration on the Generation of Antitumor Reactivity in Patients with Metastatic Melanoma Receiving a Peptide Vaccine. J. Immunol. 1999, 163, 1690–1695. [Google Scholar] [CrossRef] [PubMed]
- Tuyaerts, S.; Aerts, J.L.; Corthals, J.; Neyns, B.; Heirman, C.; Breckpot, K.; Thielemans, K.; Bonehill, A. Current approaches in dendritic cell generation and future implications for cancer immunotherapy. Cancer Immunol. Immunother. 2007, 56, 1513–1537. [Google Scholar]
- Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [Google Scholar] [CrossRef]
- Han, J.; Bhatta, R.; Liu, Y.; Bo, Y.; Elosegui-Artola, A.; Wang, H. Metabolic glycan labeling immobilizes dendritic cell membrane and enhances antitumor efficacy of dendritic cell vaccine. Nat. Commun. 2023, 14, 5049. [Google Scholar] [CrossRef]
- Carreno, B.M.; Magrini, V.; Becker-Hapak, M.; Kaabinejadian, S.; Hundal, J.; Petti, A.A.; Ly, A.; Lie, W.R.; Hildebrand, W.H.; Mardis, E.R.; et al. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science 2015, 348, 803–808. [Google Scholar] [CrossRef] [PubMed]
- Morse, M.A.; Clay, T.M.; Hobeika, A.C.; Osada, T.; Khan, S.; Chui, S.; Niedzwiecki, D.; Panicali, D.; Schlom, J.; Lyerly, H.K. Phase I study of immunization with dendritic cells modified with fowlpox encoding carcinoembryonic antigen and costimulatory molecules. Clin. Cancer Res. 2005, 11, 3017–3024. [Google Scholar] [CrossRef] [PubMed]
- Anguille, S.; Smits, E.L.; Lion, E.; Van Tendeloo, V.F.; Berneman, Z.N. Clinical use of dendritic cells for cancer therapy. Lancet Oncol. 2014, 15, 257–267. [Google Scholar]
- Antonarakis, E.S.; Small, E.J.; Petrylak, D.P.; Quinn, D.I.; Kibel, A.S.; Chang, N.N.; Dearstyne, E.; Harmon, M.; Campogan, D.; Haynes, H.; et al. Antigen-specific CD8 lytic phenotype induced by sipuleucel-T in hormone-sensitive or castration-resistant prostate cancer and association with overall survival. Clin. Cancer Res. 2018, 24, 4662–4671. [Google Scholar] [CrossRef] [PubMed]
- Carballido, E.; Fishman, M. Sipuleucel-T: Prototype for development of anti-tumor vaccines. Curr. Oncol. Rep. 2011, 13, 112–119. [Google Scholar] [CrossRef]
- Polyzoidis, S.; Ashkan, K. DCVax®-L—Developed by Northwest Biotherapeutics. Hum. Vaccines Immunother. 2014, 10, 3139–3145. [Google Scholar] [CrossRef]
- Guo, C.; Manjili, M.H.; Subjeck, J.R.; Sarkar, D.; Fisher, P.B.; Wang, X.Y. Therapeutic cancer vaccines. In Past, Present, and Future, 1st ed.; Advances in Cancer Research; Elsevier Inc.: Amsterdam, The Netherlands, 2013; Volume 119, pp. 421–475. [Google Scholar] [CrossRef]
- Shu, S.; Zheng, R.; Lee, W.T.; Cohen, P.A. Immunogenicity of dendritic-tumor fusion hybrids and their utility in cancer immunotherapy. Crit. Rev. Immunol. 2007, 27, 463–483. [Google Scholar] [CrossRef]
- Emens, L.A.; Jaffee, E.M. Leveraging the activity of tumor vaccines with cytotoxic chemotherapy. Cancer Res. 2005, 65, 8059–8064. [Google Scholar] [CrossRef]
- Principe, D.R.; Korc, M.; Kamath, S.D.; Munshi, H.G.; Rana, A. Trials and tribulations of pancreatic cancer immunotherapy. Cancer Lett. 2021, 504, 1–14. [Google Scholar] [CrossRef]
- Ward, J.E.; McNeel, D.G. GVAX: An allogeneic, whole-cell, GM-CSF-secreting cellular immunotherapy for the treatment of prostate cancer. Expert Opin. Biol. Ther. 2007, 7, 1893–1902. [Google Scholar] [CrossRef]
- Petricciani, J.; Koren, E.; Morton, D. Analysis of the in vivo proliferative capacity of a whole cell cancer vaccine. Biologicals 2016, 44, 60–63. [Google Scholar] [CrossRef] [PubMed]
- Di Pietro, A.; Tosti, G.; Ferrucci, P.F.; Testori, A. Oncophage: Step to the future for vaccine therapy in melanoma. Expert Opin. Biol. Ther. 2008, 8, 1973–1984. [Google Scholar] [CrossRef] [PubMed]
- Jacobsohn, K.M.; Wood, C.G. Adjuvant Therapy for Renal Cell Carcinoma. Semin. Oncol. 2006, 33, 576–582. [Google Scholar] [CrossRef]
- Itoh, K.; Yamada, A.; Mine, T.; Noguchi, M. Recent advances in cancer vaccines: An overview. Jpn. J. Clin. Oncol. 2009, 39, 73–80. [Google Scholar] [CrossRef] [PubMed]
- Mittendorf, E.A.; Ardavanis, A.; Litton, J.K.; Shumway, N.M.; Hale, D.F.; Murray, J.L.; Perez, S.A.; Ponniah, S.; Baxevanis, C.N.; Papamichail, M.; et al. Primary analysis of a prospective, randomized, single-blinded phase II trial evaluating the HER2 peptide GP2 vaccine in breast cancer patients to prevent recurrence. Oncotarget 2016, 7, 66192–66201. [Google Scholar] [CrossRef] [PubMed]
- Lepisto, A.J.; Moser, A.J.; Zeh, H.; Lee, K.; Bartlett, D.; McKolanis, J.R.; Geller, B.A.; Schmotzer, A.; Potter, D.P.; Whiteside, T.; et al. A phase I/II study of a MUC1 peptide pulsed autologous dendritic cell vaccine as adjuvant therapy in patients with resected pancreatic and biliary tumors. Cancer Ther. 2008, 6, 955–964. [Google Scholar]
- Kübler, H.; Scheel, B.; Gnad-Vogt, U.; Miller, K.; Schultze-Seemann, W.; Dorp, F.V.; Parmiani, G.; Hampel, C.; Wedel, S.; Trojan, L.; et al. Self-adjuvanted mRNA vaccination in advanced prostate cancer patients: A first-in-man phase I/IIa study. J. Immunother. Cancer 2015, 3, 26. [Google Scholar] [CrossRef]
- Kaufman, H.L.; Kim, D.W.; Deraffele, G.; Mitcham, J.; Coffin, R.S.; Kim-Schulze, S. Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Ann. Surg. Oncol. 2010, 17, 718–730. [Google Scholar] [CrossRef]
- Hwang, T.-H.; Moon, A.; Burke, J.; Ribas, A.; Stephenson, J.; Breitbach, C.J.; Daneshmand, M.; De Silva, N.; Parato, K.; Diallo, J.-S.; et al. A mechanistic proof-of-concept clinical trial with JX-594, a targeted multi-mechanistic oncolytic poxvirus, in patients with metastatic melanoma. Mol. Ther. 2011, 19, 1913–1922. [Google Scholar] [CrossRef]
- Liau, L.M.; Ashkan, K.; Tran, D.D.; Campian, J.L.; Trusheim, J.E.; Cobbs, C.S.; Heth, J.A.; Salacz, M.; Taylor, S.; D’andre, S.D.; et al. First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. J. Transl. Med. 2018, 16, 142. [Google Scholar] [CrossRef]
- Lutz, E.; Yeo, C.J.; Lillemoe, K.D.; Biedrzycki, B.; Kobrin, B.; Herman, J.; Sugar, E.; Piantadosi, S.; Cameron, J.L.; Solt, S.; et al. A lethally irradiated allogeneic granulocyte-macrophage colony stimulating factor-secreting tumor vaccine for pancreatic adenocarcinoma: A phase II trial of safety, efficacy, and immune activation. Ann. Surg. 2011, 253, 328–335. [Google Scholar] [PubMed]
- Morton, D.L.; Hsueh, E.C.; Essner, R.; Foshag, L.J.; O’Day, S.J.; Bilchik, A.; Gupta, R.K.; Hoon, D.S.; Ravindranath, M.; Nizze, J.A.; et al. Prolonged survival of patients receiving active immunotherapy with Canvaxin therapeutic polyvalent vaccine after complete resection of melanoma metastatic to regional lymph nodes. Ann. Surg. 2002, 236, 438–449. [Google Scholar] [CrossRef] [PubMed]
- Wood, C.; Srivastava, P.; Bukowski, R.; Lacombe, L.; Gorelov, A.I.; Gorelov, S.; Mulders, P.; Zielinski, H.; Hoos, A.; Teofilovici, F.; et al. An adjuvant autologous therapeutic vaccine (HSPPC-96; vitespen) versus observation alone for patients at high risk of recurrence after nephrectomy for renal cell carcinoma: A multicentre, open-label, randomised phase III trial. Lancet 2008, 372, 145–154. [Google Scholar] [CrossRef] [PubMed]
- Testori, A.; Richards, J.; Whitman, E.; Mann, G.B.; Lutzky, J.; Camacho, L.; Parmiani, G.; Tosti, G.; Kirkwood, J.M.; Hoos, A.; et al. Phase III comparison of vitespen, an autologous tumor-derived heat shock protein gp96 peptide complex vaccine, with physician’s choice of treatment for stage IV melanoma: The C-100-21 study group. J. Clin. Oncol. 2008, 26, 955–962. [Google Scholar] [CrossRef] [PubMed]
- Lawler, S.E.; Speranza, M.C.; Cho, C.F.; Chiocca, E.A. Oncolytic viruses in cancer treatment a review. JAMA Oncol. 2017, 3, 841–849. [Google Scholar] [CrossRef]
- Lin, M.J.; Svensson-Arvelund, J.; Lubitz, G.S.; Marabelle, A.; Melero, I.; Brown, B.D.; Brody, J.D. Cancer vaccines: The next immunotherapy frontier. Nat. Cancer 2022, 3, 911–926. [Google Scholar] [CrossRef]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef]
- Topalian, S.L.; Drake, C.G.; Pardoll, D.M. Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell 2015, 27, 450–461. [Google Scholar] [CrossRef]
- Cheever, M.A.; Allison, J.P.; Ferris, A.S.; Finn, O.J.; Hastings, B.M.; Hecht, T.T.; Mellman, I.; Prindiville, S.A.; Viner, J.L.; Weiner, L.M.; et al. The prioritization of cancer antigens: A National Cancer Institute pilot project for the acceleration of translational research. Clin. Cancer Res. 2009, 15, 5323–5337. [Google Scholar] [CrossRef]
- O’Hagan, D.T.; Fox, C.B. New generation adjuvants—From empiricism to rational design. Vaccine 2015, 33, B14–B20. [Google Scholar] [CrossRef]
- Kawai, T.; Akira, S. Toll-like Receptors and Their Crosstalk with Other Innate Receptors in Infection and Immunity. Immunity 2011, 34, 637–650. [Google Scholar] [CrossRef] [PubMed]
- Coffman, R.L.; Sher, A.; Seder, R.A. Vaccine adjuvants: Putting innate immunity to work. Immunity 2010, 33, 492–503. [Google Scholar] [CrossRef] [PubMed]
- Reed, S.G.; Orr, M.T.; Fox, C.B. Key roles of adjuvants in modern vaccines. Nat. Med. 2013, 19, 1597–1608. [Google Scholar] [CrossRef] [PubMed]
- Kocic, G.; Hadzi-Djokic, J.; Colic, M.; Veljkovic, A.; Tomovic, K.; Roumeliotis, S.; Smelcerovic, A.; Liakopoulos, V. The Role of Nucleases Cleaving TLR3, TLR7/8 and TLR9 Ligands, Dicer RNase and miRNA/piRNA Proteins in Functional Adaptation to the Immune Escape and Xenophagy of Prostate Cancer Tissue. Int. J. Mol. Sci. 2023, 24, 509. [Google Scholar] [CrossRef] [PubMed]
- Zhuang, W.-R.; Wang, Y.; Nie, W.; Lei, Y.; Liang, C.; He, J.; Zuo, L.; Huang, L.-L.; Xie, H.-Y. Bacterial outer membrane vesicle based versatile nanosystem boosts the efferocytosis blockade triggered tumor-specific immunity. Nat. Commun. 2023, 14, 1675. [Google Scholar] [CrossRef] [PubMed]
- Garland, K.M.; Sheehy, T.L.; Wilson, J.T. Chemical and Biomolecular Strategies for STING Pathway Activation in Cancer Immunotherapy. Chem. Rev. 2022, 122, 5977–6039. [Google Scholar] [CrossRef]
- Li, A.; Yi, M.; Qin, S.; Song, Y.; Chu, Q.; Wu, K. Activating cGAS-STING pathway for the optimal effect of cancer immunotherapy. J. Hematol. Oncol. 2019, 12, 35. [Google Scholar] [CrossRef]
- Hegde, P.S.; Chen, D.S. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020, 52, 17–35. [Google Scholar] [CrossRef]
- Patel, S.A.; Minn, A.J. Combination Cancer Therapy with Immune Checkpoint Blockade: Mechanisms and Strategies. Immunity 2018, 48, 417–433. [Google Scholar] [CrossRef]
- Ribas, A.; Dummer, R.; Puzanov, I.; VanderWalde, A.; Andtbacka, R.H.I.; Michielin, O.; Olszanski, A.J.; Malvehy, J.; Cebon, J.; Fernandez, E.; et al. Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration and Improves Anti-PD-1 Immunotherapy. Cell 2017, 170, 1109–1119.e10. [Google Scholar] [CrossRef]
- Gandhi, L.; Rodríguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in Metastatic Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 2078–2092. [Google Scholar] [CrossRef] [PubMed]
- Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef] [PubMed]
- Motzer, R.J.; Penkov, K.; Haanen, J.; Rini, B.; Albiges, L.; Campbell, M.T.; Venugopal, B.; Kollmannsberger, C.; Negrier, S.; Uemura, M.; et al. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2019, 380, 1103–1115. [Google Scholar] [CrossRef] [PubMed]
- Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef]
- Morse, M.A.; Gwin, W.R.; Mitchell, D.A. Vaccine Therapies for Cancer: Then and Now. Target. Oncol. 2021, 16, 121–152. [Google Scholar] [CrossRef]
- Igarashi, Y.; Sasada, T. Cancer Vaccines: Toward the Next Breakthrough in Cancer Immunotherapy. J. Immunol. Res. 2020, 2020, 5825401. [Google Scholar] [CrossRef]
- Postow, M.A.; Sidlow, R.; Hellmann, M.D. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N. Engl. J. Med. 2018, 378, 158–168. [Google Scholar] [CrossRef]
- Dusetzina, S.B.; Winn, A.N.; Abel, G.A.; Huskamp, H.A.; Keating, N.L. Cost sharing and adherence to tyrosine kinase inhibitors for patients with chronic myeloid leukemia. J. Clin. Oncol. 2014, 32, 306–311. [Google Scholar] [CrossRef]
- Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1–Positive Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef]
- Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef]
- Zraik, I.M.; Heß-Busch, Y. Management of chemotherapy side effects and their long-term sequelae. Urol. Ausg. A 2021, 60, 862–871. [Google Scholar] [CrossRef] [PubMed]
- Obeid, M.; Tesniere, A.; Ghiringhelli, F.; Fimia, G.M.; Apetoh, L.; Perfettini, J.-L.; Castedo, M.; Mignot, G.; Panaretakis, T.; Casares, N.; et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat. Med. 2007, 13, 54–61. [Google Scholar] [CrossRef] [PubMed]
- Wainwright, D.A.; Dey, M.; Chang, A.; Lesniak, M.S. Targeting tregs in malignant brain cancer: Overcoming IDO. Front. Immunol. 2013, 4, 116. [Google Scholar] [CrossRef] [PubMed]
- Ma, Z.; Zhang, W.; Dong, B.; Xin, Z.; Ji, Y.; Su, R.; Shen, K.; Pan, J.; Wang, Q.; Xue, W. Docetaxel remodels prostate cancer immune microenvironment and enhances checkpoint inhibitorbased immunotherapy. Theranostics 2022, 12, 4965–4979. [Google Scholar] [CrossRef] [PubMed]
- Sasso, M.S.; Lollo, G.; Pitorre, M.; Solito, S.; Pinton, L.; Valpione, S.; Bastiat, G.; Mandruzzato, S.; Bronte, V.; Marigo, I.; et al. Low dose gemcitabine-loaded lipid nanocapsules target monocytic myeloid-derived suppressor cells and potentiate cancer immunotherapy. Biomaterials 2016, 96, 47–62. [Google Scholar] [CrossRef]
- Hauner, K.; Maisch, P.; Retz, M. Nebenwirkungen der Chemotherapie. Urol. Ausg. A 2017, 56, 472–479. [Google Scholar] [CrossRef]
- Farkona, S.; Diamandis, E.P.; Blasutig, I.M. Cancer immunotherapy: The beginning of the end of cancer? BMC Med. 2016, 14, 73. [Google Scholar] [CrossRef]
- Emens, L.A.; Cruz, C.; Eder, J.P.; Braiteh, F.; Chung, C.; Tolaney, S.M.; Kuter, I.; Nanda, R.; Cassier, P.A.; Delord, J.P.; et al. Long-term Clinical Outcomes and Biomarker Analyses of Atezolizumab Therapy for Patients with Metastatic Triple-Negative Breast Cancer: A Phase 1 Study. JAMA Oncol. 2019, 5, 74–82. [Google Scholar] [CrossRef]
- Hinshaw, D.C.; Shevde, L.A. The tumor microenvironment innately modulates cancer progression. Cancer Res. 2019, 79, 4557–4567. [Google Scholar] [CrossRef]
- Abshire, D.; Lang, M.K. The Evolution of Radiation Therapy in Treating Cancer. Semin. Oncol. Nurs. 2018, 34, 151–157. [Google Scholar] [CrossRef]
- Dewan, M.Z.; Galloway, A.E.; Kawashima, N.; Dewyngaert, J.K.; Babb, J.S.; Formenti, S.C.; Demaria, S. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin. Cancer Res. 2009, 15, 5379–5388. [Google Scholar] [CrossRef] [PubMed]
- Golden, E.B.; Demaria, S.; Schiff, P.B.; Chachoua, A.; Formenti, S.C. An abscopal response to radiation and ipilimumab in a patient with metastatic non-small cell lung cancer. Cancer Immunol. Res. 2013, 1, 365–372. [Google Scholar] [CrossRef] [PubMed]
- Weichselbaum, R.R.; Liang, H.; Deng, L.; Fu, Y.X. Radiotherapy and immunotherapy: A beneficial liaison? Nat. Rev. Clin. Oncol. 2017, 14, 365–379. [Google Scholar] [CrossRef] [PubMed]
- Gupta, A.; Probst, H.C.; Vuong, V.; Landshammer, A.; Muth, S.; Yagita, H.; Schwendener, R.; Pruschy, M.; Knuth, A.; van den Broek, M. Radiotherapy Promotes Tumor-Specific Effector CD8+ T Cells via Dendritic Cell Activation. J. Immunol. 2012, 189, 558–566. [Google Scholar] [CrossRef]
- Barker, H.E.; Paget, J.T.E.; Khan, A.A.; Harrington, K.J. The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat. Rev. Cancer 2015, 15, 409–425. [Google Scholar] [CrossRef]
- Twyman-Saint Victor, C.; Rech, A.J.; Maity, A.; Rengan, R.; Pauken, K.E.; Stelekati, E.; Benci, J.L.; Xu, B.; Dada, H.; Odorizzi, P.M.; et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015, 520, 373–377. [Google Scholar] [CrossRef]
- Lugade, A.A.; Moran, J.P.; Gerber, S.A.; Rose, R.C.; Frelinger, J.G.; Lord, E.M. Local Radiation Therapy of B16 Melanoma Tumors Increases the Generation of Tumor Antigen-Specific Effector Cells That Traffic to the Tumor. J. Immunol. 2005, 174, 7516–7523. [Google Scholar] [CrossRef]
- Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Yokoi, T.; Chiappori, A.; Lee, K.H.; De Wit, M.; et al. Durvalumab after Chemoradiotherapy in Stage III Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 377, 1919–1929. [Google Scholar] [CrossRef]
- Galluzzi, L.; Humeau, J.; Buqué, A.; Zitvogel, L.; Kroemer, G. Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat. Rev. Clin. Oncol. 2020, 17, 725–741. [Google Scholar] [CrossRef]
- Shuel, S.L. Targeted cancer therapies. Can. Fam. Physician 2022, 68, 515–518. [Google Scholar] [CrossRef]
- Huang, R.-Y.; Eppolito, C.; Lele, S.; Shrikant, P.; Matsuzaki, J.; Odunsi, K. LAG3 and PD1 co-inhibitory molecules collaborate to limit CD8+ T cell signaling and dampen antitumor immunity in a murine ovarian cancer model. Oncotarget 2015, 6, 27359–27377. [Google Scholar] [CrossRef]
- Beatty, G.L.; Gladney, W.L. Immune escape mechanisms as a guide for cancer immunotherapy. Clin. Cancer Res. 2015, 21, 687–692. [Google Scholar] [CrossRef] [PubMed]
- Holohan, C.; Van Schaeybroeck, S.; Longley, D.B.; Johnston, P.G. Cancer drug resistance: An evolving paradigm. Nat. Rev. Cancer 2013, 13, 714–726. [Google Scholar] [CrossRef] [PubMed]
- Zitvogel, L.; Galluzzi, L.; Smyth, M.J.; Kroemer, G. Mechanism of Action of Conventional and Targeted Anticancer Therapies: Reinstating Immunosurveillance. Immunity 2013, 39, 74–88. [Google Scholar] [CrossRef] [PubMed]
- Yun, C.O.; Hong, J.W.; Yoon, A.R. Current clinical landscape of oncolytic viruses as novel cancer immunotherapeutic and recent preclinical advancements. Front. Immunol. 2022, 13, 953410. [Google Scholar] [CrossRef]
- Parato, K.A.; Senger, D.; Forsyth, P.A.J.; Bell, J.C. Recent progress in the battle between oncolytic viruses and tumours. Nat. Rev. Cancer 2005, 5, 965–976. [Google Scholar] [CrossRef]
- Wang, W.; Liu, S.; Dai, P.; Yang, N.; Wang, Y.; Giese, R.A.; Merghoub, T.; Wolchok, J.; Deng, L. Elucidating mechanisms of antitumor immunity mediated by live oncolytic vaccinia and heat-inactivated vaccinia. J. Immunother. Cancer 2021, 9, e002569. [Google Scholar] [CrossRef]
- Lin, J.; Sun, S.; Zhao, K.; Gao, F.; Wang, R.; Li, Q.; Zhou, Y.; Zhang, J.; Li, Y.; Wang, X.; et al. Oncolytic Parapoxvirus induces Gasdermin E-mediated pyroptosis and activates antitumor immunity. Nat. Commun. 2023, 14, 224. [Google Scholar] [CrossRef]
- Enekegho, L.; Stuart, D. Smart Oncolytic Adenovirotherapy to Induce Killing of Cancer Cells and Elicit Antitumor Immunity. Eureka 2022, 7, 1–18. [Google Scholar] [CrossRef]
- Zuo, S.; Wei, M.; Xu, T.; Kong, L.; He, B.; Wang, S.; Wang, S.; Wu, J.; Dong, J.; Wei, J. An engineered oncolytic vaccinia virus encoding a single-chain variable fragment against TIGIT induces effective antitumor immunity and synergizes with PD-1 or LAG-3 blockade. J. Immunother. Cancer 2021, 9, e002843. [Google Scholar] [CrossRef]
- Das, K.; Belnoue, E.; Rossi, M.; Hofer, T.; Danklmaier, S.; Nolden, T.; Schreiber, L.-M.; Angerer, K.; Kimpel, J.; Hoegler, S.; et al. A modular self-adjuvanting cancer vaccine combined with an oncolytic vaccine induces potent antitumor immunity. Nat. Commun. 2021, 12, 5195. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Shu, Y.; Hu, S.; Qi, Z.; Chen, Y.; Ma, J.; Wang, Y.; Cheng, P. In Situ Tumor Vaccine Expressing Anti-CD47 Antibody Enhances Antitumor Immunity. Front. Oncol. 2022, 12, 897561. [Google Scholar] [CrossRef] [PubMed]
- Hofer, T.; Rossi, M.; Carboni, S.; Di Berardino Besson, W.; von Laer, D.; Wollmann, G.; Derouazi, M.; Santiago-Raber, M.L. Heterologous Prime-Boost Vaccination with a Peptide-Based Vaccine and Viral Vector Reshapes Dendritic Cell, CD4+ and CD8+ T Cell Phenotypes to Improve the Antitumor Therapeutic Effect. Cancers 2021, 13, 6107. [Google Scholar] [CrossRef] [PubMed]
- Bartlett, D.L.; Liu, Z.; Sathaiah, M.; Ravindranathan, R.; Guo, Z.; He, Y.; Guo, Z.S. Oncolytic viruses as therapeutic cancer vaccines. Mol. Cancer 2013, 12, 103. [Google Scholar] [CrossRef]
- Russell, S.J.; Peng, K.W. Oncolytic Virotherapy: A Contest between Apples and Oranges. Mol. Ther. 2017, 25, 1107–1116. [Google Scholar] [CrossRef]
- Fukuhara, H.; Ino, Y.; Todo, T. Oncolytic virus therapy: A new era of cancer treatment at dawn. Cancer Sci. 2016, 107, 1373–1379. [Google Scholar] [CrossRef]
- Pol, J.; Kroemer, G.; Galluzzi, L. First oncolytic virus approved for melanoma immunotherapy. Oncoimmunology 2016, 5, e1115641. [Google Scholar] [CrossRef]
- Harrington, K.; Freeman, D.J.; Kelly, B.; Harper, J.; Soria, J.C. Optimizing oncolytic virotherapy in cancer treatment. Nat. Rev. Drug Discov. 2019, 18, 689–706. [Google Scholar] [CrossRef]
- Goradel, N.H.; Baker, A.T.; Arashkia, A.; Ebrahimi, N.; Ghorghanlu, S.; Negahdari, B. Oncolytic virotherapy: Challenges and solutions. Curr. Probl. Cancer 2021, 45, 100639. [Google Scholar] [CrossRef]
- Mondal, M.; Guo, J.; He, P.; Zhou, D. Recent advances of oncolytic virus in cancer therapy. Hum. Vaccines Immunother. 2020, 16, 2389–2402. [Google Scholar] [CrossRef]
- Niemann, J.; Kühnel, F. Oncolytic viruses: Adenoviruses. Virus Genes 2017, 53, 700–706. [Google Scholar] [CrossRef] [PubMed]
- Moaven, O.; Mangieri, C.W.; Stauffer, J.A.; Anastasiadis, P.Z.; Borad, M.J. Strategies to Develop Potent Oncolytic Viruses and Enhance Their Therapeutic Efficacy. JCO Precis. Oncol. 2021, 1, 733–743. [Google Scholar] [CrossRef] [PubMed]
- Gubin, M.M.; Zhang, X.; Schuster, H.; Caron, E.; Ward, J.P.; Noguchi, T.; Ivanova, Y.; Hundal, J.; Arthur, C.D.; Krebber, W.J.; et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 2014, 515, 577–581. [Google Scholar] [CrossRef] [PubMed]
- Wong, S.Q.; Fellowes, A.; Doig, K.; Ellul, J.; Bosma, T.J.; Irwin, D.; Vedururu, R.; Tan, A.Y.-C.; Weiss, J.; Chan, K.S.; et al. Assessing the clinical value of targeted massively parallel sequencing in a longitudinal, prospective population-based study of cancer patients. Br. J. Cancer 2015, 112, 1411–1420. [Google Scholar] [CrossRef] [PubMed]
- McGranahan, N.; Swanton, C. Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future. Cell 2017, 168, 613–628. [Google Scholar] [CrossRef]
- Miao, D.; Van Allen, E.M. Genomic determinants of cancer immunotherapy. Curr. Opin. Immunol. 2016, 41, 32–38. [Google Scholar] [CrossRef]
- Łuksza, M.; Riaz, N.; Makarov, V.; Balachandran, V.P.; Hellmann, M.D.; Solovyov, A.; Rizvi, N.A.; Merghoub, T.; Levine, A.J.; Chan, T.A.; et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature 2017, 551, 517–520. [Google Scholar] [CrossRef]
- O’Donnell, T.J.; Rubinsteyn, A.; Bonsack, M.; Riemer, A.B.; Laserson, U.; Hammerbacher, J. MHCflurry: Open-Source Class I MHC Binding Affinity Prediction. Cell Syst. 2018, 7, 129–132.e4. [Google Scholar] [CrossRef]
- Kalaora, S.; Wolf, Y.; Feferman, T.; Barnea, E.; Greenstein, E.; Reshef, D.; Tirosh, I.; Reuben, A.; Patkar, S.; Levy, R.; et al. Combined analysis of antigen presentation and T-cell recognition reveals restricted immune responses in melanoma. Cancer Discov. 2018, 8, 1366–1375. [Google Scholar] [CrossRef]
- Bjerregaard, A.M.; Nielsen, M.; Hadrup, S.R.; Szallasi, Z.; Eklund, A.C. MuPeXI: Prediction of neo-epitopes from tumor sequencing data. Cancer Immunol. Immunother. 2017, 66, 1123–1130. [Google Scholar] [CrossRef]
- Lu, Y.C.; Robbins, P.F. Cancer immunotherapy targeting neoantigens. Semin. Immunol. 2016, 28, 22–27. [Google Scholar] [CrossRef]
- Dubensky, T.W.; Reed, S.G. Adjuvants for cancer vaccines. Semin. Immunol. 2010, 22, 155–161. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.Z.; Jou, J.; Cohen, E. Vaccine strategies for human papillomavirus-associated head and neck cancers. Cancers 2022, 14, 33. [Google Scholar] [CrossRef] [PubMed]
- Heathman, T.R.; Nienow, A.W.; McCall, M.J.; Coopman, K.; Kara, B.; Hewitt, C.J. The translation of cell-based therapies: Clinical landscape and manufacturing challenges. Regen. Med. 2015, 10, 49–64. [Google Scholar] [CrossRef] [PubMed]
- Pollock, J.; Coffman, J.; Ho, S.V.; Farid, S.S. Integrated continuous bioprocessing: Economic, operational, and environmental feasibility for clinical and commercial antibody manufacture. Biotechnol Prog. 2017, 33, 854–866. [Google Scholar] [CrossRef]
- Zhang, X.; Sharma, P.K.; Peter Goedegebuure, S.; Gillanders, W.E. Personalized cancer vaccines: Targeting the cancer mutanome. Vaccine 2017, 35, 1094–1100. [Google Scholar] [CrossRef]
- Hilf, N.; Kuttruff-Coqui, S.; Frenzel, K.; Bukur, V.; Stevanović, S.; Gouttefangeas, C.; Platten, M.; Tabatabai, G.; Dutoit, V.; Van Der Burg, S.H.; et al. Actively personalized vaccination trial for newly diagnosed glioblastoma. Nature 2019, 565, 240–245. [Google Scholar] [CrossRef]
- Kloor, M.; Von Knebel Doeberitz, M. The immune biology of microsatellite-unstable cancer. Trends Cancer 2016, 2, 121–133. [Google Scholar] [CrossRef]
- Yarchoan, M.; Johnson, B.A.; Lutz, E.R.; Laheru, D.A.; Jaffee, E.M. Targeting neoantigens to augment antitumour immunity. Nat. Rev. Cancer 2017, 17, 209–222. [Google Scholar] [CrossRef]
- Palucka, K.; Banchereau, J. Dendritic-Cell-Based Therapeutic Cancer Vaccines. Immunity 2013, 39, 38–48. [Google Scholar] [CrossRef]
- Saung, M.T.; Ke, X.; Howard, G.P.; Zheng, L.; Mao, H.Q. Particulate carrier systems as adjuvants for cancer vaccines. Biomater. Sci. 2019, 7, 4873–4887. [Google Scholar] [CrossRef] [PubMed]
- Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef] [PubMed]
- Postow, M.A.; Callahan, M.K.; Wolchok, J.D. Immune checkpoint blockade in cancer therapy. J. Clin. Oncol. 2015, 33, 1974–1982. [Google Scholar] [CrossRef] [PubMed]
- Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61. [Google Scholar] [CrossRef]
- Snyder, A.; Makarov, V.; Merghoub, T.; Yuan, J.; Zaretsky, J.M.; Desrichard, A.; Walsh, L.A.; Postow, M.A.; Wong, P.; Ho, T.S.; et al. Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. N. Engl. J. Med. 2014, 371, 2189–2199. [Google Scholar] [CrossRef]
- Mitchell, S.A. Cancer-Related Fatigue: State of the Science. PMR 2010, 2, 364–383. [Google Scholar] [CrossRef]
- Vanpouille-Box, C.; Alard, A.; Aryankalayil, M.J.; Sarfraz, Y.; Diamond, J.M.; Schneider, R.J.; Inghirami, G.; Coleman, C.N.; Formenti, S.C.; Demaria, S. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 2017, 8, 15618. [Google Scholar] [CrossRef]
- Van Tendeloo, V.F.; Van de Velde, A.; Van Driessche, A.; Cools, N.; Anguille, S.; Ladell, K.; Gostick, E.; Vermeulen, K.; Pieters, K.; Nijs, G.; et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms’ tumor 1 antigen-targeted dendritic cell vaccination. Proc. Natl. Acad. Sci. USA 2010, 107, 13824–13829. [Google Scholar] [CrossRef]
- Rosenblatt, J.; Glotzbecker, B.; Mills, H.; Vasir, B.; Tzachanis, D.; Levine, J.D.; Joyce, R.M.; Wellenstein, K.; Keefe, W.; Schickler, M.; et al. PD-1 blockade by CT-011, anti-PD-1 antibody, enhances ex vivo t-cell responses to autologous dendritic cell/myeloma fusion vaccine. J. Immunother. 2011, 34, 409–418. [Google Scholar] [CrossRef]
- Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef]
- Schuster, S.J.; Bishop, M.R.; Tam, C.S.; Waller, E.K.; Borchmann, P.; McGuirk, J.P.; Jäger, U.; Jaglowski, S.; Andreadis, C.; Westin, J.R.; et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2019, 380, 45–56. [Google Scholar] [CrossRef] [PubMed]
- Hartmann, J.; Schüßler-Lenz, M.; Bondanza, A.; Buchholz, C.J. Clinical development of CAR T cells—Challenges and opportunities in translating innovative treatment concepts. EMBO Mol. Med. 2017, 9, 1183–1197. [Google Scholar] [CrossRef] [PubMed]
- Sanmamed, M.F.; Fernández-Landázuri, S.; Rodríguez, C.; Zárate, R.; Lozano, M.D.; Zubiri, L.; Perez-Gracia, J.L.; Martín-Algarra, S.; González, A. Quantitative cell-free circulating BRAFV600E mutation analysis by use of droplet digital PCR in the follow-up of patients with melanoma being treated with BRAF inhibitors. Clin. Chem. 2015, 61, 297–304. [Google Scholar] [CrossRef] [PubMed]
- Chen, D.S.; Mellman, I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013, 39, 1–10. [Google Scholar] [CrossRef]
- Shae, D.; Becker, K.W.; Christov, P.; Yun, D.S.; Lytton-Jean, A.K.R.; Sevimli, S.; Ascano, M.; Kelley, M.; Johnson, D.B.; Balko, J.M.; et al. Endosomolytic polymersomes increase the activity of cyclic dinucleotide STING agonists to enhance cancer immunotherapy. Nat. Nanotechnol. 2019, 14, 269–278. [Google Scholar] [CrossRef]
- Guedan, S.; Ruella, M.; June, C.H. Emerging Cellular Therapies for Cancer. Annu. Rev. Immunol. 2019, 37, 145–171. [Google Scholar] [CrossRef]
- Melero, I.; Berman, D.M.; Aznar, M.A.; Korman, A.J.; Gracia, J.L.P.; Haanen, J. Evolving synergistic combinations of targeted immunotherapies to combat cancer. Nat. Rev. Cancer 2015, 15, 457–472. [Google Scholar] [CrossRef]
- Palucka, A.K.; Coussens, L.M. The Basis of Oncoimmunology. Cell 2016, 164, 1233–1247. [Google Scholar] [CrossRef]
- Wu, X.; Thi, V.L.D.; Huang, Y.; Billerbeck, E.; Saha, D.; Hoffmann, H.-H.; Wang, Y.; Silva, L.A.V.; Sarbanes, S.; Sun, T.; et al. Intrinsic Immunity Shapes Viral Resistance of Stem Cells. Cell 2018, 172, 423–438.e25. [Google Scholar] [CrossRef]
- Gatti-Mays, M.E.; Redman, J.M.; Collins, J.M.; Bilusic, M. Cancer vaccines: Enhanced immunogenic modulation through therapeutic combinations. Hum. Vaccines Immunother. 2017, 13, 2561–2574. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Liu, D.; Che, X.; Wang, X.; Ma, C.; Wu, G. Tumor Vaccines: Unleashing the Power of the Immune System to Fight Cancer. Pharmaceuticals 2023, 16, 1384. https://doi.org/10.3390/ph16101384
Liu D, Che X, Wang X, Ma C, Wu G. Tumor Vaccines: Unleashing the Power of the Immune System to Fight Cancer. Pharmaceuticals. 2023; 16(10):1384. https://doi.org/10.3390/ph16101384
Chicago/Turabian StyleLiu, Dequan, Xiangyu Che, Xiaoxi Wang, Chuanyu Ma, and Guangzhen Wu. 2023. "Tumor Vaccines: Unleashing the Power of the Immune System to Fight Cancer" Pharmaceuticals 16, no. 10: 1384. https://doi.org/10.3390/ph16101384
APA StyleLiu, D., Che, X., Wang, X., Ma, C., & Wu, G. (2023). Tumor Vaccines: Unleashing the Power of the Immune System to Fight Cancer. Pharmaceuticals, 16(10), 1384. https://doi.org/10.3390/ph16101384