Therapeutic Vaccines for Hematological Cancers: A Scoping Review of This Immunotherapeutic Approach as Alternative to the Treatment of These Malignancies
Abstract
:1. Introduction
2. Materials and Methods
- ➢
- Pubmed: Free Full text, Full Text, 10 years;
- ➢
- Science Direct: Title, abstract or author-specified keywords, 10 years;
- ➢
- Scopus: Title, abstract or author-specified keywords, 10 years;
- ➢
- Web of Science: Open Access, Article, English, 10 years.
3. Results and Discussion
3.1. Pathology
3.2. Vaccine Formulation, Therapeutic Approaches, and Study Follow-Up
3.3. Study Stage Development
3.4. Assays and Techniques Employed to Vaccine Evaluation
3.4.1. Preclinical Studies Overview
3.4.2. Clinical Studies Overview
3.5. Vaccine Treatment Outcomes
3.6. Study Limitations
3.7. Restrictions of the Scoping Review Process
4. Conclusions and Perspectives
Supplementary Materials
Author Contributions
Funding
Conflicts of Interest
References
- Zhang, N.; Wu, J.; Wang, Q.; Liang, Y.; Li, X.; Chen, G.; Ma, L.; Liu, X.; Zhou, F. Global Burden of Hematologic Malignancies and Evolution Patterns over the Past 30 Years. Blood Cancer J. 2023, 13, 82. [Google Scholar] [CrossRef]
- Dieguez, G.; Ferro, C.; Rotter, D. The Cost Burden of Blood Cancer Care. A Longitudinal Analysis of Commercially Insured Patients Diagnosed with Blood Cancer; Milliman: Seattle, WA, USA, 2018. [Google Scholar]
- Freireich, E.J. The Society of Hematologic Oncology (SOHO): Moving Forward in the Battle against Hematologic Malignancies. Clin. Lymphoma Myeloma Leuk. 2012, 12, 385. [Google Scholar] [CrossRef] [PubMed]
- Tang, L.; Huang, Z.; Mei, H.; Hu, Y. Immunotherapy in Hematologic Malignancies: Achievements, Challenges and Future Prospects. Signal Transduct. Target. Ther. 2023, 8, 306. [Google Scholar] [CrossRef] [PubMed]
- Olson, B.; Li, Y.; Lin, Y.; Liu, E.T.; Patnaik, A. Mouse Models for Cancer Immunotherapy Research. Cancer Discov. 2018, 8, 1358–1365. [Google Scholar] [CrossRef] [PubMed]
- Sweeney, C.; Vyas, P. The Graft-Versus-Leukemia Effect in AML. Front. Oncol. 2019, 9, 1217. [Google Scholar] [CrossRef]
- Piesche, M.; Ho, V.T.; Kim, H.; Nakazaki, Y.; Nehil, M.; Yaghi, N.K.; Kolodin, D.; Weiser, J.; Altevogt, P.; Kiefel, H.; et al. Angiogenic Cytokines Are Antibody Targets during Graft-versus-Leukemia Reactions. Clin. Cancer Res. 2015, 21, 1010. [Google Scholar] [CrossRef]
- Warren, E.H.; Deeg, H.J. Dissecting Graft-versus-Leukemia from Graft-versus-Host-Disease Using Novel Strategies. Tissue Antigens 2013, 81, 183–193. [Google Scholar] [CrossRef]
- Biavati, L.; Noonan, K.; Luznik, L.; Borrello, I. Activated Allogeneic Donor-Derived Marrow-Infiltrating Lymphocytes Display Measurable In Vitro Antitumor Activity. J. Immunother. 2019, 42, 73. [Google Scholar] [CrossRef]
- Peres, C.; Matos, A.I.; Moura, L.I.F.; Acúrcio, R.C.; Carreira, B.; Pozzi, S.; Vaskovich-Koubi, D.; Kleiner, R.; Satchi-Fainaro, R.; Florindo, H.F. Preclinical Models and Technologies to Advance Nanovaccine Development. Adv. Drug Deliv. Rev. 2021, 172, 148–182. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Slezak, A.J.; Chang, K.; Beckman, T.N.; Refvik, K.C.; Alpar, A.T.; Lauterbach, A.L.; Solanki, A.; Kwon, J.W.; Gomes, S.; Mansurov, A.; et al. Cysteine-Binding Adjuvant Enhances Survival and Promotes Immune Function in a Murine Model of Acute Myeloid Leukemia. Blood Adv. 2024, 8, 1747–1759. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Ceppi, M.; Hettich, M.; Teichgräber, V.; Driessen, W.; Tuerck, D.; Marrer-Berger, E.; Evers, S.; Crameri, F.; Gomes, B.; Bachl, J.; et al. Tumor-Bearing Non-Human Primates: An Unrivaled Model for Translational Cancer Immunology Research. Cancer Res. 2020, 80, 6135. [Google Scholar] [CrossRef]
- Hoos, A.; Parmiani, G.; Hege, K.; Sznol, M.; Loibner, H.; Eggermont, A.; Urba, W.; Blumenstein, B.; Sacks, N.; Keilholz, U.; et al. A Clinical Development Paradigm for Cancer Vaccines and Related Biologics. J. Immunother. 2007, 30, 1–15. [Google Scholar] [CrossRef]
- Kudrin, A. Cancer Vaccines: What Do We Need to Measure in Clinical Trials? Hum. Vaccin. Immunother. 2014, 10, 3236–3240. [Google Scholar] [CrossRef]
- Tricco, A.C.; Lillie, E.; Zarin, W.; O’Brien, K.K.; Colquhoun, H.; Levac, D.; Moher, D.; Peters, M.D.J.; Horsley, T.; Weeks, L.; et al. PRISMA Extension for Scoping Reviews (PRISMA-ScR): Checklist and Explanation. Ann. Intern. Med. 2018, 169, 467–473. [Google Scholar] [CrossRef]
- Hardwick, N.; Buchan, S.; Ingram, W.; Khan, G.; Vittes, G.; Rice, J.; Pulford, K.; Mufti, G.; Stevenson, F.; Guinn, B.A. An Analogue Peptide from the Cancer/Testis Antigen PASD1 Induces CD8+ T Cell Responses against Naturally Processed Peptide. Cancer Immun. 2013, 13, 16. [Google Scholar]
- Vincent, K.; Hardy, M.P.; Trofimov, A.; Laumont, C.M.; Sriranganadane, D.; Hadj-Mimoune, S.; Salem Fourati, I.; Soudeyns, H.; Thibault, P.; Perreault, C. Rejection of Leukemic Cells Requires Antigen-Specific T Cells with High Functional Avidity. Biol. Blood Marrow Transpl. 2014, 20, 37–45. [Google Scholar] [CrossRef]
- Uttenthal, B.; Martinez-Davila, I.; Ivey, A.; Craddock, C.; Chen, F.; Virchis, A.; Kottaridis, P.; Grimwade, D.; Khwaja, A.; Stauss, H.; et al. Wilms’ Tumour 1 (WT1) Peptide Vaccination in Patients with Acute Myeloid Leukaemia Induces Short-Lived WT1-Specific Immune Responses. Br. J. Haematol. 2014, 164, 366–375. [Google Scholar] [CrossRef]
- Li, R.; Qian, J.; Zhang, W.; Fu, W.; Du, J.; Jiang, H.; Zhang, H.; Zhang, C.; Xi, H.; Yi, Q.; et al. Human Heat Shock Protein-Specific Cytotoxic T Lymphocytes Display Potent Antitumour Immunity in Multiple Myeloma. Br. J. Haematol. 2014, 166, 690–701. [Google Scholar] [CrossRef]
- Yao, Y.; Wang, C.; Wei, W.; Shen, C.; Deng, X.; Chen, L.; Ma, L.; Hao, S. Dendritic Cells Pulsed with Leukemia Cell-Derived Exosomes More Efficiently Induce Antileukemic Immunities. PLoS ONE 2014, 9, e91463. [Google Scholar] [CrossRef] [PubMed]
- Suehiro, Y.; Hasegawa, A.; Iino, T.; Sasada, A.; Watanabe, N.; Matsuoka, M.; Takamori, A.; Tanosaki, R.; Utsunomiya, A.; Choi, I.; et al. Clinical Outcomes of a Novel Therapeutic Vaccine with Tax Peptide-Pulsed Dendritic Cells for Adult T Cell Leukaemia/Lymphoma in a Pilot Study. Br. J. Haematol. 2015, 169, 356–367. [Google Scholar] [CrossRef] [PubMed]
- McCann, K.J.; Godeseth, R.; Chudley, L.; Mander, A.; Di Genova, G.; Lloyd-Evans, P.; Kerr, J.P.; Malykh, V.B.; Jenner, M.W.; Orchard, K.H.; et al. Idiotypic DNA Vaccination for the Treatment of Multiple Myeloma: Safety and Immunogenicity in a Phase I Clinical Study. Cancer Immunol. Immunother. 2015, 64, 1021–1032. [Google Scholar] [CrossRef] [PubMed]
- Le Pogam, C.; Patel, S.; Gorombei, P.; Guerenne, L.; Krief, P.; Omidvar, N.; Tekin, N.; Bernasconi, E.; Sicre, F.; Schlageter, M.-H.; et al. DNA-Mediated Adjuvant Immunotherapy Extends Survival in Two Different Mouse Models of Myeloid Malignancies. Oncotarget 2015, 6, 32494. [Google Scholar] [CrossRef]
- Kobayashi, T.; Doff, B.L.; Rearden, R.C.; Leggatt, G.R.; Mattarollo, S.R. NKT Cell-Targeted Vaccination plus Anti-4–1BB Antibody Generates Persistent CD8 T Cell Immunity against B Cell Lymphoma. Oncoimmunology 2015, 4, e990793. [Google Scholar] [CrossRef]
- Sugata, K.; Yasunaga, J.I.; Mitobe, Y.; Miura, M.; Miyazato, P.; Kohara, M.; Matsuoka, M. Protective Effect of Cytotoxic T Lymphocytes Targeting HTLV-1 BZIP Factor. Blood 2015, 126, 1095–1105. [Google Scholar] [CrossRef]
- Manlove, L.S.; Schenkel, J.M.; Manlove, K.R.; Pauken, K.E.; Williams, R.T.; Vezys, V.; Farrar, M.A. Heterologous Vaccination and Checkpoint Blockade Synergize to Induce Anti-Leukemia Immunity. J. Immunol. 2016, 196, 4793. [Google Scholar] [CrossRef]
- Willemen, Y.; Van den Bergh, J.M.J.; Bonte, S.M.; Anguille, S.; Heirman, C.; Stein, B.M.H.; Goossens, H.; Kerre, T.; Thielemans, K.; Peeters, M.; et al. The Tumor-Associated Antigen RHAMM (HMMR/CD168) Is Expressed by Monocyte-Derived Dendritic Cells and Presented to T Cells. Oncotarget 2016, 7, 73960. [Google Scholar] [CrossRef]
- Shah, N.N.; Loeb, D.M.; Khuu, H.; Stroncek, D.; Ariyo, T.; Raffeld, M.; Delbrook, C.; Mackall, C.L.; Wayne, A.S.; Fry, T.J. Induction of Immune Response after Allogeneic Wilms’ Tumor 1 Dendritic Cell Vaccination and Donor Lymphocyte Infusion in Patients with Hematologic Malignancies and Post-Transplantation Relapse. Biol. Blood Marrow Transpl. 2016, 22, 2149. [Google Scholar] [CrossRef]
- Khoury, H.J.; Collins, R.H.; Blum, W.; Stiff, P.S.; Elias, L.; Lebkowski, J.S.; Reddy, A.; Nishimoto, K.P.; Sen, D.; Wirth, E.D.; et al. Immune Responses and Long-Term Disease Recurrence Status after Telomerase-Based Dendritic Cell Immunotherapy in Patients with Acute Myeloid Leukemia. Cancer 2017, 123, 3061–3072. [Google Scholar] [CrossRef]
- Huang, F.; Wan, J.; Hu, W.; Hao, S. Enhancement of Anti-Leukemia Immunity by Leukemia–Derived Exosomes Via Downregulation of TGF-Β1 Expression. Cell. Physiol. Biochem. 2018, 44, 240–254. [Google Scholar] [CrossRef] [PubMed]
- Kobayashi, Y.; Sakura, T.; Miyawaki, S.; Toga, K.; Sogo, S.; Heike, Y. A New Peptide Vaccine OCV-501: In Vitro Pharmacology and Phase 1 Study in Patients with Acute Myeloid Leukemia. Cancer Immunol. Immunother. 2017, 66, 851. [Google Scholar] [CrossRef]
- Chen, J.; Zhang, M.; Zhou, F.; Wang, J.; Niu, B.; Zhang, W.G. Immunological Effects of Vaccines Combined with Granulocyte Colony-Stimulating Factor on a Murine WEHI-3 Leukemia Model. Oncol. Lett. 2017, 13, 2323–2329. [Google Scholar] [CrossRef] [PubMed]
- Lu, C.; Meng, S.; Jin, Y.; Zhang, W.; Li, Z.; Wang, F.; Wang-Johanning, F.; Wei, Y.; Liu, H.; Tu, H.; et al. A Novel Multi-Epitope Vaccine from MMSA-1 and DKK1 for Multiple Myeloma Immunotherapy. Br. J. Haematol. 2017, 178, 413–426. [Google Scholar] [CrossRef]
- Montico, B.; Lapenta, C.; Ravo, M.; Martorelli, D.; Muraro, E.; Zeng, B.; Comaro, E.; Spada, M.; Donati, S.; Santini, S.M.; et al. Exploiting a New Strategy to Induce Immunogenic Cell Death to Improve Dendritic Cell-Based Vaccines for Lymphoma Immunotherapy. Oncoimmunology 2017, 6, e1356964. [Google Scholar] [CrossRef]
- Anguille, S.; Van De Velde, A.L.; Smits, E.L.; Van Tendeloo, V.F.; Juliusson, G.; Cools, N.; Nijs, G.; Stein, B.; Lion, E.; Van Driessche, A.; et al. Dendritic Cell Vaccination as Postremission Treatment to Prevent or Delay Relapse in Acute Myeloid Leukemia. Blood 2017, 130, 1713. [Google Scholar] [CrossRef]
- Pradhan, P.; Leleux, J.; Liu, J.; Roy, K. A Simple, Clinically Relevant Therapeutic Vaccine Shows Long-Term Protection in an Aggressive, Delayed-Treatment B Lymphoma Model. JCI Insight 2017, 2, e92522. [Google Scholar] [CrossRef]
- Ho, V.T.; Kim, H.T.; Bavli, N.; Mihm, M.; Pozdnyakova, O.; Piesche, M.; Daley, H.; Reynolds, C.; Souders, N.C.; Cutler, C.; et al. Vaccination with Autologous Myeloblasts Admixed with GM-K562 Cells in Patients with Advanced MDS or AML after Allogeneic HSCT. Blood Adv. 2017, 1, 2269. [Google Scholar] [CrossRef]
- Ueda, Y.; Ogura, M.; Miyakoshi, S.; Suzuki, T.; Heike, Y.; Tagashira, S.; Tsuchiya, S.; Ohyashiki, K.; Miyazaki, Y. Phase 1/2 Study of the WT1 Peptide Cancer Vaccine WT4869 in Patients with Myelodysplastic Syndrome. Cancer Sci. 2017, 108, 2445–2453. [Google Scholar] [CrossRef]
- Liu, T.T.; Wu, Y.; Niu, T. Human DKK1 and Human HSP70 Fusion DNA Vaccine Induces an Effective Anti-Tumor Efficacy in Murine Multiple Myeloma. Oncotarget 2017, 9, 178–191. [Google Scholar] [CrossRef]
- Nakata, J.; Nakajima, H.; Hayashibara, H.; Imafuku, K.; Morimoto, S.; Fujiki, F.; Motooka, D.; Okuzaki, D.; Hasegawa, K.; Hosen, N.; et al. Extremely Strong Infiltration of WT1-Specific CTLs into Mouse Tumor by the Combination Vaccine with WT1-Specific CTL and Helper Peptides. Oncotarget 2018, 9, 36029. [Google Scholar] [CrossRef] [PubMed]
- Maslak, P.G.; Dao, T.; Bernal, Y.; Chanel, S.M.; Zhang, R.; Frattini, M.; Rosenblat, T.; Jurcic, J.G.; Brentjens, R.J.; Arcila, M.E.; et al. Phase 2 Trial of a Multivalent WT1 Peptide Vaccine (Galinpepimut-S) in Acute Myeloid Leukemia. Blood Adv. 2018, 2, 224. [Google Scholar] [CrossRef]
- van de Loosdrecht, A.A.; van Wetering, S.; Santegoets, S.J.A.M.; Singh, S.K.; Eeltink, C.M.; den Hartog, Y.; Koppes, M.; Kaspers, J.; Ossenkoppele, G.J.; Kruisbeek, A.M.; et al. A Novel Allogeneic Off-the-Shelf Dendritic Cell Vaccine for Post-Remission Treatment of Elderly Patients with Acute Myeloid Leukemia. Cancer Immunol. Immunother. 2018, 67, 1505. [Google Scholar] [CrossRef]
- Kerage, D.; Soon, M.S.F.; Doff, B.L.; Kobayashi, T.; Nissen, M.D.; Lam, P.Y.; Leggatt, G.R.; Mattarollo, S.R. Therapeutic Vaccination with 4–1BB Co-Stimulation Eradicates Mouse Acute Myeloid Leukemia. Oncoimmunology 2018, 7, e1486952. [Google Scholar] [CrossRef]
- Shi, Y.; Dincheva-Vogel, L.; Ayemoba, C.E.; Fung, J.P.; Bergamaschi, C.; Pavlakis, G.N.; Farzaneh, F.; Gaensler, K.M.L. IL-15/IL-15Rα/CD80-Expressing AML Cell Vaccines Eradicate Minimal Residual Disease in Leukemic Mice. Blood Adv. 2018, 2, 3177–3192. [Google Scholar] [CrossRef]
- Liu, H.; Zha, Y.; Choudhury, N.; Malnassy, G.; Fulton, N.; Green, M.; Park, J.H.; Nakamura, Y.; Larson, R.A.; Salazar, A.M.; et al. WT1 Peptide Vaccine in Montanide in Contrast to Poly ICLC, Is Able to Induce WT1-Specific Immune Response with TCR Clonal Enrichment in Myeloid Leukemia. Exp. Hematol. Oncol. 2018, 7, 1. [Google Scholar] [CrossRef]
- Chen, Z.; You, L.; Wang, L.; Huang, X.; Liu, H.; Wei, J.Y.; Zhu, L.; Qian, W. Dual Effect of DLBCL-Derived EXOs in Lymphoma to Improve DC Vaccine Efficacy in Vitro While Favor Tumorgenesis in Vivo. J. Exp. Clin. Cancer Res. 2018, 37, 190. [Google Scholar] [CrossRef]
- Robinson, T.M.; Prince, G.T.; Thoburn, C.; Warlick, E.; Ferguson, A.; Kasamon, Y.L.; Borrello, I.M.; Hess, A.; Smith, B.D. Pilot Trial of K562/GM-CSF Whole-Cell Vaccination in MDS Patients. Leuk. Lymphoma 2018, 59, 2801–2811. [Google Scholar] [CrossRef]
- Thomas, S.K.; Cha, S.-C.; Smith, D.L.; Kim, K.H.; Parshottam, S.R.; Rao, S.; Popescu, M.; Lee, V.Y.; Neelapu, S.S.; Kwak, L.W. Phase I Study of an Active Immunotherapy for Asymptomatic Phase Lymphoplasmacytic Lymphoma with DNA Vaccines Encoding Antigen-Chemokine Fusion: Study Protocol. BMC Cancer 2018, 18, 187. [Google Scholar] [CrossRef]
- Wang, D.; Huang, X.F.; Hong, B.; Song, X.T.; Hu, L.; Jiang, M.; Zhang, B.; Ning, H.; Li, Y.; Xu, C.; et al. Efficacy of Intracellular Immune Checkpoint-Silenced DC Vaccine. JCI Insight 2018, 3, e98368. [Google Scholar] [CrossRef]
- Martínez-Torres, A.C.; Calvillo-Rodríguez, K.M.; Uscanga-Palomeque, A.C.; Gómez-Morales, L.; Mendoza-Reveles, R.; Caballero-Hernández, D.; Karoyan, P.; Rodríguez-Padilla, C. PKHB1 Tumor Cell Lysate Induces Antitumor Immune System Stimulation and Tumor Regression in Syngeneic Mice with Tumoral T Lymphoblasts. J. Oncol. 2019, 2019, 9852361. [Google Scholar] [CrossRef] [PubMed]
- Shin, A.-R.; Lee, S.-E.; Choi, H.; Sohn, H.-J.; Cho, H.-I.; Kim, T.-G. An Effective Peptide Vaccine Strategy Circumventing Clonal MHC Heterogeneity of Murine Myeloid Leukaemia. Br. J. Cancer 2020, 123, 919–931. [Google Scholar] [CrossRef] [PubMed]
- Lichtenegger, F.S.; Schnorfeil, F.M.; Rothe, M.; Deiser, K.; Altmann, T.; Bücklein, V.L.; Köhnke, T.; Augsberger, C.; Konstandin, N.P.; Spiekermann, K.; et al. Toll-like Receptor 7/8-Matured RNA-Transduced Dendritic Cells as Post-Remission Therapy in Acute Myeloid Leukaemia: Results of a Phase I Trial. Clin. Transl. Immunol. 2020, 9, e1117. [Google Scholar] [CrossRef] [PubMed]
- Hu, W.; Huang, F.; Ning, L.; Hao, J.; Wan, J.; Hao, S. Enhanced Immunogenicity of Leukemia-Derived Exosomes via Transfection with Lentiviral Vectors Encoding Costimulatory Molecules. Cell. Oncol. 2020, 43, 889–900. [Google Scholar] [CrossRef] [PubMed]
- Zhong, G.; Jin, G.; Zeng, W.; Yu, C.; Li, Y.; Zhou, J.; Zhang, L.; Yu, L. Conjugation of TLR7 Agonist Combined with Demethylation Treatment Improves Whole-Cell Tumor Vaccine Potency in Acute Myeloid Leukemia. Int. J. Med. Sci. 2020, 17, 2346–2356. [Google Scholar] [CrossRef]
- Habault, J.; Kaci, A.; Pasquereau-Kotula, E.; Fraser, C.; Chomienne, C.; Dombret, H.; Braun, T.; Pla, M.; Poyet, J.L. Prophylactic and Therapeutic Antileukemic Effects Induced by the AAC-11-Derived Peptide RT53. Oncoimmunology 2020, 9, 1728871. [Google Scholar] [CrossRef]
- Jørgensen, N.G.; Klausen, U.; Grauslund, J.H.; Helleberg, C.; Aagaard, T.G.; Do, T.H.; Ahmad, S.M.; Olsen, L.R.; Klausen, T.W.; Breinholt, M.F.; et al. Peptide Vaccination Against PD-L1 With IO103 a Novel Immune Modulatory Vaccine in Multiple Myeloma: A Phase I First-in-Human Trial. Front. Immunol. 2020, 11, 595035. [Google Scholar] [CrossRef]
- Johnson, D.T.; Zhou, J.; Kroll, A.V.; Fang, R.H.; Yan, M.; Xiao, C.; Chen, X.; Kline, J.; Zhang, L.; Zhang, D.E. Acute Myeloid Leukemia Cell Membrane-Coated Nanoparticles for Cancer Vaccination Immunotherapy. Leukemia 2022, 36, 994–1005. [Google Scholar] [CrossRef]
- Almshayakhchi, R.; Nagarajan, D.; Vadakekolathu, J.; Guinn, B.A.; Reeder, S.; Brentville, V.; Metheringham, R.; Pockley, A.G.; Durrant, L.; McArdle, S. A Novel HAGE/WT1-ImmunoBody® Vaccine Combination Enhances Anti-Tumour Responses When Compared to Either Vaccine Alone. Front. Oncol. 2021, 11, 636977. [Google Scholar] [CrossRef]
- Stroopinsky, D.; Liegel, J.; Bhasin, M.; Cheloni, G.; Thomas, B.; Bhasin, S.; Panchal, R.; Ghiasuddin, H.; Rahimian, M.; Nahas, M.; et al. Leukemia Vaccine Overcomes Limitations of Checkpoint Blockade by Evoking Clonal T-Cell Responses in a Murine Acute Myeloid Leukemia Model. Haematologica 2021, 106, 1330. [Google Scholar] [CrossRef]
- Rovida, A.; Maccalli, C.; Scarfo, L.; Dellabona, P.; Stamatopoulos, K.; Ghia, P. Exploiting B-Cell Receptor Stereotypy to Design Tailored Immunotherapy in Chronic Lymphocytic Leukemia. Clin. Cancer Res. 2021, 27, 729–739. [Google Scholar] [CrossRef] [PubMed]
- Webster, J.A.; Robinson, T.M.; Blackford, A.L.; Warlick, E.; Ferguson, A.; Borrello, I.; Zahurak, M.; Jones, R.J.; Smith, B.D. A Randomized, Phase II Trial of Adjuvant Immunotherapy with Durable TKI-Free Survival in Patients with Chronic Phase CML. Leuk. Res. 2021, 111, 106737. [Google Scholar] [CrossRef] [PubMed]
- Tusup, M.; Läuchli, S.; Jarzebska, N.T.; French, L.E.; Chang, Y.T.; Vonow-Eisenring, M.; Su, A.; Kündig, T.M.; Guenova, E.; Pascolo, S. MRNA-Based Anti-TCR CDR3 Tumour Vaccine for T-Cell Lymphoma. Pharmaceutics 2021, 13, 1040. [Google Scholar] [CrossRef] [PubMed]
- Fujii, S.-I.; Kawamata, T.; Shimizu, K.; Nakabayashi, J.; Yamasaki, S.; Iyoda, T.; Shinga, J.; Nakazato, H.; Sanpei, A.; Kawamura, M.; et al. Reinvigoration of Innate and Adaptive Immunity via Therapeutic Cellular Vaccine for Patients with AML. Mol. Ther. Oncolytics 2022, 27, 315–332. [Google Scholar] [CrossRef]
- Klausen, U.; Grauslund, J.H.; Jørgensen, N.G.D.; Ahmad, S.M.; Jonassen, M.; Weis-Banke, S.E.; Martinenaite, E.; Pedersen, L.B.; Lisle, T.L.; Gang, A.O.; et al. Anti-PD-L1/PD-L2 Therapeutic Vaccination in Untreated Chronic Lymphocytic Leukemia Patients with Unmutated IgHV. Front. Oncol. 2022, 12, 1023015. [Google Scholar] [CrossRef]
- Su, H.; Imai, K.; Jia, W.; Li, Z.; DiCioccio, R.A.; Serody, J.S.; Poe, J.C.; Chen, B.J.; Doan, P.L.; Sarantopoulos, S. Alphavirus Replicon Particle Vaccine Breaks B Cell Tolerance and Rapidly Induces IgG to Murine Hematolymphoid Tumor Associated Antigens. Front. Immunol. 2022, 13, 865486. [Google Scholar] [CrossRef]
- Du, Z.; Huang, Z.; Chen, X.; Jiang, G.; Peng, Y.; Feng, W.; Huang, N. Modified Dendritic Cell-Derived Exosomes Activate Both NK Cells and T Cells through the NKG2D/NKG2D-L Pathway to Kill CML Cells with or without T315I Mutation. Exp. Hematol. Oncol. 2022, 11, 36. [Google Scholar] [CrossRef]
- Kreutmair, S.; Pfeifer, D.; Waterhouse, M.; Takács, F.; Graessel, L.; Döhner, K.; Duyster, J.; Illert, A.L.; Frey, A.V.; Schmitt, M.; et al. First-in-Human Study of WT1 Recombinant Protein Vaccination in Elderly Patients with AML in Remission: A Single-Center Experience. Cancer Immunol. Immunother. 2022, 71, 2913–2928. [Google Scholar] [CrossRef]
- Minagawa, H.; Hashii, Y.; Nakajima, H.; Fujiki, F.; Morimoto, S.; Nakata, J.; Shirakawa, T.; Katayama, T.; Tsuboi, A.; Ozono, K. Enhanced Antitumor Activity of a Novel, Oral, Helper Epitope-Containing WT1 Protein Vaccine in a Model of Murine Leukemia. BMC Cancer 2023, 23, 167. [Google Scholar] [CrossRef]
- Nakagawa, N.; Hashii, Y.; Kayama, H.; Okumura, R.; Nakajima, H.; Minagawa, H.; Morimoto, S.; Fujiki, F.; Nakata, J.; Shirakawa, T.; et al. An Oral WT1 Protein Vaccine Composed of WT1-Anchored, Genetically Engineered Bifidobacterium Longum Allows for Intestinal Immunity in Mice with Acute Myeloid Leukemia. Cancer Immunol. Immunother. 2023, 72, 39–53. [Google Scholar] [CrossRef]
- Shimizu, K.; Ueda, S.; Kawamura, M.; Aoshima, H.; Satoh, M.; Nakabayashi, J.; Fujii, S.I. Combination of Cancer Vaccine with CD122-Biased IL-2/Anti-IL-2 Ab Complex Shapes the Stem-like Effector NK and CD8+ T Cells against Tumor. J. Immunother. Cancer 2023, 11, e006409. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Zhou, M.; Wang, L.; Huang, L.; Chen, X.; Sun, X.; Liu, T. Evaluation of the Safety and Efficiency of Cytotoxic T Cell Therapy Sensitized by Tumor Antigens Original from T-ALL-iPSC in Vivo. Cancer Innov. 2023, 3, e95. [Google Scholar] [CrossRef] [PubMed]
- Preisler, H.D. The leukemias. Disease-a-Month 1994, 40, 529–579. [Google Scholar] [CrossRef]
- Pelcovits, A.; Niroula, R. Acute Myeloid Leukemia: A Review. Rhode Isl. Med. J. 2020, 103, 38–40. [Google Scholar]
- Liquori, A.; Ibañez, M.; Sargas, C.; Sanz, M.Á.; Barragán, E.; Cervera, J. Acute Promyelocytic Leukemia: A Constellation of Molecular Events around a Single PML-RARA Fusion Gene. Cancers 2020, 12, 624. [Google Scholar] [CrossRef]
- Iyer, S.G.; Elias, L.; Stanchina, M.; Watts, J. The Treatment of Acute Promyelocytic Leukemia in 2023: Paradigm, Advances, and Future Directions. Front. Oncol. 2023, 12, 1062524. [Google Scholar] [CrossRef]
- Harrison, C.J. Acute Lymphoblastic Leukemia. Clin. Lab. Med. 2011, 31, 631–647. [Google Scholar] [CrossRef]
- Aureli, A.; Marziani, B.; Venditti, A.; Sconocchia, T.; Sconocchia, G. Acute Lymphoblastic Leukemia Immunotherapy Treatment: Now, Next, and Beyond. Cancers 2023, 15, 3346. [Google Scholar] [CrossRef]
- Mojtahedi, H.; Yazdanpanah, N.; Rezaei, N. Chronic Myeloid Leukemia Stem Cells: Targeting Therapeutic Implications. Stem Cell Res. Ther. 2021, 12, 603. [Google Scholar] [CrossRef]
- Rinaldi, I.; Winston, K. Chronic Myeloid Leukemia, from Pathophysiology to Treatment-Free Remission: A Narrative Literature Review. J. Blood Med. 2023, 14, 261–277. [Google Scholar] [CrossRef]
- Hallek, M. Chronic Lymphocytic Leukemia: 2020 Update on Diagnosis, Risk Stratification and Treatment. Am. J. Hematol. 2019, 94, 1266–1287. [Google Scholar] [CrossRef] [PubMed]
- Griggio, V.; Perutelli, F.; Salvetti, C.; Boccellato, E.; Boccadoro, M.; Vitale, C.; Coscia, M. Immune Dysfunctions and Immune-Based Therapeutic Interventions in Chronic Lymphocytic Leukemia. Front. Immunol. 2020, 11, 594556. [Google Scholar] [CrossRef] [PubMed]
- Kumar, S.K.; Rajkumar, V.; Kyle, R.A.; Van Duin, M.; Sonneveld, P.; Mateos, M.V.; Gay, F.; Anderson, K.C. Multiple Myeloma. Nat. Rev. Dis. Prim. 2017, 3, 17046. [Google Scholar] [CrossRef]
- Gulla, A.; Morelli, E.; Samur, M.K.; Botta, C.; Hideshima, T.; Bianchi, G.; Fulciniti, M.; Malvestiti, S.; Prabhala, R.H.; Talluri, S.; et al. Bortezomib Induces Anti-Multiple Myeloma Immune Response Mediated by CGAS/STING Pathway Activation. Blood Cancer Discov. 2021, 2, 468–483. [Google Scholar] [CrossRef]
- Lin, P.; Ren, Y.; Yan, X.; Luo, Y.; Zhang, H.; Kesarwani, M.; Bu, J.; Zhan, D.; Zhou, Y.; Tang, Y.; et al. The High NRF2 Expression Confers Chemotherapy Resistance Partly through Up-Regulated DUSP1 in Myelodysplastic Syndromes. Haematologica 2019, 104, 485–496. [Google Scholar] [CrossRef]
- Hellström-Lindberg, E.; Tobiasson, M.; Greenberg, P. Myelodysplastic Syndromes: Moving towards Personalized Management. Haematologica 2020, 105, 1765–1779. [Google Scholar] [CrossRef]
- Mugnaini, E.N.; Ghosh, N. Lymphoma. Prim. Care 2016, 43, 661–675. [Google Scholar] [CrossRef]
- Jamil, A.; Mukkamalla, S.K.R. Lymphoma. In Essential Paediatric Surgery: A Practical Guide; CRC Press: Boca Raton, FL, USA, 2023; pp. 344–347. [Google Scholar] [CrossRef]
- Che, Y.; Ding, X.; Xu, L.; Zhao, J.; Zhang, X.; Li, N.; Sun, X. Advances in the Treatment of Hodgkin’s Lymphoma (Review). Int. J. Oncol. 2023, 62, 61. [Google Scholar] [CrossRef]
- Schnell, S.; Knierim, E.; Bittigau, P.; Kreye, J.; Hauptmann, K.; Hundsdoerfer, P.; Morales-Gonzalez, S.; Schuelke, M.; Nikolaus, M. Hodgkin Lymphoma Cell Lines and Tissues Express MGluR5: A Potential Link to Ophelia Syndrome and Paraneoplastic Neurological Disease. Cells 2023, 12, 606. [Google Scholar] [CrossRef]
- Moubadder, L.; McCullough, L.E.; Flowers, C.R.; Koff, J.L. Linking Environmental Exposures to Molecular Pathogenesis in Non-Hodgkin Lymphoma Subtypes. Cancer Epidemiol. Biomark. Prev. 2020, 29, 1844–1855. [Google Scholar] [CrossRef]
- Francisco, L.F.V.; da Silva, R.N.; Oliveira, M.A.; dos Santos Neto, M.F.; Gonçalves, I.Z.; Marques, M.M.C.; Silveira, H.C.S. Occupational Exposures and Risks of Non-Hodgkin Lymphoma: A Meta-Analysis. Cancers 2023, 15, 2600. [Google Scholar] [CrossRef] [PubMed]
- Cook, L.B.; Fuji, S.; Hermine, O.; Bazarbachi, A.; Ramos, J.C.; Ratner, L.; Horwitz, S.; Fields, P.; Tanase, A.; Bumbea, H.; et al. Revised Adult T-Cell Leukemia-Lymphoma International Consensus Meeting Report. J. Clin. Oncol. 2019, 37, 677–687. [Google Scholar] [CrossRef] [PubMed]
- Tsukasaki, K.; Marçais, A.; Nasr, R.; Kato, K.; Fukuda, T.; Hermine, O.; Bazarbachi, A. Diagnostic Approaches and Established Treatments for Adult T Cell Leukemia Lymphoma. Front. Microbiol. 2020, 11, 533939. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.J.; Tokumori, F.C.; Del Guzzo, C.; Kim, J.; Ruan, J. Update on T-Cell Lymphoma Epidemiology. Curr. Hematol. Malig. Rep. 2024, 19, 93–103. [Google Scholar] [CrossRef]
- Li, J.; Zou, J.; Wan, X.; Sun, C.; Peng, F.; Chu, Z.; Hu, Y. The Role of Noncoding RNAs in B-Cell Lymphoma. Front. Oncol. 2020, 10, 577890. [Google Scholar] [CrossRef]
- Armengol, M.; Santos, J.C.; Fernández-serrano, M.; Profitós-pelejà, N.; Ribeiro, M.L.; Roué, G. Immune-Checkpoint Inhibitors in B-Cell Lymphoma. Cancers 2021, 13, 214. [Google Scholar] [CrossRef]
- Bailey, N.G.; Elenitoba-Johnson, K.S.J. Impact of Genetics on Mature Lymphoid Leukemias and Lymphomas. Cold Spring Harb. Perspect. Med. 2020, 10, a035444. [Google Scholar] [CrossRef]
- Onaindia, A.; Santiago-Quispe, N.; Iglesias-Martinez, E.; Romero-Abrio, C. Molecular Update and Evolving Classification of Large B-Cell Lymphoma. Cancers 2021, 13, 3352. [Google Scholar] [CrossRef]
- Kaseb, H.; Gonzalez-Mosquera, L.F.; Parsi, M.; Mewawalla, P. Lymphoplasmacytic Lymphoma. In Atlas of Differential Diagnosis in Neoplastic Hematopathology; CRC Press: Boca Raton, FL, USA, 2023; pp. 275–286. [Google Scholar] [CrossRef]
- Fujii, K. New Therapies and Immunological Findings in Cutaneous T-Cell Lymphoma. Front. Oncol. 2018, 8, 198. [Google Scholar] [CrossRef]
- Ødum, A.W.F.; Geisler, C. Vitamin D in Cutaneous T-Cell Lymphoma. Cells 2024, 13, 503. [Google Scholar] [CrossRef]
- da Melo, A.R.S.; de Macêdo, L.S.; da Invenção, M.C.V.; de Moura, I.A.; da Gama, M.A.T.M.; de Melo, C.M.L.; Silva, A.J.D.; de Batista, M.V.A.; de Freitas, A.C. Third-Generation Vaccines: Features of Nucleic Acid Vaccines and Strategies to Improve Their Efficiency. Genes 2022, 13, 2287. [Google Scholar] [CrossRef] [PubMed]
- Lu, B.; Lim, J.M.; Yu, B.; Song, S.; Neeli, P.; Sobhani, N.; K, P.; Bonam, S.R.; Kurapati, R.; Zheng, J.; et al. The Next-Generation DNA Vaccine Platforms and Delivery Systems: Advances, Challenges and Prospects. Front. Immunol. 2024, 15, 1332939. [Google Scholar] [CrossRef]
- Zhang, G.; Tang, T.; Chen, Y.; Huang, X.; Liang, T. MRNA Vaccines in Disease Prevention and Treatment. Signal Transduct. Target. Ther. 2023, 8, 365. [Google Scholar] [CrossRef] [PubMed]
- Lee, K.-W.; Yam, J.W.P.; Lee, K.-W.; Wai, J.; Yam, P.; Mao, X. Dendritic Cell Vaccines: A Shift from Conventional Approach to New Generations. Cells 2023, 12, 2147. [Google Scholar] [CrossRef] [PubMed]
- Versteeg, L.; Almutairi, M.M.; Hotez, P.J.; Pollet, J. Enlisting the MRNA Vaccine Platform to Combat Parasitic Infections. Vaccines 2019, 7, 122. [Google Scholar] [CrossRef]
- Fu, C.; Zhou, L.; Mi, Q.S.; Jiang, A. DC-Based Vaccines for Cancer Immunotherapy. Vaccines 2020, 8, 706. [Google Scholar] [CrossRef]
- Kozak, M.; Hu, J. The Integrated Consideration of Vaccine Platforms, Adjuvants, and Delivery Routes for Successful Vaccine Development. Vaccines 2023, 11, 695. [Google Scholar] [CrossRef]
- Hajiaghapour Asr, M.; Dayani, F.; Saedi Segherloo, F.; Kamedi, A.; Neill, A.O.; MacLoughlin, R.; Doroudian, M. Lipid Nanoparticles as Promising Carriers for MRNA Vaccines for Viral Lung Infections. Pharmaceutics 2023, 15, 1127. [Google Scholar] [CrossRef]
- Sanmamed, M.F.; Chester, C.; Melero, I.; Kohrt, H. Defining the Optimal Murine Models to Investigate Immune Checkpoint Blockers and Their Combination with Other Immunotherapies. Ann. Oncol. 2016, 27, 1190–1198. [Google Scholar] [CrossRef]
- Spreafico, F.; Fernandez, C.V.; Brok, J.; Nakata, K.; Vujanic, G.; Geller, J.I.; Gessler, M.; Maschietto, M.; Behjati, S.; Polanco, A.; et al. Wilms Tumour. Nat. Rev. Dis. Primers 2021, 7, 75. [Google Scholar] [CrossRef]
- Seok, J.; Warren, H.S.; Alex, G.C.; Michael, N.M.; Henry, V.B.; Xu, W.; Richards, D.R.; McDonald-Smith, G.P.; Gao, H.; Hennessy, L.; et al. Genomic Responses in Mouse Models Poorly Mimic Human Inflammatory Diseases. Proc. Natl. Acad. Sci. USA 2013, 110, 3507–3512. [Google Scholar] [CrossRef]
- Takao, K.; Miyakawa, T. Genomic Responses in Mouse Models Greatly Mimic Human Inflammatory Diseases. Proc. Natl. Acad. Sci. USA 2015, 112, 1167–1172. [Google Scholar] [CrossRef] [PubMed]
- Cogels, M.M.; Rouas, R.; Ghanem, G.E.; Martinive, P.; Awada, A.; Van Gestel, D.; Krayem, M. Humanized Mice as a Valuable Pre-Clinical Model for Cancer Immunotherapy Research. Front. Oncol. 2021, 11, 784947. [Google Scholar] [CrossRef] [PubMed]
- Kruse, B.; Buzzai, A.C.; Shridhar, N.; Braun, A.D.; Gellert, S.; Knauth, K.; Pozniak, J.; Peters, J.; Dittmann, P.; Mengoni, M.; et al. CD4+ T Cell-Induced Inflammatory Cell Death Controls Immune-Evasive Tumours. Nature 2023, 618, 1033–1040. [Google Scholar] [CrossRef]
- Deycmar, S.; Gomes, B.; Charo, J.; Ceppi, M.; Cline, J.M. Spontaneous, Naturally Occurring Cancers in Non-Human Primates as a Translational Model for Cancer Immunotherapy. J. Immunother. Cancer 2023, 11, e005514. [Google Scholar] [CrossRef]
- Tarantal, A.F.; Noctor, S.C.; Hartigan-O’connor, D.J. Nonhuman Primates in Translational Research. Annu. Rev. Anim. Biosci. 2022, 10, 441–468. [Google Scholar] [CrossRef]
- How Vaccines Are Developed and Approved for Use|Vaccines & Immunizations|CDC. Available online: https://www.cdc.gov/vaccines/basics/how-developed-approved.html?CDC_AAref_Val=https://www.cdc.gov/vaccines/basics/test-approve.html (accessed on 28 October 2024).
- Vaccine Safety & Availability|FDA. Available online: https://www.fda.gov/vaccines-blood-biologics/safety-availability-biologics/vaccine-safety-availability (accessed on 28 October 2024).
- Reyes, C.; Patarroyo, M.A. Adjuvants Approved for Human Use: What Do We Know and What Do We Need to Know for Designing Good Adjuvants? Eur. J. Pharmacol. 2023, 945, 175632. [Google Scholar] [CrossRef]
- Zhao, T.; Cai, Y.; Jiang, Y.; He, X.; Wei, Y.; Yu, Y.; Tian, X. Vaccine Adjuvants: Mechanisms and Platforms. Signal Transduct. Target. Ther. 2023, 8, 283. [Google Scholar] [CrossRef]
- Ols, S.; Yang, L.; Thompson, E.A.; Pushparaj, P.; Tran, K.; Liang, F.; Lin, A.; Eriksson, B.; Karlsson Hedestam, G.B.; Wyatt, R.T.; et al. Route of Vaccine Administration Alters Antigen Trafficking but Not Innate or Adaptive Immunity. Cell Rep. 2020, 30, 3964–3971.e7. [Google Scholar] [CrossRef]
- Reis E Sousa, C. Dendritic Cells in a Mature Age. Nat. Rev. Immunol. 2006, 6, 476–483. [Google Scholar] [CrossRef]
- Banchereau, J.; Briere, F.; Caux, C.; Davoust, J.; Lebecque, S.; Liu, Y.J.; Pulendran, B.; Palucka, K. Immunobiology of Dendritic Cells. Annu. Rev. Immunol. 2000, 18, 767–811. [Google Scholar] [CrossRef] [PubMed]
- Raskov, H.; Orhan, A.; Christensen, J.P.; Gögenur, I. Cytotoxic CD8+ T Cells in Cancer and Cancer Immunotherapy. Br. J. Cancer 2020, 124, 359–367. [Google Scholar] [CrossRef] [PubMed]
- Fučíková, J.; Rožková, D.; Ulčová, H.; Budinský, V.; Sochorová, K.; Pokorná, K.; Bartůňková, J.; Špíšek, R. Poly I: C-Activated Dendritic Cells That Were Generated in CellGro for Use in Cancer Immunotherapy Trials. J. Transl. Med. 2011, 9, 223. [Google Scholar] [CrossRef]
- Wang, H.; Kaur, G.; Sankin, A.I.; Chen, F.; Guan, F.; Zang, X. Immune Checkpoint Blockade and CAR-T Cell Therapy in Hematologic Malignancies. J. Hematol. Oncol. 2019, 12, 59. [Google Scholar] [CrossRef]
- Conlon, K.C.; Miljkovic, M.D.; Waldmann, T.A. Cytokines in the Treatment of Cancer. J. Interferon Cytokine Res. 2019, 39, 6. [Google Scholar] [CrossRef]
- Maris, M.B.; Niederwieser, D.; Sandmaier, B.M.; Storer, B.; Stuart, M.; Maloney, D.; Petersdorf, E.; McSweeney, P.; Pulsipher, M.; Woolfrey, A.; et al. HLA-Matched Unrelated Donor Hematopoietic Cell Transplantation after Nonmyeloablative Conditioning for Patients with Hematologic Malignancies. Blood 2003, 102, 2021–2030. [Google Scholar] [CrossRef]
- Lica, J.J.; Pradhan, B.; Safi, K.; Jakóbkiewicz-Banecka, J.; Hellmann, A. Promising Therapeutic Strategies for Hematologic Malignancies: Innovations and Potential. Molecules 2024, 29, 4280. [Google Scholar] [CrossRef]
- Xie, Q.; Ding, J.; Chen, Y. Role of CD8+ T Lymphocyte Cells: Interplay with Stromal Cells in Tumor Microenvironment. Acta Pharm. Sin. B 2021, 11, 1365. [Google Scholar] [CrossRef]
- Tay, R.E.; Richardson, E.K.; Toh, H.C. Revisiting the Role of CD4+ T Cells in Cancer Immunotherapy—New Insights into Old Paradigms. Cancer Gene Ther. 2020, 28, 5–17. [Google Scholar] [CrossRef]
- Guy, B. The Perfect Mix: Recent Progress in Adjuvant Research. Nat. Rev. Microbiol. 2007, 5, 505–517. [Google Scholar] [CrossRef]
- De Waele, J.; Verhezen, T.; van der Heijden, S.; Berneman, Z.N.; Peeters, M.; Lardon, F.; Wouters, A.; Smits, E.L.J.M. A Systematic Review on Poly(I:C) and Poly-ICLC in Glioblastoma: Adjuvants Coordinating the Unlocking of Immunotherapy. J. Exp. Clin. Cancer Res. 2021, 40, 213. [Google Scholar] [CrossRef] [PubMed]
- Mathias, F.A.S.; Ostolin, T.L.V.D.P.; Reis, L.E.S.; Cardoso, J.M.d.O.; De Brito, R.C.F.; Aguiar Soares, R.D.d.O.; Roatt, B.M.; Vieira, P.M.d.A.; Reis, A.B. The Use of an Adjuvant System Improves Innate and Adaptive Immune Response When Associated with a Leishmania (Viannia) Braziliensis Antigen in a Vaccine Candidate against L. (Leishmania) Infantum Infection. Vaccines 2023, 11, 395. [Google Scholar] [CrossRef]
Ag | Vaccine Type | Pathology | Study Stage | Patients Profile | Disease Stage | Model | Challenge Via | Vaccine Scheme | Immunization Via | Associated Treatment | Follow Up | Assay | Efficacy | Limitations | Ref. |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
PASD1 antigen | DNA vaccine | AML | preclinical | N/A | N/A | HHD mice | N/A | 1 dose | IM | N/A | N/A | (1) In silico-Bioinformatics; (2) Ex vivo; (3) ELISA; (4) ELISPOT; (5) Flow cytometry | PASD1-derived peptides binding to MHC-I; Stimulation of IFN-γ production by CD8+ T cells; Effector CD8+ T cells for the lysis of PASD1-expressing cells. | Limitations associated with the preclinical nature of the study; Requirement for transgenic animal models to enhance translational relevance; Absence of in vivo challenge experiments, restricting the assessment of protective efficacy; Limited scope of immunological evaluations to characterize the vaccine-induced response. | [18] |
Peptides derived from leukemia associated Ags (LAAs) and minor histocompatibility Ags (miHAs) | DC vaccine | TCL | preclinical | N/A | N/A | B10, C57BL/10 and C57BL/6 mice | IV | 2 doses, 1 week interval | IV | N/A | 100 days | (1) Ex vivo; (2) In vitro; (3) ELISPOT; (4) Flow cytometry | Enhanced survival rate observed in experimental models; Increased frequency of antigen-specific T cells producing IFN-γ; Antigens with higher abundance did not consistently elicit the most robust immune response. | Limitations inherent to the preclinical nature of the study; Absence of in vivo challenge experiments, limiting the evaluation of real-world applicability. | [19] |
Peptides derived from WT1 and pan-DR binding peptide epitope (PADRE) | Peptide vaccine | AML | clinical-phase 1 and 2 | HLA-A*0201+ low risk patients | CR1, CR2, PR and slow progression | N/A | N/A | 5 doses, 3 weeks interval | SC | N/A | 41 months | (1) Safety and cytotoxicity; (2) Ex vivo; (3) Flow cytometry; (4) ELISPOT; (5) Molecular analyses | Safe formulations for administration in patients; Increase in specific CD8+ T cells up to 12 weeks; Increase in IFN-γ-producing CD8+ T cells in 6/7 patients; 2/5 eligible patients showed reduction in WT1 mRNA in PBMC. | Preliminary study with limited data available; Insufficient conclusive evidence regarding the treatment’s impact on disease progression in patients. | [20] |
Synthetic HSP (heat-shock protein) peptides | Peptide vaccine | MM | preclinical | MM patients | N/A | HLA-A2.1-tg and NOD/SCID mice | SC | 1 dose | SC | N/A | 2 weeks | (1) In silico-Bioinformatics; (2) Ex vivo; (3) Molecular assays; (4) In vitro; (5) Flow cytometry; (6) Overall survival | MM cells show increased expression of HSPs; Increased proliferation of IFN-γ-producing CD8+ T cells; Increased lysis of MM cells by CD8+ T effector memory cells; Reduced tumor growth and increased survival rate. | Limitations inherent to the preclinical nature of the study; Necessity of employing transgenic animal models for further validation. | [21] |
L1210 or K-562 Leukemic cell exosomes (LEXs) | DC vaccine | ALL | preclinical | N/A | N/A | DBA/2 mice | SC | 1 dose | SC | N/A | 70 days | (1) Imaging analysis; (2) Flow cytometry; (3) Ex vivo | LEXs are important antigenic sources; Their phagocytosis by DCs is efficient and stable even after 72 h; Increased cytotoxic capacity in animals; Prophylactic potential with increased resistance to disease establishment after vaccination; Increased survival rate. | Limitations due to the preclinical nature of the study; Limited evaluation of the immunological response to the vaccine. | [22] |
Synthetic peptides Tax11-19 (LLFGYPVYV) and Tax301-309 (SFHSLHLLY) | DC vaccine | ATL | clinical-phase 1 | HLA-A*0201, A*24:02 or A*11:01 patients | SD or PR | N/A | N/A | 3 doses, 2 weeks interval | SC | N/A | 14 months | (1) Ex vivo; (2) Safety and cytotoxicity; (3) Flow cytometry; (4) Molecular analysis | Safe vaccine formulation for administration in patients; Despite the fluctuation between patients, there was an increase in cytotoxic cells producing IFN-γ; Improvement in patient prognosis. | Small sample size; Limited conclusive data on the impact of treatment on disease progression in patients. | [23] |
DNA encoding the patient-specific idiotype with the VH and VL region genes | DNA vaccine | MM | clinical-phase 1 | MM patients who have already undergone chemotherapy and immunotherapy protocols | CR, PR, or SD | N/A | N/A | 6 doses, 1 to 4 weeks interval | IM | N/A | 52 weeks | (1) Molecular analyses; (2) ELISA; (3) ELISPOT; (4) Overall survival | Safe vaccine formulation for administration in patients; Increased production of specific IgG; 11/14 patients remained in remission after 52 weeks; 7/14 patients after 52 weeks remained without detection of disease biomarkers. | Limited immunological assessments of vaccine response. | [24] |
Nonspecific immunogenic DNA sequences inserted into a vector | DNA vaccine | APL and MDS | preclinical | N/A | N/A | FVB/N mice | IV | 3 doses, 20 days interval | IM | Vitamin-All-trans retinoic acic (ATRA) | 200 days | (1) Molecular analyses; (2) Overall survival; (3) Histological analysis; (4) ELISA; (5) Flow cytometry | Increased survival rate; Reduction in the number of immature blasts in the bone marrow; The production of specific IgG for both vaccines was variable in the animals; Both the isolated vaccines and those associated with treatment showed an increase in memory T cells, cytotoxic T cells, and IFN-γ production. | Limitations arising from the preclinical nature of the study; Limited comprehensive immunological assessments of the vaccine response. | [25] |
Eu-myc cell whole antigen | Cell lysate vaccine | BCL | preclinical | N/A | N/A | C57BL/6 and B6. SJLxOT-I e IFN-gKO mice | IV | 1 dose | IV | mAB-anti-4-1BB | 180 days | (1) Elisa; (2) Ex vivo; (3) Flow cytometry | The vaccine + antibody combination promoted the greatest increase in survival rate; The vaccine promoted long-lasting protection after a new challenge; An increase in cytotoxic T cells producing IFN-γ was observed; The importance of these cells in disease control was demonstrated by depletion and transfer assays. | Limitations due to the preclinical nature of the study; Requirement for the use of transgenic animals. | [26] |
HBZ protein | Recombinant virus vaccine | ATL | preclinical | N/A | N/A | C57BL/6, NOD/SCID mice and Rhesus monkeys | IP | 6 doses, 3–4 weeks interval | ID | N/A | 45 days | (1) Flow cytometry; (2) Ex vivo; (3) ELISA | Vaccination induced increased production of pro-inflammatory cytokines by T cells; Immunization was protective with increased cytotoxic activity; The vaccine induced the formation of memory T cells. | Limitations due to this being a preclinical study; Need to utilize transgenic animals; Difficulty in employing non-human primates. | [27] |
Bap (BCR-ABL) peptide | Peptide vaccine and Recombinant virus vaccine | ALL | preclinical | N/A | N/A | C57BL/6, Cdkn2a-/-, Foxp3-GFP, Ifng-/- and OT-I x Rag2-/-. mice | IV | 3 doses, 7 days interval | IV or IP | mAB-anti-PDL1, anti-CTLA | 80 days | (1) Flow cytometry; (2) Overall survival; (3) Ex vivo; (4) ELISPOT | Activation of CD4+ T cells by MHC-II molecules influences disease progression; The use of blocking antibodies promoted a modest increase in survival; Vaccination induced the formation of multifunctional cytokine-producing T cells and memory T cells; The cytokine IFN-γ is important in the anti-leukemic response. | Limitations due to this being a preclinical study; Need for the use transgenic animals. | [28] |
mRNA RHAMM (hyaluronic acid)or mRNA WT1 | DC vaccine | AML | clinical-phase 1 | AML Patients in remission who have already undergone chemotherapy protocol | CR1 | N/A | N/A | 4 doses, 2 weeks interval | ID | N/A | 8 weeks | (1) Ex vivo; (2) Molecular analysis; (3) Flow cytometry; (4) ELISA | Activation of CD4+ T cells by MHC-II molecules influences disease progression; The use of blocking antibodies promoted a modest increase in survival; Vaccination induced the formation of multifunctional cytokine-producing T cells and memory T cells; The cytokine IFN-γ is important in the anti-leukemic response. | Limited immunological assessments of vaccine response. | [29] |
Peptides derived from WT1 | DC vaccine | ALL, AML and HL | clinical-phase 1 and 2 | HLA-A2 patients in relapse after allo-HSCT expressing WT1 | Relapse | N/A | N/A | 6 doses, 2 weeks interval | SC or ID | Cell inoculation—Donnor lymphocytes | 36 weeks | (1) ELISPOT; (2) DTH | Vaccine formulation safe for administration in patients; Despite segment loss, 3 patients presented DTH; Vaccination induced IFN-γ production. | Limited immunological assessments of vaccine response; Early study, limited conclusive data on the effect of treatment on disease progression in patients. | [30] |
mRNA hTERT (human telomerase reverse transcriptase) | DC vaccine | AML | clinical-phase 2 | Patients with AML in first or second remission | CR1 or CR2 | N/A | N/A | Cycle 1: 6 doses, 1 week interval; cycle 2: 6 doses, 2 weeks interval | ID | N/A | 2 years | (1) Histological analysis; (2) ELISPOT; (3) DTH; (4) Molecular analysis | 74% of vaccinated patients remained in remission during the study follow-up period; In 69% of patients in remission, specific T cells developed; Patients who were in second remission remained in this state for 24 months; In 5/7 patients over 60 years of age, T cell responses were observed, and there was no recurrence of the disease during the follow-up period. | Loss of patient follow-up throughout the study. | [31] |
Leukemic tumor cell exosomes (LEX) transfected with a lentiviral vector containing a shRNA (small harpin RNA) sequence | Exosome vaccine | ALL | preclinical | N/A | N/A | DBA/2 mice | SC | 3 doses, 5–7 days interval | SC | N/A | 50 days | (1) Imaging analysis; (2) Molecular analysis; (3) Flow cytomety; (4) Ex vivo; (5) Overall survival | In vitro, LEX TGF-B1si captured by DCs induces pro-inflammatory cytokines and CD4+ T cell proliferation; Immunization induces increased cytotoxic activity of cytotoxic T lymphocytes and NK cells; In vivo, the presence of LEX TGF-β1si induced increased protection against tumor establishment; Increased survival rate. | Limitations due to this being a preclinical study. | [32] |
Synthetic peptide OCV-501 derived from WT1 | Peptide vaccine | AML | preclinical and clinical-phase 1 | AML patients HLA-DRB1*01:01, *04:05, *15:01, *15:02, *08:03, or *09:01 in remission who have already undergone chemotherapy protocol | CR | N/A | N/A | 4 doses, 4 weeks interval | SC | N/A | 50 days | (1) In vitro; (2) Ex vivo; (3) Flow cytometry; (4) ELISA; (5) Analytical chemistry assay; (6) Safety and cytotoxicity; (7) Molecular assays | In vitro, the vaccine peptide induced the differentiation and proliferation of Th1 cells from PBMC; An increase in specific cytotoxic T cells was also observed; In clinical studies, the vaccine formulation was safe for administration to patients; During the observation period, patients did not present relapse; Vaccination induced a DTH response in patients; 4/9 patients presented reduced WT1 mRNA expression (MRD). | This is an initial study with limited conclusive data on the impact of treatment on disease progression in patients. | [33] |
Inactivated WEHI-3 total leukemic cell antigen | Cell lysate vaccine | AML | preclinical | N/A | N/A | Balb/c mice | IV | 4 doses, 3 days interval | SC | Cytokine-G-CSF | 50 days | (1) Ex vivo; (2) In vitro; (3) Imaging analysis; (4) ELISA; (5) Flow cytometry; (6) Overall survival | G-CSF associated with the vaccine induced the reversal of the immunosuppressive tumor microenvironment by reducing Treg cells in the bone marrow and increasing these cells in the peripheral blood; The treatment increased the survival rate of vaccinated animals. | Limitations due to it being a preclinical study. | [34] |
MMSA-1 (multiple myeloma special antigen) and DKKP-1 (Dickkopft-(1)) | Peptide vaccine | MM | preclinical and clinical-phase 1 | MM patients | N/A | SCID mice | SC | 3 doses, 1 week interval | SC | N/A | 60 days | (1) Molecular analysis; (2) ELISA; (3) Ex vivo; (4) Flow cytometry; (5) Overall survival; (6) Hematological assays; (7) Imaging analysis | MMSA-1 expression is essential in the establishment of the disease; In vitro MMSA-1 peptides induced the formation of specific cytotoxic T cells from PBMC of patients; In vivo it was found that the vaccine induced an increase in cytotoxic T cells producing IFN-γ and IL-2 and a reduction in Treg cells; Vaccination led to a reduction in tumor volume and bone degradation with an increase in the survival rate. | Limitations due to this being a preclinical study; Need to use transgenic animals. | [35] |
Tumor cell lysate (TCL) | DLBCL and MCL | preclinical | N/A | N/A | NOD/SCID mice | SC | 3 doses, 1 week interval | IP | Cytokine-IFN-α; Vitamin-retinoic acid | 43 days | (1) Flow cytometry; (2) ELISA; (3) Ex vivo; (4) Imaging analysis | In vitro, DCs incubated with TCL-RA-IFN-α induced tumor cell death by inducing CRT expression in them and increasing phagocytosis of these cells by DCs; Increased immunogenicity of cytotoxic T cells; In vivo, the vaccine led to inhibition of tumor growth. | Limitations due to this being a preclinical study; Need to use transgenic animals. | [36] | |
WT1 mRNA | DC vaccine | AML | clinical-phase 2 | AML patients at high risk of relapse | CR | N/A | N/A | 16 doses, 2 weeks interval | ID | N/A | 10 years | (1) Molecular analysis; (2) ELISA; (3) Flow cytometry; (4) DTH assay; (5) Histological analysis | Vaccination induced molecular remission in 9 patients by reducing WT1 transcripts in blood and bone marrow; 2/9 patients went from partial to complete remission; Vaccination led to an increase in the frequency of IFN-γ and TNF-α-producing cytotoxic T lymphocytes and increased survival rate. | Limited immunological assessments of vaccine response. | [37] |
A20 tumor cell lysate | Cell lysate vaccine | DLBCL | preclinical | N/A | N/A | Balb/c mice | IP | Schedule 1: 3 doses, days 1, 7, and 15; Schedule 2: 3 doses, days 7, 10, and 14 | SC | Glycolipid-α-GalCer | 160 days | (1) Flow cytometry; (2) ELISA; (3) Ex vivo; (4) Imaging analysis; (5) Multiplex assay | Vaccination induced protection with increased animal survival; There was an increase in NK cells and cytotoxic T cells producing IFN-γ; The association with α-GalCer intensified the cellular response and reduced the number of Tregs. | Limitations arise from the study being preclinical in nature, restricting direct applicability to clinical settings. | [38] |
Whole antigen of K-562 cells expressing GM-CSF and autologous tumor cells | Cell lysate vaccine | MDS and AML | clinical-phase 1 | Patients with advanced MDS or high-risk AML | Advanced disease, high risk | N/A | N/A | Cycle 1: 3 doses, 1 week interval; Cycle 2: 3 doses, 2 week interval | SC/ID | Cell inoculation-HSCT | 6 years | (1) Hematology assay; (2) DTH assay; (3) Flow cytometry; (4) ELISA | Safe vaccine formulation for administration in patients, 39% of patients presented relapse-free survival during 5 years of follow-up; Vaccination induced an increase in CD4+ and CD8+ cells in the initial 2 months of treatment; Biopsies of the vaccination site confirmed the influx of DCs, macrophages, neutrophils and lymphocytes. | Loss of patient follow-up throughout the study; Limited immunological assessments of vaccine response. | [39] |
Synthetic peptide WT4869 derived from WT1 | Peptide vaccine | MDS | clinical-phase 1 and 2 | MDS patients unresponsive to conventional treatments | N/A | N/A | N/A | Escalonade dose 3 + 3, 2 weeks interval | ID | N/A | 3 years | (1) Safety and cytotoxicity; (2) DTH assay; (3) Flow cytometry; (4) Molecular assay; (5) Ex vivo | The vaccine formulation presented grade 3 or higher adverse effects in 19 of the 26 patients; Vaccination promoted an improvement in the hematological condition of the patients; The patients presented a DTH response and 11/25 patients presented an increase in cytotoxic T cells; There was an increase in the mean survival rate of 64.71%; At the end of the study, 13/26 patients died. | Limited immunological assessments of vaccine response; Adverse effects of vaccination. | [40] |
DKKP-1 (Dickkopft-(1)) antigen and HSP70 | DNA vaccine | MM | preclinical | N/A | N/A | Balb/c | SC | 3 doses, interval not reported | IM | N/A | 100 days | (1) Safety and cytotoxicity; (4) Histological analysis; (3) Flow cytometry; (4) Molecular analysis; (5) Ex vivo | The vaccine formulation induced tumor regression in both prophylactic and therapeutic models; vaccination promoted an increase in IFN-γ-producing CD4+ and CD8+ cells and a decrease in T-reg cells in the spleen; it enhanced CTL activity and antigen-specific antibody responses; the number of apoptotic tumor cells was increased in the vaccinated group. | Limitations due to the preclinical nature of the study. | [41] |
Synthetic peptides derived from WT1 | Peptide vaccine | AML | preclinical | N/A | N/A | C57BL/6J CD45.1 and CD45.2 mice | SC | 1 dose | SC | N/A | N/A | (1) Histological analysis; (2) Flow cytometry; (3) Molecular analysis; (4) In silico-Bioinformatics | Vaccination with 2 peptides promoted an increase in CD4+ T cells and mainly CD8+ T cells and DCs with induction of necrotic lesions in the tumor; Vaccination also induced an increase in the production of IFN-γ and TNF-α by specific CD4+ and CD8+ T cells in addition to effector memory and resident memory T cells. | Limitations due to this being a preclinical study; Need to use transgenic animals. | [42] |
Synthetic peptides derived from WT1 | Peptide vaccine | AML | clinical-phase 2 | AML patients in remission who have already undergone chemotherapy protocol | CR1 | N/A | N/A | 6 doses, 2 weeks interval | SC | Cytokine-G-CSF | 80 months | (1) Hematological analysis; (2) Molecular analysis; (3) Flow cytometry; (4) ELISPOT | Safe vaccine formulation for administration in patients; Vaccination promoted an increase in the overall survival rate (67.6 months); The mean disease-free survival was 16.9 months; 11/22 patients remained alive at the end of the study, and 9 of these remained in remission; Vaccination induced an increase in specific IFN-γ-producing CD8+ T cells and the proliferation of CD4+ T cells. | Loss of patient follow-up throughout the study. | [43] |
Whole antigens associated with AML | DC vaccine | AML | clinical-phase 1 | AML patients in remission who have already undergone chemotherapy protocol | CR2, de novo CR1, smolerding AML | N/A | N/A | 4 doses, 2 weeks interval | ID | N/A | 2 years | (1) Ex vivo; (2) Flow cytometry; (3) In vitro; (4) Safety and cytotoxicity | Vaccine formulation safe for administration in patients; After 126 days of follow-up, 9/12 patients remained alive and 6/12 patients remained in remission; Vaccination induced an increase in CD4+, CD8+, NK T cells, and DCs at the immunization site; Cytokine evaluation showed the induction of a Th1 or mixed response with production of IFN-γ, IL-2, IL-4, IL-6, IL-10, and IL-17. | Limited immunological assessments of vaccine response; Early study with limited data, resulting in insufficient conclusive evidence on the impact of treatment on disease progression in patients. | [44] |
Whole antigens associated with AML | Cell lysate vaccine | AML | preclinical | N/A | N/A | C57BL/6 mice | IV | 1 dose | IV | mAB-anti-PDL1, anti-4-1BB | 120 days | (1) Imaging analysis; (2) Flow cytometry; (3) Overall survival | Vaccination associated with anti-4-1BB mAB promoted the cure of 100% of the animals; There was an increase in central and effector memory NK and CD8+ T cells; The adoptive transfer of effector memory T cells to sick animals confirmed the importance of this population in controlling the disease. | Limitations due to it being a preclinical study. | [45] |
Whole antigens of parental leukemic cells | Cell lysate vaccine | AML | preclinical | N/A | N/A | C3H mice | IV | 3 doses, 2 weeks interval | SC | N/A | 450 days | (1) Flow cytometry; (2) ELISA; (3) Ex vivo; (4) In vitro; (5) ELISPOT; (6) Overall survival; Molecular analysis | Vaccination induced proliferation of CD4+ and CD8+ T cells, increased cytolytic activity and IFN-γ production; There was an increase in the survival rate of the animals. | Limitations due to it being a preclinical study. | [46] |
Synthetic peptides derived from WT1 | Peptide vaccine | AML | clinical-phase 1 | HLA-A*02 patients with MDS or AML | CR ou CRi | N/A | N/A | 6 doses, 2 weeks interval | SC | N/A | 14 months | (1) Molecular analyis; (2) ELISPOT; (3) Ex vivo | Safe vaccine formulation for administration in patients; Vaccination induces an increase in CD8+ T cells with IFN-γ production for both AML and MDS patients. | Limited immunological assessments of vaccine response; As this is an early-stage study, there is limited conclusive data on the impact of treatment on disease progression in patients. | [47] |
Tumor cell exosomes | DC vaccine | DLBCL | preclinical | N/A | N/A | C57BL/6, Balb/c and NOD/SCID mice | SC | 3 doses, 2 weeks interval | IV | N/A | 56 days | (1) Imaging analysis; (2) Flow cytometry; (3) Ex vivo; (4) Molecular analysis | Vaccination stimulated the proliferation of specific cytotoxic T cells with production of TNF-α and IL-6 and anti-tumor response. | Limitations due to being a preclinical study; Necessity for the use of transgenic animals. | [48] |
Whole antigen of K-562 cell | Cell lysate vaccine | MDS | clinical phase 1 | Patients with any subtype of MDS who have not been treated for at least 2 months and have not undergone HSCT | N/A | N/A | N/A | 5 doses, weeks 0, 3, 6, 9 and 17 | ID | N/A | 22 weeks | (1) Ex vivo; (2) Multiplex assay; Molecular assay | Vaccine formulation safe for administration in patients; Vaccination was not able to induce cell proliferation in in vitro culture samples from all patients; Patients who responded were observed to have an increase in IL-6 in cell cultures; TCR sequencing analysis of a single patient showed that the vaccine induced a polyclonal expansion of CD4+ and CD8+ T cells. | Limited immunological assessments of vaccine response; In the early stages of study, there are limited conclusive data on the impact of treatment on disease progression in patients. | [49] |
scFv-CCL20 plasmid DNA | DNA vaccine | LPL | clinical phase 1 | Asymptomatic patients not treated with LPL | N/A | N/A | N/A | 3 doses, 1 month interval | ID | N/A | 1 year | (1) Ex vivo; (2) ELISPOT (3) Hematological analysis; (4) Histological analysis; (5) Safety and cytotoxicity | Safe vaccine formulation for administration to patients. | Limited immunological assessments of vaccine response; In the early stages of study, there are limited conclusive data on the impact of treatment on disease progression in patients. | [50] |
Survivin-MUC1 protein | DC vaccine | AML and ALL | preclinical, clinical-phase 1 and 2 | Patients with relapse after allo-HSCT | Relapse | Rhesus monkeys | N/A | Patients: 4 doses, 2 days interval; animals 1 dose | SC-humans/IM-monkeys | N/A | 10 years (humans) 14 days (animal) | (1) Flow cytometry; (2) ELISA; (3) Ex vivo; (4) ELISPOT; (5) Hematological analysis; (6) Molecular analysis; (7) Histological analysis | In vitro, it was observed that Ad-siSSF was able to activate DCs and that Ad-siSSF-DCs activated cytotoxic T cells; In vivo, vaccination promoted an increase in IFN-γ, increasing the survival rate of animals; Vaccination also induced a reduction in WT1 expression and a reduction in MRD. | Difficulty in using non-human primates. | [51] |
L5178Y-R tumor cell lysate (TCL) treated with PKHB1 (PKHB1-TCL) | Cell lysate vaccine | ALL | preclinical | N/A | N/A | Balb/c mice | SC | 4 doses, 3 days interval | SC | N/A | 150 days | (1) Ex vivo; (2) In vitro; (3) Flow cytometry; (4) Overall survival | In vitro, the vaccine peptide induced tumor cell lysis, dendritic cell activation, and production of IFN-γ, TNF-α, and IL-2 and T cell activation; In vivo, vaccination induced tumor reduction (80%), memory cell production and increased survival rate. When challenged again, the immunized animals protected 100% of the animals. | Limitations due to it being a preclinical study. | [52] |
PLK1 derived peptides | DC vaccine | AML | preclinical | N/A | N/A | C57BL/6 mice | SC | 2 doses, 1 week interval | IV | mAB-anti-PDL1 | 100 days | (1) Ex vivo; (2) Flow cytometry; (3) ELISPOT; (4) In vitro; (5) Overall survival; (6) Molecular analysis | 2/8 vaccine peptides showed stimulation and expansion of cytotoxic T cells producing IFN-γ. Vaccination with these isolated peptides induced tumor reduction; The immunized animals proved resistant to the establishment of the disease after a new challenge; Vaccination was also able to protect the animals against more than one type of tumor. There was an increase in the survival rate. The association of the vaccine with anti-PD-L1 treatment further increased cytotoxic T cells with greater activity. The association of the peptides promoted better vaccine performance than the isolated vaccines. | Limitations due to it being a preclinical study. | [53] |
mRNA encoding WT1, PRABE and (CMV)pp65 antigens | DC vaccine | AML | clinical-phase 1 | AML patients in remission who have already undergone chemotherapy protocol | CR | N/A | N/A | 10 doses total: 4 doses, 1 week interval + 1 dose, 2 weeks interval + 5 doses, 4 weeks interval | ID | N/A | 3 years | (1) Ex vivo; (2) Flow cytometry; (3) DTH assay; (4) Histological analysis; (5) ELISPOT; (6) ELISA; 7 Safety and cytotoxicity | Safe vaccine formulation for administration in patients; Vaccination promoted a 50% recurrence rate; Patients under 65 years of age had a better immune response with the presence of specific T cells. | Small sample size, there is not much conclusive data on the effect of treatment on disease progression in patients. | [54] |
Exosomes from tumor cells expressing CD80/86 | Exossome vaccine | ALL | preclinical | N/A | N/A | DBA/2 mice | SC | 3 doses, 1 week interval | SC | N/A | 81 days | (1) Flow cytometry; (2) Imaging analysis; (3) Molecular analysis; (4) ELISA; (5) Ex vivo; (6) Overall survival | Vaccination reduced tumor size and increased survival rate; immunized animals showed greater proliferation of CD4+ T cells, greater activity of cytotoxic T cells, production of pro-inflammatory cytokines and maturation of DCs. | Limitations due to it being a preclinical study. | [55] |
Whole antigens expressed on U937 cells | Cell lysate vaccine | AML | preclinical | N/A | N/A | Balb/c mice | SC | 4 doses, 1 week interval | SC | N/A | 30 days | (1) Ex vivo; (2) In vitro; (3) Flow cytometry; (4) ELISA; (5) Molecular analysis; (6) Overall survival | Vaccination promoted in vitro the activation of DCs and CTLs with production of pro-inflammatory cytokines; It was observed that the vaccine was able to reduce tumor size and increase the survival rate of treated animals. | Limitations due to it being a preclinical study. | [56] |
Synthetic peptide RT53 | Peptide-pulsed splenocyte vaccine | APL | preclinical | N/A | N/A | FVB/N mice | IV | 1 dose | SC | N/A | 260 days | (1) Flow cytometry; (2) Ex vivo; (3) ELISA; (4) Overall survival | The RT53 peptide promoted the death of leukemic cells in vitro; Vaccination induced an increase in the survival rate; Both the therapeutic and prophylactic approaches were able to protect the animals from the development of the disease with an important role of CD4+ T cells. | Limitations due to this being a preclinical study; Need to use transgenic animals; Limited immunological evaluations of the vaccine response. | [57] |
Long peptide (19aa) IO103 | Peptide vaccine | MM | clinical-phase 1 | MM patients who have already undergone chemotherapy protocol in the last 6 months and allo-HSCT | N/A | N/A | N/A | 15 total doses: 6 doses, 2 weeks interval + 9 doses, 4 weeks interval | SC | N/A | 3 years | (1) Ex vivo; (2) DTH assay; (3) ELISPOT; (4) Flow cytometry; (5) Multiplex assay | Safe vaccine formulation for administration in patients; Patients showed DTH response; Lymphocytes from the biopsy site were able to produce IFN-γ and TNF-α; 8/10 patients remained alive at the end of the study. | Limited immunological assessments of vaccine response; Small sample size, there is not much conclusive data on the effect of treatment on disease progression in patients. | [58] |
Antigens from the cell membrane of leukemic cells | Cell-membrane-coated nanoparticle vaccine | AML | preclinical | N/A | N/A | C57BL/6 mice | IV | 3 doses, 2 ou 7 dias de intervalo | SC | N/A | 12 weeks | (1) In vitro; (2) Overall survival; (3) Imaging analysis; (4) Flow cytometry; (5) Ex vivo; (6) ELISPOT; (7) Overall survival | Vaccination induced the activation of specific IFN-γ-producing T cells; the formation of central and effector memory cells and an increase in the survival rate were also observed. | Limitations due to it being a preclinical study. | [59] |
WT1 and HAGE derived peptides | DNA vaccine | CML | preclinical | N/A | N/A | HHDII-DR1 transgenic mice | SC | 3 doses, 1 week interval | ID | N/A | 60 days | (1) In vitro; (2) Ex vivo; (3) ELISPOT; (4) Imaging analysis; (5) Flow cytometry | Vaccination induced the activation of specific IFN-γ-producing T cells; the formation of central and effector memory cells and an increase in the survival rate were also observed. | Limitations due to this being a preclinical study; Absence of a specific model for studying the disease in vivo. | [60] |
Whole antigens expressed in leukemic cells | DC vaccine | AML | preclinical | N/A | N/A | C57BL/6 mice | IV | 1 dose | SC | mAB-anti-PDL1, anti-TIM3, anti-RGMb | 180 days | (1) Ex vivo; (2) Flow cytometry; (3) Molecular analysis | Vaccination associated with anti-PD-1, anti-TIM3, and anti-RGMb mABs showed 100% survival for 90 days; In a second challenge, these animals maintained 100% survival; Vaccination induced an increase in IFN-γ-producing cytotoxic T lymphocytes, an increase in memory T cells and a reduction in Treg cells. | Limitations due to it being a preclinical study. | [61] |
Synthetic peptides derived from BCRs | DC vaccine | CLL | preclinical and clinical-phase 1 | Patients with severe and progressive CLL | Progressive | C57BL/6N mice | IP | 1 dose | ID | N/A | 200 days | (1) Ex vivo; (2) ELISPOT; (3) Flow cytometry; (4) ELISA | Vaccination promoted an increase in IFN-γ-producing T cells and increased cytotoxicity when cultured with Eu-TCL1 cells; T cells from vaccinated mice were able to produce IFN-γ when cultured with other tumor clones; There was an increase in the survival rate. | Limitations due to it being a preclinical study; Only a prophylactic study, absence of an in vivo therapeutic study. | [62] |
Whole antigens expressed in K562/GM-CSF cells | Cell lysate vaccine | CML | clinical-phase 2 | CML patients on chemotherapy protocol | Chronic phase | N/A | N/A | Up to 17 doses, 3 weeks interval | ID | Cytokines-IFN-α, GM-CSF | 60 months | (1) Molecular assay; (2) Safety and cytotoxicity; (3) Flow cytometry | Vaccination promoted an increase in IFN-γ-producing T cells and increased cytotoxicity when cultured with Eu-TCL1 cells; T cells from vaccinated mice were able to produce IFN-γ when cultured with other tumor clones; There was an increase in the survival rate. | Loss of patient follow-up throughout the study; The vaccine was not very effective. | [63] |
mRNA encoding the CDR3 region of the TCR hypervariable chain | mRNA vaccine | TCL and CTCL | preclinical | N/A | N/A | C57BL/6 mice | SC | 2 doses, 1 week interval | IV | N/A | N/A | (1) Flow cytometry; (2) ELISA; (3) Overall survival | Vaccination was able to induce a specific immune response against malignant T cells and promoted tumor reduction. | Limitations due to it being a preclinical study; Conducted as a prophylactic study with no in vivo therapeutic assessment. | [64] |
WT1 antigen | Cell vector vaccine | AML | clinical-phase 1 | AML patients in remission who have already undergone chemotherapy or are refractory | Relapse or refractory | N/A | N/A | 2 doses, 1 week interval | IV | N/A | 12 months | (1) Molecular assay; (2) Safety and cytotoxicity; (3) Flow cytometry; (4) Hematological analysis; (5) ELISPOT; (6) In silico-Bioinformatics | Vaccination was able to induce a specific immune response against malignant T cells and promoted tumor reduction. | Loss of patient follow-up throughout the study; Adverse effects of vaccination. | [65] |
Synthetic peptides derived from PD-L1 and PD-L2 | Peptide vaccine | CLL | clinical-phase 1 and 2 | Untreated CLL patients with IgHV gene | Progressive | N/A | N/A | 9 total doses: 6 doses, 2 weeks interval + 3 doses, 1 month interval | N/A | N/A | 52 weeks | (1) Safety and cytotoxicity; (2) ELISPOT; (3) Flow cytometry | Safe vaccine formulation for administration in patients; 17/19 patients remained with stable disease during follow-up; Vaccination induced an increase in IFN-γ-producing T cells and effector memory T cells. | Loss of patient follow-up throughout the study. | [66] |
VEE virus RNA replicating particles encoding FLT3 | Recombinant virus vaccine | AML and BCL | preclinical | N/A | N/A | Balb/c and B6. SJL mice | IV and SC | Scheme 1: 2 doses, 2 weeks interval; Scheme 2: 3 doses, 2 weeks interval | SC | N/A | 42 days | (1) Flow cytometry; (2) ELISA; (3) Molecular analysis | Vaccination induced an increase in specific IgG-producing B cells, cytotoxic T cells; the increase in specific responsive antibodies bound to tumor cells attenuated tumor growth. | Limitations due to this being a preclinical study; Limited immunological assessments of the vaccine response. | [67] |
Exosomes from dendritic cells stimulated with leukemic cell lysate | Exosome vaccine | CML | preclinical | N/A | N/A | Balb/c mice | IV | 14 doses, 2 days interval | ID | N/A | 180 days | (1) In vitro; (2) Imaging analysis; (3) Flow cytometry; (4) Ex vivo; (5) ELISPOT; (6) ELISA; (7) Hematological analysis; (8) Overall survival | In vitro, it was observed that DEX presented high affinity with T and NK cells, with increased activation, proliferation, and cytotoxic potential with production of IFN-γ and TNF; In vivo, vaccination protected 100% against the development of the disease. | Limitations due to it being a preclinical study; Absence of a specific model for studying the disease in vivo. | [68] |
WT1-A10 protein derived from WT1 | Protein vaccine | AML | clinical-phase 1 | AML patients expressing WT1 transcripts in blasts | CR, PR and CRi | N/A | N/A | Cycle 1: 6 doses, 2 weeks interval; Cycle 2: 6 doses, 3 weeks interval; Cycle 3: 4 doses, 6 weeks interval; Cycle 4: 4 doses, 3 months interval; Cycle 5: 4 doses, 6 months interval | IM | N/A | 75 months | (1) Molecular analysis; (2) In silico Bioinformatics; (3) ELISA; (4) Flow cytometry | Safe formulation for administration in patients; Vaccination induced an increase in the humoral response with production of specific antibodies and cellular response with an increase in CD4+ and CD8+ T cells; WT1 expression was reduced in vaccinated patients and consequent absence of MRD. | Small sample size, there are not many conclusive data on the effect of treatment on disease progression in patients. | [69] |
WT1 protein expressed by B. longum | Recombinant bacterial vaccine | AML | preclinical | N/A | N/A | C57BL/6 mice | SC | 10 total doses: 1 dose/day for 5 days/week + 1 dose/day for 5 days/week | Oral | N/A | 24 days | (1) Ex vivo; (2) Flow cytometry; (3) Overall survival | Vaccination associated with B. longum 420/2656 showed tumor reduction; There was an increase in cytotoxic T lymphocytes in the peripheral blood and an increase in effector memory T cells; Vaccination induced an increase in CD4+ T cells in the tumor infiltrate. | Limitations due to it being a preclinical study. | [70] |
WT1 protein expressed by B. longum | Recombinant bacterial vaccine | AML | preclinical | N/A | NA | C57BL/6 mice | SC | 10 total doses: 1 dose/day for 5 days/week + 1 dose/day for 5 days/week | Oral | N/A | 30 days | (1) Flow cytometry; (2) Histological analaysis; (3) Ex vivo; (4) ELISA | Vaccination promoted tumor reduction due to the presence of cytotoxic T cells; An increase in cytotoxic T cells was observed in the peripheral blood in addition to the increase in specific IgG antibodies; Vaccination showed an increase in DCs and CD4+ T cells producing IFN-γ. | Limitations due to it being a preclinical study. | [71] |
WT1 protein and OVA | mRNA vaccine | AML | preclinical | N/A | N/A | C57BL/6 mice | IV | 1 dose | IV | Cytokines-IL-2 | 120 days | (1) Flow cytometry; (2) Molecular analysis; (3) ELISPOT; (4) Overall survival | Vaccination induced activation of NK, iNK, CD4+ T, and cytotoxic T cells with increased production of IFN-γ, in addition to effector memory cells; There was an increase in the survival rate of the animals. | Limitations due to it being a preclinical study. | [72] |
Cell lysate of W10-iPSCs | DC vaccine and T cell vaccine | ALL | preclinical | N/A | N/A | NOD/SCID mice | IV | 3 doses, 1 week interval | IV | N/A | 45 days | (1) Ex vivo; (2) Overall survival; (3) Flow cytometry; (4) Multiplex analysis; (5) Biochemical analyses | Vaccination did not present acute toxicity in animals; There was a reduction in circulating tumor cells and an increase in T cells in peripheral blood; Vaccination induced an increase in the cytokines IL-6, IFN-γ, and TNF and a reduction in cellular exhaustion factors. | Limitations due to this being a preclinical study; Limited immunological assessments of the vaccine response. | [73] |
Vaccine Classification | Nº of Articles | References | Advantages | Disadvantages |
---|---|---|---|---|
1st generation | 14 | [26,34,36,38,39,44,45,46,49,52,56,59,61,63] | Simplified formulation | Risk of infection |
Strong immune response | Not suitable for immunocompromised | |||
Multivalent | ||||
2nd generation | 16 | [20,21,23,30,33,35,40,42,43,47,53,57,58,62,66,69] | Easy antigen modification | Less immunogenic |
Non-infectious | Necessity of adjuvants | |||
Stable | Time-consuming production | |||
3rd generation | 5 | [18,24,25,41,50,60] | Easy antigen modification | High cost of production |
Non-infectious | Potential risk of genetic integration | |||
Well-tolerated | Necessity of improved logistics of storage and transportation | |||
Strong immune response | ||||
Constitutional antigen expression | ||||
4th generation | 4 | [65,73] | Non-infectious | Fast degradation by RNAses |
Readily designed | Necessity of delivery vehicles | |||
Mass production capability | Drugs can impact mRNA metabolism | |||
Strong T and B cell responses | ||||
DC vaccines | 16 | [19,22,23,29,30,31,36,37,44,48,51,53,54,61,62,73] | Relatively simple to obtain DCs from monocytes (MoDCs) | Require MHC match for antigen presentation |
Ready-to-use when acquired from allogeneic transplant | Migratory capability is affected by long-term cultures | |||
Increase the chances of cross-presentation of targeted antigens | Depending on source (e.g., from CD34+ hematopoietic precursors), they provide a heterogeneous population | |||
Can be obtained from different sources and provide a gamut of possibilities for antigen presentation/immune system activation | In vitro cultured MoDCs shown some differences from natural DCs |
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. |
© 2025 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
Mathias, F.A.S.; Carvalho, M.G.R.; Ruiz, J.C. Therapeutic Vaccines for Hematological Cancers: A Scoping Review of This Immunotherapeutic Approach as Alternative to the Treatment of These Malignancies. Vaccines 2025, 13, 114. https://doi.org/10.3390/vaccines13020114
Mathias FAS, Carvalho MGR, Ruiz JC. Therapeutic Vaccines for Hematological Cancers: A Scoping Review of This Immunotherapeutic Approach as Alternative to the Treatment of These Malignancies. Vaccines. 2025; 13(2):114. https://doi.org/10.3390/vaccines13020114
Chicago/Turabian StyleMathias, Fernando Augusto Siqueira, Maria Gabriela Reis Carvalho, and Jeronimo Conceição Ruiz. 2025. "Therapeutic Vaccines for Hematological Cancers: A Scoping Review of This Immunotherapeutic Approach as Alternative to the Treatment of These Malignancies" Vaccines 13, no. 2: 114. https://doi.org/10.3390/vaccines13020114
APA StyleMathias, F. A. S., Carvalho, M. G. R., & Ruiz, J. C. (2025). Therapeutic Vaccines for Hematological Cancers: A Scoping Review of This Immunotherapeutic Approach as Alternative to the Treatment of These Malignancies. Vaccines, 13(2), 114. https://doi.org/10.3390/vaccines13020114