Revisão Acesso aberto Revisado por pares

Personalized Cancer Vaccines: Clinical Landscape, Challenges, and Opportunities

2020; Elsevier BV; Volume: 29; Issue: 2 Linguagem: Inglês

10.1016/j.ymthe.2020.09.038

ISSN

1525-0024

Autores

Colby S. Shemesh, Joy Hsu, Iraj Hosseini, Ben‐Quan Shen, Anand Rotte, Patrick Twomey, Sandhya Girish, Benjamin Wu,

Tópico(s)

Cancer Immunotherapy and Biomarkers

Resumo

Tremendous innovation is underway among a rapidly expanding repertoire of promising personalized immune-based treatments. Therapeutic cancer vaccines (TCVs) are attractive systemic immunotherapies that activate and expand antigen-specific CD8+ and CD4+ T cells to enhance anti-tumor immunity. Our review highlights key issues impacting TCVs in clinical practice and reports on progress in development. We review the mechanism of action, immune-monitoring, dosing strategies, combinations, obstacles, and regulation of cancer vaccines. Most trials of personalized TCVs are ongoing and represent diverse platforms with predominantly early investigations of mRNA, DNA, or peptide-based targeting strategies against neoantigens in solid tumors, with many in combination immunotherapies. Multiple delivery systems, routes of administration, and dosing strategies are used. Intravenous or intramuscular administration is common, including delivery by lipid nanoparticles. Absorption and biodistribution impact antigen uptake, expression, and presentation, affecting the strength, speed, and duration of immune response. The emerging trials illustrate the complexity of developing this class of innovative immunotherapies. Methodical testing of the multiple potential factors influencing immune responses, as well as refined quantitative methodologies to facilitate optimal dosing strategies, could help resolve uncertainty of therapeutic approaches. To increase the likelihood of success in bringing these medicines to patients, several unique development challenges must be overcome. Tremendous innovation is underway among a rapidly expanding repertoire of promising personalized immune-based treatments. Therapeutic cancer vaccines (TCVs) are attractive systemic immunotherapies that activate and expand antigen-specific CD8+ and CD4+ T cells to enhance anti-tumor immunity. Our review highlights key issues impacting TCVs in clinical practice and reports on progress in development. We review the mechanism of action, immune-monitoring, dosing strategies, combinations, obstacles, and regulation of cancer vaccines. Most trials of personalized TCVs are ongoing and represent diverse platforms with predominantly early investigations of mRNA, DNA, or peptide-based targeting strategies against neoantigens in solid tumors, with many in combination immunotherapies. Multiple delivery systems, routes of administration, and dosing strategies are used. Intravenous or intramuscular administration is common, including delivery by lipid nanoparticles. Absorption and biodistribution impact antigen uptake, expression, and presentation, affecting the strength, speed, and duration of immune response. The emerging trials illustrate the complexity of developing this class of innovative immunotherapies. Methodical testing of the multiple potential factors influencing immune responses, as well as refined quantitative methodologies to facilitate optimal dosing strategies, could help resolve uncertainty of therapeutic approaches. To increase the likelihood of success in bringing these medicines to patients, several unique development challenges must be overcome. Therapeutic cancer vaccines (TCVs) have been heavily investigated in clinical trials for the past 50 years as investigational immunotherapies that aim to elicit new, or strengthen existing, CD8+ cytotoxic T cell lymphocyte (CTL) tumor antigen-specific responses.1Finn O.J. The dawn of vaccines for cancer prevention.Nat. Rev. Immunol. 2018; 18: 183-194Crossref PubMed Scopus (95) Google Scholar,2Falzone L. Salomone S. Libra M. Evolution of cancer pharmacological treatments at the turn of the third millennium.Front. Pharmacol. 2018; 9: 1300Crossref PubMed Scopus (243) Google Scholar As TCVs target antigens predominantly associated with tumor cells, this approach can be safer than other therapies by avoiding off-target effects. TCVs have evolved as a promising class of drugs in the immuno-oncology space, and they comprise a diverse set of antigens, adjuvants, delivery vectors, and administration methods.3Lopes A. Vandermeulen G. Préat V. Cancer DNA vaccines: current preclinical and clinical developments and future perspectives.J. Exp. Clin. Cancer Res. 2019; 38: 146Crossref PubMed Scopus (108) Google Scholar Historically, hundreds of TCV clinical trials including dozens of pivotal investigations were largely unsuccessful in demonstrating a clear clinical benefit.4Rahma O.E. Gammoh E. Simon R.M. Khleif S.N. Is the "3+3" dose-escalation phase I clinical trial design suitable for therapeutic cancer vaccine development? A recommendation for alternative design.Clin. Cancer Res. 2014; 20: 4758-4767Crossref PubMed Scopus (32) Google Scholar,5Tan A.C.L. Goubier A. Kohrt H.E. A quantitative analysis of therapeutic cancer vaccines in phase 2 or phase 3 trial.J. Immunother. Cancer. 2015; 3: 48Crossref PubMed Scopus (21) Google Scholar This is likely due to a combination of factors not limited to (1) suboptimal antigens, (2) lack of effective adjuvants, (3) poorly immunogenic platforms, and (4) an insufficient number of CTLs entering the tumor due to immunosuppression related to high disease burden, poor immune fitness, or an immunosuppressive tumor microenvironment.6van der Burg S.H. Correlates of immune and clinical activity of novel cancer vaccines.Semin. Immunol. 2018; 39: 119-136Crossref PubMed Scopus (28) Google Scholar,7Tran T. Blanc C. Granier C. Saldmann A. Tanchot C. Tartour E. Therapeutic cancer vaccine: building the future from lessons of the past.Semin. Immunopathol. 2019; 41: 69-85Crossref PubMed Scopus (28) Google Scholar Renewed investment and innovation are now underway, among a rapidly expanding repertoire of advanced TCV platforms. Among recent advances are personalized neoantigen-based TCVs with selective individualized antigens and new combination approaches to enhance immune activities compared to conventional TCVs against shared antigens.8Wirth T.C. Kühnel F. Neoantigen Targeting—dawn of a new era in cancer immunotherapy?.Front. Immunol. 2017; 8: 1848Crossref PubMed Scopus (50) Google Scholar, 9Zhang X. Sharma P.K. Peter Goedegebuure S. Gillanders W.E. Personalized cancer vaccines: targeting the cancer mutanome.Vaccine. 2017; 35: 1094-1100Crossref PubMed Scopus (43) Google Scholar, 10Sahin U. Türeci Ö. Personalized vaccines for cancer immunotherapy.Science. 2018; 359: 1355-1360Crossref PubMed Scopus (343) Google Scholar Given that neoantigen load has been correlated with response to existing immunotherapies,11Desrichard A. Snyder A. Chan T.A. Cancer neoantigens and applications for immunotherapy.Clin. Cancer Res. 2016; 22: 807-812Crossref PubMed Scopus (136) Google Scholar,12Łuksza 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: 517-520Crossref PubMed Scopus (292) Google Scholar these represent compelling targets for personalizing TCVs to enhance activity. More than 799 TCVs are in the global drug development pipeline as of 2019, with more than 400 active clinical trials.13Xin Yu J. Hubbard-Lucey V.M. Tang J. Immuno-oncology drug development goes global.Nat. Rev. Drug Discov. 2019; 18: 899-900Crossref PubMed Scopus (94) Google Scholar Of these, at least 23 are personalized vaccination approaches, which are well suited to investigate therapeutically as custom-tailored medicines in patients. Our review aims to provide a detailed account of the key components and common mechanisms of action of TCVs, while focusing on personalized TCVs, including (1) assessing the current clinical trial landscapes, (2) summarizing vaccination strategies, combination immunotherapies, common obstacles in development, and the regulatory framework of personalized TCVs, and, lastly, (3) by providing insight into additional development aspects important for the clinical development of personalized TCVs. Many of the mechanisms of action and biology relevant for TCVs also apply to personalized TCVs. To successfully induce anti-tumor T cell responses in the human body, TCVs act on and exploit multiple aspects of cancer immunity, including cancer antigen presentation, T cell priming and activation, recognition of cancer cells by T cells, and several effector mechanisms to eliminate tumor cells.14Chen D.S. Mellman I. Oncology meets immunology: the cancer-immunity cycle.Immunity. 2013; 39: 1-10Abstract Full Text Full Text PDF PubMed Scopus (2786) Google Scholar TCVs engage both innate and adaptive immunity with the use of an adjuvant and antigen to trigger an innate and adaptive response, respectively. Nonspecific innate immune responses are activated via pattern recognition receptors, such as Toll-like receptors, that recognize and respond to pathogen- or damage-associated molecular patterns. Engagement of these receptors activates transcription factor nuclear factor κB (NF-κB), stimulates cytokine and chemokine production, and recruits and activates lymphocytes.15Rezaei N. Keshavarz-Fathi M. Vaccines for Cancer Immunotherapy: An Evidence-Based Review on Current Status and Future Perspectives. Academic Press, 2018Google Scholar To induce adaptive CTL-mediated anti-tumor responses, TCVs must assist in (1) presentation and recognition of immunogenic tumor antigens by antigen-presenting cells (APCs); (2) recruitment, antigen processing, and maturation of APCs; (3) induced expression of T cell costimulatory signals and cytokines by APCs; (4) interaction of APCs with the adaptive immune system to prime and activate CD8+ T cells; and, lastly, (5) localization of these elements to the tumor.16Song Q. Zhang C.D. Wu X.H. Therapeutic cancer vaccines: from initial findings to prospects.Immunol. Lett. 2018; 196: 11-21Crossref PubMed Scopus (36) Google Scholar, 17Coventry B.J. Therapeutic vaccination immunomodulation: forming the basis of all cancer immunotherapy.Ther. Adv. Vaccines Immunother. 2019; 7 (2515135519862234)PubMed Google Scholar, 18Zhang J. Shi Z. Xu X. Yu Z. Mi J. The influence of microenvironment on tumor immunotherapy.FEBS J. 2019; 286: 4160-4175Crossref PubMed Scopus (35) Google Scholar A schematic of common TCV immune mechanisms to induce human anti-tumor T cell responses is provided in Figure 1. Through these processes TCVs can generate long-lasting immunological memory capable of controlling tumor growth and inhibiting relapse and metastasis. In preclinical studies, it has been shown that long-lived memory T cell responses regenerate effector T cells to eliminate tumor cells.19Romero P. Banchereau J. Bhardwaj N. Cockett M. Disis M.L. Dranoff G. Gilboa E. Hammond S.A. Hershberg R. Korman A.J. et al.The human vaccines project: a roadmap for cancer vaccine development.Sci. Transl. Med. 2016; 8: 334ps9Crossref PubMed Scopus (107) Google Scholar Unfortunately, TCVs have rarely met the criteria among the number of biological processes that must be engaged for a TCV to be efficacious; however, new approaches hold promise for improved performance.20Wong K.K. Li W.A. Mooney D.J. Dranoff G. Advances in therapeutic cancer vaccines.Adv. Immunol. 2016; 130: 191-249Crossref PubMed Scopus (66) Google Scholar To strengthen anti-tumor immunity, TCVs must activate tumor-associated antigen-specific CTLs, and thus targeting neoantigens expressed on a tumor cell surface via the use of a personalized TCV may be an effective strategy. The choice of target antigen is a major determinant of immunogenicity and takes advantage of distinctions between tumor and normal cells. Numerous approaches are used to identify one or multiple antigens for a TCV. These include selection of overexpressed or dysregulated tumor-associated proteins, such as the melanoma antigen gene (MAGE), New York esophageal squamous cell carcinoma 1 (NY-ESO01), or human epidermal growth factor receptor 2 (HER2) proteins, or by personalized approaches targeting cancer-associated neoantigens that arise by mutations found in a specific tumor.16Song Q. Zhang C.D. Wu X.H. Therapeutic cancer vaccines: from initial findings to prospects.Immunol. Lett. 2018; 196: 11-21Crossref PubMed Scopus (36) Google Scholar,21Jäger D. Jäger E. Knuth A. Immune responses to tumour antigens: implications for antigen specific immunotherapy of cancer.J. Clin. Pathol. 2001; 54: 669-674Crossref PubMed Scopus (149) Google Scholar Most TCVs to date have been directed against the first type, i.e., antigens overexpressed in malignant cells and expressed at lower levels in healthy tissues.1Finn O.J. The dawn of vaccines for cancer prevention.Nat. Rev. Immunol. 2018; 18: 183-194Crossref PubMed Scopus (95) Google Scholar CD4+ T cells play a complex and pivotal helper role orchestrating cancer immunity by regulating and priming antigen-specific CD8+ T cells (boosting function, magnitude, quality, persistence, and memory); additionally, these cells provide protective immunity through effector function, cytokine secretion, and activation of tumoricidal macrophages.22Pardoll D.M. Topalian S.L. The role of CD4+ T cell responses in antitumor immunity.Curr. Opin. Immunol. 1998; 10: 588-594Crossref PubMed Scopus (563) Google Scholar, 23Borst J. Ahrends T. Bąbała N. Melief C.J.M. Kastenmüller W. CD4+ T cell help in cancer immunology and immunotherapy.Nat. Rev. Immunol. 2018; 18: 635-647Crossref PubMed Scopus (391) Google Scholar, 24Tay 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; (Published online May 24, 2020)https://doi.org/10.1038/s41417-020-0183-xCrossref PubMed Scopus (94) Google Scholar Interferon (IFN)-γ release by CD4+ T cells is required for elimination of tumor cells25Mumberg D. Monach P.A. Wanderling S. Philip M. Toledano A.Y. Schreiber R.D. Schreiber H. CD4+ T cells eliminate MHC class II-negative cancer cells in vivo by indirect effects of IFN-γ.Proc. Natl. Acad. Sci. USA. 1999; 96: 8633-8638Crossref PubMed Scopus (295) Google Scholar,26Alspach E. Lussier D.M. Schreiber R.D. Interferon γ and its important roles in promoting and inhibiting spontaneous and therapeutic cancer immunity.Cold Spring Harb. Perspect. Biol. 2019; 11: a028480Crossref PubMed Scopus (104) Google Scholar, and immune attack against tumors depend on both CD4+ and CD8+ T cells, with depletion of either subset limiting tumor inhibition.27Alspach E. Lussier D.M. Miceli A.P. Kizhvatov I. DuPage M. Luoma A.M. Meng W. Lichti C.F. Esaulova E. Vomund A.N. et al.MHC-II neoantigens shape tumour immunity and response to immunotherapy.Nature. 2019; 574: 696-701Crossref PubMed Scopus (219) Google Scholar Mechanistic insights into the crosstalk between T lymphocyte subsets, as well as future work to optimize and modulate CD4+ T cells against specific tumor antigens, are likely crucial determinants for improved clinical response of personalized TCVs.27Alspach E. Lussier D.M. Miceli A.P. Kizhvatov I. DuPage M. Luoma A.M. Meng W. Lichti C.F. Esaulova E. Vomund A.N. et al.MHC-II neoantigens shape tumour immunity and response to immunotherapy.Nature. 2019; 574: 696-701Crossref PubMed Scopus (219) Google Scholar, 28Sillito F. Holler A. Stauss H.J. Engineering CD4+ T cells to enhance cancer immunity.Cells. 2020; 9: 1721Crossref Scopus (1) Google Scholar, 29Knutson K.L. Disis M.L. Tumor antigen-specific T helper cells in cancer immunity and immunotherapy.Cancer Immunol. Immunother. 2005; 54: 721-728Crossref PubMed Scopus (487) Google Scholar, 30Ostroumov D. Fekete-Drimusz N. Saborowski M. Kühnel F. Woller N. CD4 and CD8 T lymphocyte interplay in controlling tumor growth.Cell. Mol. Life Sci. 2018; 75: 689-713Crossref PubMed Scopus (166) Google Scholar, 31Kreiter 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: 692-696Crossref PubMed Scopus (640) Google Scholar TCVs currently in development rely on various methods of antigen delivery, including cell-based, protein/peptide-based, RNA- or DNA-based, and viral/bacterial-based approaches. Each of these has unique considerations related to manufacturing, delivery, antigen selection, immunogenicity, and tolerability.1Finn O.J. The dawn of vaccines for cancer prevention.Nat. Rev. Immunol. 2018; 18: 183-194Crossref PubMed Scopus (95) Google Scholar A wide spectrum of adjuvants may also be used to enhance immune responses.32Temizoz B. Kuroda E. Ishii K.J. Vaccine adjuvants as potential cancer immunotherapeutics.Int. Immunol. 2016; 28: 329-338Crossref PubMed Scopus (138) Google Scholar The optimal adjuvant must contain attributes to produce more immunity than the antigen alone and succeed at increasing cell-mediated immunity to an optimal amplitude, specificity, and effector profile, some of which can be optimized preclinically.32Temizoz B. Kuroda E. Ishii K.J. Vaccine adjuvants as potential cancer immunotherapeutics.Int. Immunol. 2016; 28: 329-338Crossref PubMed Scopus (138) Google Scholar,33Vermaelen K. Vaccine strategies to improve anti-cancer cellular immune responses.Front. Immunol. 2019; 10: 8Crossref PubMed Scopus (69) Google Scholar Most adjuvants activate damage- or pathogen-associated pattern recognition receptors, initiating a cascade of innate immune response aiding presentation of antigens on APCs. The multitude of diverse approaches for selection, engineering, packaging, and delivery of antigens, in conjunction with the complex biology, give rise to great complexity in comparing TCV platforms systematically. Neoantigen-based vaccines are individualized tumor-specific therapies,34Schumacher T.N. Scheper W. Kvistborg P. Cancer neoantigens.Annu. Rev. Immunol. 2019; 37: 173-200Crossref PubMed Scopus (176) Google Scholar typically targeting multiple tumor antigens unique to each patient. To identify and confirm expression of non-synonymous somatic mutations expressed in the tumor for inclusion in a personalized TCV, a biopsy of tumor tissue is taken for whole-exome and RNA sequencing. Mutations are analyzed using major histocompatibility complex (MHC) class I epitope prediction algorithms and prioritized. Ranked lists of candidate antigens are further refined based on in vitro binding assay results in which synthetic peptides are tested for binding to the appropriate class I human leukocyte antigen allele of interest.9Zhang X. Sharma P.K. Peter Goedegebuure S. Gillanders W.E. Personalized cancer vaccines: targeting the cancer mutanome.Vaccine. 2017; 35: 1094-1100Crossref PubMed Scopus (43) Google Scholar Selected neoantigens are tumor-specific and, hence, unlike tumor-associated antigens, neoantigen-specific T cells are less likely to have been eliminated during development of immune self-tolerance. This enhances their immunogenicity and ability to stimulate robust T cell responses and increases the breadth and diversity of the response.8Wirth T.C. Kühnel F. Neoantigen Targeting—dawn of a new era in cancer immunotherapy?.Front. Immunol. 2017; 8: 1848Crossref PubMed Scopus (50) Google Scholar Various types of variant mutations can be targeted by neoantigen-based vaccines. These include the common use of single nucleotide variants resulting from a single nucleotide change from one base to another, or indels as an insertion or deletion of a sequence of nucleotides from the genome resulting in a frameshift mutation that may alter protein function. Neoantigens selected may be of clonal origin present in all tumor cells or subclonal, which are present only in a subset, both of which influence immunoreactivity.35McGranahan N. Furness A.J.S. Rosenthal R. Ramskov S. Lyngaa R. Saini S.K. Jamal-Hanjani M. Wilson G.A. Birkbak N.J. Hiley C.T. et al.Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade.Science. 2016; 351: 1463-1469Crossref PubMed Scopus (1632) Google Scholar Mutations can also be classified by their role in tumor growth, and either passenger mutations lacking intrinsic growth advantages, or driver mutations that provide growth advantages selected during tumor evolution, can be incorporated into personalized TCVs.36Aurisicchio L. Pallocca M. Ciliberto G. Palombo F. The perfect personalized cancer therapy: cancer vaccines against neoantigens.J. Exp. Clin. Cancer Res. 2018; 37: 86Crossref PubMed Scopus (44) Google Scholar Patients harbor extensive variability in tumor neoantigen expression and clonality that gives rise to evasion of immune effectors and formation of resistance mechanisms, which are key challenges to reducing variability and increasing efficacy for immunotherapies such as TCVs.37Morris L.G. Riaz N. Desrichard A. Şenbabaoğlu Y. Hakimi A.A. Makarov V. Reis-Filho J.S. Chan T.A. Pan-cancer analysis of intratumor heterogeneity as a prognostic determinant of survival.Oncotarget. 2016; 7: 10051-10063Crossref PubMed Scopus (150) Google Scholar,38Shembrey C. Huntington N.D. Hollande F. Impact of tumor and immunological heterogeneity on the anti-cancer immune response.Cancers (Basel). 2019; 11: 1217Crossref Scopus (21) Google Scholar Tumors with high neoantigen intratumoral heterogeneity have a higher degree of branched mutations that give rise to an increased amount of subclones expressed with specific neoantigens, resulting in weaker neoantigen-specific T cell responses.39Fennemann F.L. de Vries I.J.M. Figdor C.G. Verdoes M. Attacking tumors from all sides: personalized multiplex vaccines to tackle intratumor heterogeneity.Front. Immunol. 2019; 10: 824Crossref PubMed Scopus (22) Google Scholar Provided that T cell infiltration and anti-tumor effect are related to selected antigens and the percentage of tumor cells expressing selected antigens, high fractions of subclonal neoantigens have had a negative impact on the response to immunotherapy.35McGranahan N. Furness A.J.S. Rosenthal R. Ramskov S. Lyngaa R. Saini S.K. Jamal-Hanjani M. Wilson G.A. Birkbak N.J. Hiley C.T. et al.Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade.Science. 2016; 351: 1463-1469Crossref PubMed Scopus (1632) Google Scholar Innovative multi-epitope approaches targeting more neoantigens by multiplexed personalized TCVs in addition to multi-regional tumor sampling that account for temporal changes following longitudinal liquid biopsy at follow-up may be key to combating tumor antigen heterogeneity,39Fennemann F.L. de Vries I.J.M. Figdor C.G. Verdoes M. Attacking tumors from all sides: personalized multiplex vaccines to tackle intratumor heterogeneity.Front. Immunol. 2019; 10: 824Crossref PubMed Scopus (22) Google Scholar,40Stanta G. Jahn S.W. Bonin S. Hoefler G. Tumour heterogeneity: principles and practical consequences.Virchows Arch. 2016; 469: 371-384Crossref PubMed Scopus (21) Google Scholar allowing for diverse targeting of both dominant subclones and low-abundance neoantigens to increase T cell reactivity. Following vaccination, augmentation of initial CTL responses through epitope spread leads to distinct immune responses to additional untargeted cryptic tumor antigens not present in TCVs, which have been reported in mouse and human cancers.31Kreiter 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: 692-696Crossref PubMed Scopus (640) Google Scholar,41GuhaThakurta D. Sheikh N.A. Fan L.Q. Kandadi H. Meagher T.C. Hall S.J. Kantoff P.W. Higano C.S. Small E.J. Gardner T.A. et al.Humoral immune response against nontargeted tumor antigens after treatment with sipuleucel-t and its association with improved clinical outcome.Clin. Cancer Res. 2015; 21: 3619-3630Crossref PubMed Scopus (85) Google Scholar, 42Corbière V. Chapiro J. Stroobant V. Ma W. Lurquin C. Lethé B. van Baren N. Van den Eynde B.J. Boon T. Coulie P.G. Antigen spreading contributes to MAGE vaccination-induced regression of melanoma metastases.Cancer Res. 2011; 71: 1253-1262Crossref PubMed Scopus (135) Google Scholar, 43Disis M.L. Gooley T.A. Rinn K. Davis D. Piepkorn M. Cheever M.A. Knutson K.L. Schiffman K. Generation of T-cell immunity to the HER-2/neu protein after active immunization with HER-2/neu peptide-based vaccines.J. Clin. Oncol. 2002; 20: 2624-2632Crossref PubMed Scopus (372) Google Scholar, 44Butterfield L.H. Ribas A. Dissette V.B. Amarnani S.N. Vu H.T. Oseguera D. Wang H.J. Elashoff R.M. McBride W.H. Mukherji B. et al.Determinant spreading associated with clinical response in dendritic cell-based immunotherapy for malignant melanoma.Clin. Cancer Res. 2003; 9: 998-1008PubMed Google Scholar, 45Wierecky J. Müller M.R. Wirths S. Halder-Oehler E. Dörfel D. Schmidt S.M. Häntschel M. Brugger W. Schröder S. Horger M.S. et al.Immunologic and clinical responses after vaccinations with peptide-pulsed dendritic cells in metastatic renal cancer patients.Cancer Res. 2006; 66: 5910-5918Crossref PubMed Scopus (191) Google Scholar These become additional targets of ongoing immune response important in control and protection against heterogeneous tumors in a robust, durable, and adaptive process that broadens and expands over time.46Gulley J.L. Madan R.A. Pachynski R. Mulders P. Sheikh N.A. Trager J. Drake C.G. Role of antigen spread and distinctive characteristics of immunotherapy in cancer treatment.J. Natl. Cancer Inst. 2017; 109: djw261Crossref Scopus (102) Google Scholar Correlations between epitope spread and tumor regression show significant value in immune response diversification by TCV therapies.47Ribas A. Timmerman J.M. Butterfield L.H. Economou J.S. Determinant spreading and tumor responses after peptide-based cancer immunotherapy.Trends Immunol. 2003; 24: 58-61Abstract Full Text Full Text PDF PubMed Scopus (89) Google Scholar Foundational pre-clinical work of tumor neoantigens led to the first proof-of-concept study in mice, which revealed that mice treated with a peptide-based neoantigen vaccine against mLama4 and mAlg8 using poly(IC) as an adjuvant conferred strong anti-tumor immunity and high response rates in treated animals.48Gubin 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: 577-581Crossref PubMed Scopus (1259) Google Scholar Further supportive preclinical efforts in other tumor models using other neoantigen-based vaccines have since confirmed anti-tumor immunity through de novo CD8+ T cell responses that are capable of inducing tumor rejection of aggressive tumors in mice, conferring a survival benefit.49Duperret E.K. Perales-Puchalt A. Stoltz R. G H H. Mandloi N. Barlow J. Chaudhuri A. Sardesai N.Y. Weiner D.B. A synthetic DNA, multi-neoantigen vaccine drives predominately MHC class I CD8+ T-cell responses, impacting tumor challenge.Cancer Immunol. Res. 2019; 7: 174-182Crossref PubMed Scopus (35) Google Scholar The anti-tumor efficacy of neoantigen-based cancer vaccines has been established with the first neoantigen vaccine clinical trials in patients with melanoma, and later in patients with glioblastoma, validating the potential benefit of neoantigen-based cancer vaccines as effective and even curative cancer therapies.50Carreno 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. Linette G.P. Cancer immunotherapy. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells.Science. 2015; 348: 803-808Crossref PubMed Scopus (721) Google Scholar, 51Ott 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: 217-221Crossref PubMed Scopus (1245) Google Scholar, 52Sahin 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: 222-226Crossref PubMed Scopus (11) Google Scholar, 53Keskin 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: 234-239Crossref PubMed Scopus (480) Google Scholar Now more than a dozen companies and academic institutions have partnered to explore personalized TCVs and have many studies ongoing or poised to begin enrolling patients. Advances in DNA/RNA sequencing, epitope prediction algorithms, and artificial intelligence are helping to identify more potent neoantigens. Additionally, optimization and expansion of manufacturing capacity is underway with the goal of supporting more widespread use of personalized TCVs.54Hundal J. Miller C.A. Griffith M. Griffith O.L. Walker J. Kiwala S. Graubert A. McMichael J. Coffman A. Mardis E.R. Cancer immunogenomics: computational neoantigen identification and vaccine design.Cold Spring Harb. Symp. Quant. Biol. 2016; 81: 105-111Crossref PubMed Scopus (13) Google Scholar,55Bräunlein E. Krackhardt A.M. Identification and characterization of neoantigens as well as respective immune responses in cancer patients.Front. Immunol. 2017; 8: 1702Crossref PubMed Scopus (29) Google Scholar Many personalized TCVs have entered this space with accelerated development plans, despite considerable investment risk and uncertainty regarding the best platform given the many unproven methodologies among diverse algorithms for neoantigen prediction. Additional questions remain around optimal delivery, dosing, and identifying the best therapeutic setting.56Guo Y. Lei K. Tang L. Neoantigen vaccine delivery for personalized anticance

Referência(s)