Skip to main content

REVIEW article

Front. Immunol., 16 August 2023
Sec. Vaccines and Molecular Therapeutics

Peptide-based vaccine for cancer therapies

Updated
  • Innovative Immunological Models Unit, Istituto Nazionale Tumori - IRCCS - “Fond G. Pascale”, Naples, Italy

Different strategies based on peptides are available for cancer treatment, in particular to counter-act the progression of tumor growth and disease relapse. In the last decade, in the context of therapeutic strategies against cancer, peptide-based vaccines have been evaluated in different tumor models. The peptides selected for cancer vaccine development can be classified in two main type: tumor-associated antigens (TAAs) and tumor-specific antigens (TSAs), which are captured, internalized, processed and presented by antigen-presenting cells (APCs) to cell-mediated immunity. Peptides loaded onto MHC class I are recognized by a specific TCR of CD8+ T cells, which are activated to exert their cytotoxic activity against tumor cells presenting the same peptide-MHC-I complex. This process is defined as active immunotherapy as the host’s immune system is either de novo activated or restimulated to mount an effective, tumor-specific immune reaction that may ultimately lead to tu-mor regression. However, while the preclinical data have frequently shown encouraging results, therapeutic cancer vaccines clinical trials, including those based on peptides have not provided satisfactory data to date. The limited efficacy of peptide-based cancer vaccines is the consequence of several factors, including the identification of specific target tumor antigens, the limited immunogenicity of peptides and the highly immunosuppressive tumor microenvironment (TME). An effective cancer vaccine can be developed only by addressing all such different aspects. The present review describes the state of the art for each of such factors.

1 Introduction

In 1991, van der Bruggen et al, Ludwig’s researchers in Brussels, published in Science a groundbreaking study on tumor antigens. They described for the first time that cytotoxic T cells can selectively recognize a tumor antigen expressed by the human melanoma (MAGE Ag) (1). This article represents a pillar oftumor immunology introducing the concept that such specific antigens may be used to design and develop an effective active immunotherapy. Since the discovery of MAGE, the field has moved forward rapidly. Scientists have discovered many other tumor antigens, of which many are being tested as targets for immunotherapy and in particular for therapeutic peptide-based cancer vaccines (2, 3).

Active immunotherapy is aimed either at amplifying the existing antitumor immune response by nonspecific proinflammatory molecules/adjuvants or at eliciting a specific de novo host immune response against selected tumor antigens by cancer vaccines. In particular, peptide-based cancer vaccines are able to activate the effector adaptive immune response as well as to provide long-term acquired immunity against a “foreign” tumor antigen. Indeed, cancer cells can be distinguished from normal cells by either upregulation/overexpression of endogenous proteins or mutation of those proteins. As a result, any mutated or differentially expressed protein in cancer cells can potentially represent a vaccine target. In particular, the antigens derived from overexpressed self proteins in tumor cells are defined tumor-associated antigens (TAAs) and might be shared among patients with the same tumor (4) (5, 6). To this class of antigens belong cell lineage differentiation antigens, which are normally not expressed in adult tissue (7, 8); and cancer/germline antigens (also known as cancer/testis) (9). Since TAAs are also expressed by normal cells, they may be subject to immunological tolerance and therefore may be poorly immunogenic. In contrast, antigens arising from cancer-related nonsynonymous mutations or other genetic alterations in cancer cells, named tumor-specific antigens (TSAs), are not expressed on the surface of normal cells and are specific to each type of tumors (1012). Consequently, TSAs are not subject to central and peripheral immune tolerance and are able to trigger a specific and effective T cell response against cancer cells (13).

The transformed cells are recognized by the immune system through immunosurveillance. Indeed, new antigens, including tumor antigens, are constantly presented by antigen-presenting cells (APC) to B and T lymphocytes of the adaptive immune response. The APCs can express the peptides through the MHC class I molecule for presentation to CD8+ T cells, or MHC class II molecule for presentation to CD4+ T helper cells. The latter may differentiate in two major subtypes, Th2 and Th1, involved in inflammatory response as well as in potentiating and sustaining the activity of CD8+ T cells (CTLs), respectively (14). Consequently, activated CD8+ T cells recognize cancer antigens expressed on the surface of tumor cells and initiate the release of apoptotic factors such as Perforin, Fas Ligand and Granzymes, leading to cell-mediated cytotoxicity (15). In this regard, the basic principles for a successful therapeutic vaccination against tumors include delivery of large amounts of high immunogenic antigens to APCs, induction of strong and sustained CD4+ T helper cell and CD8+ cytotoxic T lymphocyte (CTL) responses, infiltration of the TME as well as durability and maintenance of the immune response. In particular, strategies for improving activation and maturation of APCs, aiming at a more efficient antigen presentation to elicit an optimal T cell response, are actively pursued by several groups. Such strategies are mostly focused on the identification, selection and validation of novel adjuvants able to stimulate and enhance the magnitude and durability of antigen-specific T and B cell responses (16).Several therapeutic peptide-based vaccine strategies and formulations have been evaluated in different tumor types in the last 2 decades. However, only modest effects have been reported with an overall rate of clinical benefit of around 20% (17, 18). MUC1 represents a paradigmatic example of a TAA, highly expressed by adenocarcinomas, which has been evaluated in several human clinical trials with poor efficacy. In particular, MUC1 was targeted to dendritic cells, demonstrating the induction of highly specific immune response in preclinical and in human clinical trials (19). Subsequently, the ligand mannan used to target antigens in the mannose receptor lead to maturation of dendritic cells and activation via the Toll-Like Receptor 4 (2024). The National Cancer Institute Translational Research Working Group has listed MUC1 as the second promising target in cancer research from 75 tumor-related antigens (25). The distinctive biological structure of MUC1 and its aberrant glycosylation in cancer cells make it a recognized tumor-specific antigen on epithelial tumor cells (26, 27). In particular, MUC1 consists of both a C-terminal fragment (MUC1-C), highly conserved region, and an N-terminal fragment (MUC1-N), which contains the variable number tandem repeat (VNTR) region. The latter has been described to play a critical role in the MUC1 immunogenicity.

Therapeutic MUC1−based vaccines have been tested in numerous early stage clinical trials (28); ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02134925, but none of them has shown the expected anti-tumor effect in patients. In particular, these studies indicate that both the VNTR (MUC1-N) and non-VNTR (MUC1-C) regions contribute to immune evasion by cancer cells, which needs to be taken into consideration and addressed during the further development of MUC1-based cancer vaccines (2931).

To date, the only FDA-approved therapeutic cancer vaccine is Provenge® (Sipucleucel-T) for patients with castration-resistant prostate cancer (32). However, a modest increase in overall survival is observed together with a partial tumor regression. In addition to Sipucleucel-T vaccine, other peptide-based cancer vaccines are developed for prostate cancer, such as Cancer-associated membrane carbohydrates, including ganglioside (GM2), mucin 1 (MUC1), globo H and Thompson–Friedenreich antigen. Overall, five approaches - GVAX, DCVAC/PCa, a multi-epitope peptide vaccine, sipuleucel-T and PROSTVAC (33) - have been investigated in randomized phase III trials. They all showed safety and partial immunological activity but no effective clinical efficacy (34).

Such a poor efficacy of the therapeutic cancer vaccines developed for prostate cancer is common to most of the vaccines developed for other cancers. This is the consequence of a series of events that facilitate tumor development. Among these, the immunosuppressive TME plays a central role in cancer cell immune escape, inhibiting the activation of a specific T cell immune response against tumor cells (35). Moreover, during the tumor growth, the constant pressure from the adaptive immune system, coupled with the genetic instability of cancer cells, can select cellular sub-clones with reduced immunogenicity able to evade the immune recognition and destruction. Such low immunogenic sub-clones are often affected by loss of antigen presentation, due to a defective antigen- presenting machinery (36), hampering the recognition of tumor antigens by conventional (CD4+ and CD8+) and unconventional T cells (37). Moreover, chronically stimulated CD8+ T cells infiltrating the TME may acquire an ‘exhausted’ state characterized by loss of cytolytic activity, reduced cytokine production and reduced proliferation capacity.

To circumvent such adverse components of TME and improve their efficacy, cancer vaccines should be combined with immunomodulatory drugs (namely immunocheckpoint inhibitors, ICIs) to counterbalance the intra-tumor immune suppressive factors (3840).

In the present review, we provide an overview of biological mechanisms underlying peptide-based vaccine platform developed in the last years for cancer therapy. We also describe the identification, characterization and design of such antigens, their formulation, as well as different combinatorial strategies to optimize their antigenicity and overcome the immune evasion for more specific and effective immune response.

2 Peptides for cancer vaccine development

2.1 Peptide MHC interaction

The immunodominance is a key aspect to consider when planning a vaccination strategy, in particular when the vaccine formulation is based on few and short specific epitopes. In particular, the immunodominance of T cell determinants results from several factors: 1) intrinsic characteristics of the epitope, e.g. the binding affinity to MHC molecules; 2) the presence of appropriate MHC molecules and T cell receptors at the individual level; and 3) competition between different epitopes of a given protein antigen for the available MHC binding sites. The combination of all such factors will give rise to distinct immunodominant epitopes selectively driving the T cell response.

Thus, to maximize the efficacy of a vaccine based on a mix of peptides, it is relevant to ensure a balanced T cell response for all the peptides included in the mix. Otherwise, the less immunogenic epitopes would elicit an inefficient anti-cancer T cell response, with a resulting immunological escape of tumor cells.

Epitope specificity for T cells is mediated by the T-cell receptor (TCR), which binds peptides presented in the “peptide binding groove” of class I or class II major histocompatibility complexes (MHCs, also known as human leukocyte antigen, HLA, for humans). Whole proteins are internalized and proteolyzed by APCs and the resulting short peptides are loaded onto MHCs and presented on the APC surface. Consequently, specific TCRs may bind such peptide−MHC complexes (pMHC). Peptides binding the same HLA show a signature characterized by specific aminoacid residues at the positions interacting with the MHC groove (anchor residues). In particular, HLA-A2 - restricted epitopes show the xL/IxxxxxxV/L signature, where L/I and V/L represent the anchor residues (L: Leucine; I: isoleucine; V: Valine). Indeed, their side chains are oriented toward the interior of the peptide-binding groove and mediate the anchoring of the peptide to the MHC molecule (41, 42). The residues in the other positions of the epitope point toward the TCR and mediate the specificity of the interaction between the T cell and the target pMHC complex.

However, although the peptides adopt an optimal conformation in the groove of the MHC, there is no guarantee for their immunogenicity. Therefore, in order to improve the immunogenicity of a candidate antigen it is necessary to increase both the binding and the affinity to the MHC groove and to the T cell TCR, as described below.

2.2 Short versus long synthetic peptides

The sequence length of peptide vaccines is important to promote a strong immunogenic response. Typically, peptides can be either short (8 – 11 amino acids) or long (11 – 30 amino acids) for presentation in MHC class I or II molecules, respectively.

Short peptides represent the nominal epitope capable of binding the class I MHC molecules. They are attractive for vaccine development as they are easy to synthesize and cheap to produce in clinical grade. However, short peptides may bind to MHC of non-professional APCs, including B and T lymphocytes, which lack the secondary signaling machinery and cannot provide the full range of costimulatory signals required for complete T cell activation. Moreover, such non-conventional antigen presentation takes place in non-inflamed lymph nodes and in the absence of a strong pro-inflammatory context (43). This would lead to a poor T cell response or immune tolerance (43). Short peptides are strictly HLA-type restricted and consequently have to perfectly match the patient’s HLA, limiting their use to a specific subset of patients (44, 45). Furthermore, they are susceptible to fast exopeptidase-mediated degradation, which dramatically reduces their half-life (46, 47).

Nevertheless, short peptides represent exactly the natural HLA ligands on tumor cells and offer the possibility for precise immunomonitoring analyses (4851).

Alternatively, vaccines can be based on synthetic long peptides (SLPs). They cannot bind directly the MHC molecule and needs to be internalized and processed by professional APCs for presentation on both class I and class II MHC molecules. Consequently, SLPs can activate also the CD4+ TH cell response, providing helper factors (interferon γ [IFN-γ], tumor necrosis factor α, IL-2), essential for sustaining the cytotoxic CD8+ T cell responses and inducing the immune-mediated tumor cell killing (5254) (18). Lastly, they offer the possibility of including binding or recognition motifs for improved immunogenicity (55, 56).

Several early clinical trials based on SLPs have shown a good safety profile and promising results. For example, in three independent studies, SLP vaccination, based on HPV16 E6/E7 antigens, induced high levels of specific cytotoxic T lymphocytes, positively correlated with clinical benefit in premalignant disease (57, 58) and late-stage cervical cancer (59). In particular, the efficacy of SLP vaccine was potentiated when combined with immunomodulatory drugs (anti-PD-1 immune checkpoint and platinum-based chemotherapy) (59, 60).

Overall, an important condition for success of a cancer vaccine is the induction of a robust and sustained of both specific CD8+ and CD4+ T cells response as well as the increase in the CD4+:Treg ratio, to counteract the immunosuppressive TME.

2.3 Tumor associated antigens vs tumor specific antigens

In the last years, shared tumor antigens, also called Tumor associated antigens (TAAs), have been mostly adopted for cancer vaccine development. These include ‘self-antigens’ such as differentiation antigens (e.g., tyrosinase, gp100, MART-1), cancer/testis antigens (e.g., MAGE-A1, MAGE-A3, NY-ESO-1, and PRAME) and overexpressed antigens (79, 6165). These antigenic candidates have the advantage of being shared between patients with the same tumor type, but the disadvantage of being expressed also by normal cells. Consequently, they may elicit either a limited immune response, due to self-tolerance, or a potent autoimmune response, if the self-tolerance is broken. Therefore, the appropriate balance between such two extremes needs to be reached when peptide-based vaccines are based on TAAs (65).

More recently, attention has turned to ‘neoantigens’, that comprise mutated antigens arising in tumors by non-synonymous somatic mutations, insertions/deletions (INDEL) in the coding regions, frameshifts as occurring in microsatellite-instability-high tumors (66) and human endogenous retroviruses (67). Moreover, post-translational modifications such as phosphorylation (68), glycosylation (69) or methylation contribute to express new antigens on tumor cells. These types of antigens, also named tumor specific antigens (TSAs), are distinct from the corresponding wild-type self-antigens and are not affected by immunological tolerance. Therefore, a robust specific T cell response against these antigens can be elicited (70, 71). However, TSAs are strictly private to each individual cancer patient and their identification is laborious as well as expensive (13). Moreover, they are subject to immunoediting and escape. Therefore, the neoantigen repertoire is dynamic and may evolve in the course of tumor progression, requiring a pool of neoantigens in vaccine formulation for efficacy.

3 Identification and design of tumor antigens for cancer vaccine

3.1 Isolation of human leukocyte antigen (HLA) ligands

The separation of peptides from HLA molecules is usually accomplished by acid elution using HLA-specific antibodies. This process does not destroy the structure of the peptides, which are non-covalently bound to HLA molecules. However, acid elution of HLA-presented peptides can also be performed by simply incubating living cells in medium with a low pH and harvesting HLA ligands from the supernatant (72). The major disadvantage of the latter is that the harvested supernatant contains many other materials from the cells, which may interfere with the subsequent analysis. Surgically resected tumor tissues are the most suitable source, providing the whole array of the ligands presented by the cancer cells in vivo. Passages in vitro culture are not recommended in order to avoid possible changes in the HLA ligand repertoire, due to adaptation to cell culture conditions. Whenever possible, the tumor tissue should be ideally coupled by a sample of non-tumor tissue from the same organ for selecting only tumor-specific peptides.

In addition, blood can represent a potential source of soluble HLA molecules (sHLA), which can be regularly found in both healthy and diseased individuals, deriving from death or living cells or exosomes. In cancer patients, such blood-derived sHLA molecules have also been shown to contain peptides matching those identified in cancer cells. Therefore, in case of poor availability of tumor tissue, a “liquid biopsy” could provide useful information for cancer diagnosis and monitoring as well as identification of antigenic targets useful for developing therapeutic interventions (73, 74).

3.2 Immunopeptidome for human leukocyte antigen ligands identification

One of the methods of choice used for this purpose is based on a comprehensive analysis of the HLA ligandome of cancer cells (also termed immunopeptidome) to pursue tumor antigen discovery (75). It allows to identify and validate the “natural” presentation of new or well-known TAAs by the majority of primary tumors compared to normal tissues (Figure 1A) (76, 77).

FIGURE 1
www.frontiersin.org

Figure 1 Antigen discovery strategies. Tumor antigens are discovered by (A) Mass Spectometry analysis of eluted peptides; (B) integrated multi-omics screening; (C) in silico prediction. TAAs are wt epitopes over-expressed in cancer cells compared to normal cells. TSAs are mutated neo-antigens.

HLA molecules are precipitated from the tissue’s samples and HLA-bound peptides are eluted for MS analysis (78, 79). In particular, using high-performance liquid chromatography with very narrow long columns, peptide-HLA complex mixtures (pMHC), are separated before injection into the ionization chamber of the mass spectrometer.The ionized peptides are separated according to their mass-to-charge ratio in the mass filter, and then analyzed in the mass detector for their actual mass. Starting from the mass-to-charge ratio of the fragments it is possible to calculate the original peptide sequence. However, the probability of an incorrect assignment (false discovery rate) is high (8082). A principal challenge here is the complexity of data analysis. Prior to applying the identified peptides for vaccine development, they should be validated by comparison with synthetic, isotope-labeled peptides on the same equipment (83).

The MS identification of peptides should be always coupled with exome and transcriptome analyses to match the genetic and phenotypic results. The analysis of the HLA ligandome allowed to uncover another “universe” of sources for HLA ligands, derived from untranslated or not completely translated sequences. Such HLA ligands, named “cryptic peptides” (84) can be derived from noncoding RNAs, introns, or short open reading frames (85). Some of such non-conventional antigens can be consider cancer-specific (86). In order to identify the latter antigens, the proteogenomic approach has been defined by combining conventional proteomics with next-generation sequencing (NGS) (Figure 1B). In particular, merging WGS and RNASeq analyses with Ribosome profiling (Ribo-seq), it is possible to identify an excess of novel translated unannotated open reading frames. They can be derived from alternative out-of-frame ORFs, untranslated regions (UTRs) or long non-coding RNAs. Such non-coding transcripts could generate peptide sequences that are missing in conventional protein sequence repositories. Therefore, only the combination of genomics and proteomics on the same target cancer cells can lead to the identification of novel and more immunogenic HLA ligands, expanding the repertoire of targetable epitopes for cancer immunotherapy (8688).

3.3 Human leukocyte antigen ligand prediction tools

Relevant efforts have been committed to developing computational methods capable of accurately predicting peptide binding to both MHC-I and MHC-II [reviewed in (89)]. Indeed, the analysis of the HLA ligandome has shown a great limitation in sensitivity for identifying mutated neo-antigens, especially in tumors with low mutational burden (9092). Therefore, prediction algorithms are crucial for mutated neo-antigens discovery upon transcriptomic analysis (93). A global bioinformatics consortium was established in 2016, namely Tumor Neoantigen Selection Alliance (TESLA), which includes scientists, from academia, industry, and non-profit groups. Through predictive algorithms and machine learning, the most immunogenic neoantigens can be identified for stimulating strong immune responses (94). The TESLA consortium defined standardized predictive algorithms for neoantigens discovery, allowing to create more cancer immunotherapy treatments specific to each patient (https://www.synapse.org/#!Synapse:syn21048999).

In particular, the identification of large numbers of ligands retrieved from mass spectrometry (MS) experiments and their analysis for sequence similarities has allowed the development of algorithms based on machine learning (ML) methods for prediction of the ligand affinity to a specific MHC molecule [reviewed in (95)]. Each tool is based on different prediction methods [i.e. structure-based (SB) (96), motif matrix (MM) (97, 98), sequence motif (SM), quantitative affinity matrix (QAM) (99, 100), artificial neural network (ANN) (101, 102), support vector machine (SVM) (103, 104)]. Moreover, the choice of a specific tool is based on the characteristics of the antigen (TAA vs TSA) and the MHC molecule they bind (class I or II or both). These tools are able to address the distinction between MHC binders and non-binders as well as the prediction of the binding affinity of a peptide to MHC molecules. However, these methods are not yet proficient in deterministically estimating whether a given peptide is an epitope or not (105).

Among these tools, NetMHCpan is frequently used for MHC class I molecules and NetMHCIIpan for MHC class II (106). Both algorithms cover MHC molecules from a number of different species and many HLA alleles. Over the years, they have been continually improved (107, 108), but there are still many caveats in their prediction accuracy, especially for less frequent class I HLA alleles and for class II binding peptides (109).

In very recent studies, it has been reported that peptide presentation by MHC is strongly correlated with mRNA expression of the ligand’s source protein, underlying the importance of direct correlation between the source protein abundance and MHC epitope predictions (110115). For this reason, the epitope predictions can be further improved by considering also the abundance of peptides’ source proteins. Based on such assumption, the Antigen eXpression based Epitope Likelihood-Function (AXEL-F), together with TGCA expression data, has been shown to improve the prediction of neoantigens that are recognized by T cells (116).

4 Strategies for enhancing peptide immunogenicity: peptide vaccine formulation, delivery systems and immunogenic optimization

4.1 Formulation of peptide-based vaccines

Peptide-based vaccines represent the easiest strategy for eliciting the most focused anti-tumor T cell response, but an appropriate formulation needs to be developed. Indeed, in order to elicit a robust effector T cells response, peptide-based vaccines must deliver a sufficiently high dose to avoid the induction of T cell anergy (117). Moreover, free peptides are characterized by unfavorable pharmacokinetic properties, including short half-lives and low stability in vivo, which significantly reduces their immunogenicity To overcome such limitations, they need to be formulated with adjuvants (118).

Only a few adjuvants have been approved for human use to date (119), but several new molecules are under pre-clinical and early clinical development, including cytokines, saponins, mineral salts, emulsions, bacterial exotoxins, virosomes, liposomes, and immune-stimulating complexes (120). Several studies have revealed the central role of adjuvants in stimulating the innate immunity and, downstream, the antigen-specific adaptive T cell response. In particular, the ideal target for vaccine adjuvants is represented by the pattern recognition receptors (PRRs) of the innate immune system [reviewed in (121)]. Adjuvants which improve the inflammation, the delivery of antigens to DCs and their uptake and presentation to T cells, as well as increase their stability, include aluminum salts, nanoparticles, lipid vesicles, oil- and water-based formulations, and bacterial exotoxins. Among these, only alum has had limited use in cancer vaccines due to moderate activation of Th1 and CD8+ responses (122). On the contrary, oil-in-water emulsions has been shown to activate humoral and Th1 and Th2 immune responses, enhancing recruitment of granulocytes and DCs and the antigen uptake (123). Montanide, a mineral oil-based emulsion, is the most commonly used adjuvant, increasing the delivery of antigens to DCs and presentation to T cells. It has been and continues to be used in peptide cancer vaccine formulations in melanoma and renal carcinoma clinical trials (124).

Several TLR ligands have been extensively assessed as vaccine adjuvants in preclinical as well as human clinical setting, showing their ability to elicit a balanced humoral and cellular immune response [reviewed in (125)]. TLRs comprise a family of 10 receptors (126128) each of which is capable of recognizing unique pathogen-associated molecular patterns (PAMPs). These will activate downstream genes critical to promote innate immune responses and enhance immunity against the microbe (129). TLR agonists can be incorporated into peptide-based therapeutic cancer vaccines to induce Th1 type cellular proinflammatory cytokine production, such as type I IFNs, and the efficient activation and specific expansion of antigen-specific CTL (51, 130136). Among these, TLR4 ligands are known to enable a potent activation of APCs (137). Moreover, the TLR-9 agonist synthetic CpG oligodeoxynucleotide (CpG-ODN) was approved for the first time by the United States Food and Drug Administration for application in humans, in the Heplisav-B hepatitis B vaccine (138).

4.2 Delivery systems

Another key factor for the success of a peptide-based cancer vaccines is the capability of reaching the TME before degradation and being efficiently uptaken by APCs. For these aims, PLGA and Liposomes represent two examples of drug delivery systems, which have been approved by FDA after several years of experimental testing with a proven track-record in safety and biodegradability. (Food and Drug Admistration (FDA). (2021). Available at: www.fda.gov (139). Both delivery platforms can protect peptides from degradation and control their release to the spleen and lymph nodes, which contain a higher proportion of cross-presenting DCs (140, 141). In these sites, the particles are efficiently processed by DCs inducing a strong and sustained CD4+ T helper cell and cytotoxic T lymphocyte (CTL) responses (142). Indeed, their cationic charge and small size, efficiently promote a strong attraction and active uptake by DCs (143, 144). Besides PLGA and liposomes, other nanoparticles are tested as antigen delivery system, such as micelles, mesoporous silica nanoparticles (MSNs), gold nanoparticles (AuNPs) and virus nanoparticles. The delivery and the intra-cellular trafficking of nanoparticles have been optimized by selected modification of their surface (e.g., charge, structure, dimension and hydrophobicity) (145, 146) and by incorporating cell-penetrating peptides, APC-specific cellular epitopes or immune-stimulant lipid moieties (147).

Some of the nanoparticle vaccines have been evaluated in clinical trials for different cancers [reviewed in (148150)]. Among all, liposomes have been extensively used due to their versatility, including Tecemotide— liposomes (151, 152), AS15— lipids (153), DepoVax— liposomes (154), Iscomatrix (155), Cholesteryl pullulan (CHP) nanogels (156), and virus-like nanoparticles. However, although these trials have shown induction of specific antigen immune response, none of them have resulted in a statistically significant survival benefit. Such unsatisfactory results need to be thoroughly analyzed in order to identify strategies for improvements.

Furthermore, exosomes exhibit features for application as adjuvant carriers, such as optimal size, biocompatibility, stability in systemic circulation, and target-specific delivery (157). Zitvogel et al. (158) described that tumor peptide-pulsed dendritic cells (DCs) released DEXs (exosomes derived from dendritic cells) presenting tumor antigens on the membrane, which induced in vivo CTL priming and consequent tumor growth suppression. Such pillar study was the first to support the development of a novel cell-free vaccine using exosomes and subsequently confirmed by other groups (159). Moreover, DEXs have been shown to stimulate cells of the innate immune system, such as natural killer cells, and the production of INF-g (160). DEXs show several advantages over DCs: higher stability, due to their lipid composition, and higher number of peptide-MHC I and –MHC II complexes on their surface (161163). In addition, Wolfers and colleagues have reported that the Tumor Derived Exosomes (TDEs) represent a source of T-cell cross-priming, which are able to induce a CTL anti-tumor responses in vitro and in vivo (164).

Overall, these results highlight the novel use of exosomes as adjuvanting carriers for a future cancer vaccine development.

4.3 Peptide modification to improve the immunogenicity of antigenic targets

In order to improve the immunogenicity of tumor antigens, peptides can be modified to increase their (165)affinity and binding to the presenting MHC-I (166). Analogue peptides are designed by substituting amino acid residues in the epitope sequence to improve antigenicity and immunogenicity (167). Such modified peptides (heteroclitic peptides) have been shown to induce a more potent CD8+ T cell response, and break the immunological tolerance (86, 167170). Nevertheless, Tor B Stuge et al., have demonstrated that although vaccination with heteroclitic peptides may induce strong T cell responses, the recognition efficiencies of the wild type peptides may significantly vary. Indeed, most of such immune responses show a poor cytotoxic effect against melanoma cells expressing the wild type epitope (165).

A different approach for improving the immunogenicity of natural TAAs is to generate heteroclitic peptides with mutations in the TCR-binding residues (42, 171). In this regard, our group has shown that heteroclitic peptides modified in the TCR-binding residues of melanoma specific Trp2 TAA can improve control of tumor growth in a mouse model (172). In particular, the modification of TCR-facing amino acids, significantly improve the recognition by PBMCs of the HPV E7 wt epitope expressed on TC1 mouse lung tumor cell lines. Consequently, heteroclitic peptides are able to elicit even stronger immune response, cross-reacting with the parental wild type peptide. CTL elicited by the heteroclitic peptides show potent lytic activity on target cells expressing the wt peptide as well as control of tumor growth in vivo (173).

4.4 Antigenic molecular mimicry between microorganisms’ antigens and TAAs

Another strategy for improving the immunogenicity of tumor antigens is to identify novel universal shared antigens able to overcome the immune tolerance and elicit an effective T cell immune response. In this respect, non-self antigens derived from microorganisms (microorganisms-derived antigens, MoAs) with high sequence and structural homology with TAAs (molecular mimicry) can be highly useful.

The probability of homology between MoAs and human antigens and an overlapping peptidome representation is high and can result in a cross-reacting CD8+T cell responses. Indeed, similar epitopes can be targeted by the same CD8+ T cell receptor (TCR), given that a single TCR is cross-reactive with at least 10 (6) different MHC bound peptides (174, 175). Therefore, two unrelated antigens are very likely recognized by the same TCRs if the structural conformation of the entire epitopes is saved, sharing the same or conserved TCR facing central residues (176). This process may drive the fate of cancer development, progression and eventually response to therapy.

Preliminary evidences for T cells cross-reacting with microbial antigens and the homologous TAAs have been reported (177179). A more general evidence of molecular mimicry between TAAs and viral or microbiota-derived antigens has been reported by our group (176, 180182).

Overall, if tumor cells express a TAA similar to non-self MoA, it is possible to recall memory CD8+ T cells cross-reacting with tumor antigens, able to control the tumor growth. This may at last represent a relevant selective advantage for cancer patients and may lead to a novel preventive anti-cancer vaccine strategy (183).

5 Combinatorial strategies

The limited efficacy of cancer vaccines is consequent to immune-evasive mechanisms developing in the TME. These can be effectively targeted by combining different strategies based on immunotherapy and conventional anticancer therapies, such as standard or metronomic chemotherapy (MCT) and radiotherapy (184186) as well as the combination of Immune checkpoint inhibitors (ICIs). Indeed, the immunosuppressive TME leads to a limited infiltration of cancer-specific cytotoxic T lymphocytes and/or their functional anergy/exhaustion, resulting in a reduced efficacy of cancer vaccines.

5.1 Chemotherapy and vaccines

Numerous preclinical and clinical studies have been conducted in recent years to evaluate the efficacy of combinatorial strategies in enhancing the efficacy of the cytotoxic immune response induced by cancer vaccines.

Standard and metronomic chemotherapy, such as oxaliplatin and doxorubicin, induce positive immunomodulatory effects in the TME and can enhance the antitumor immune responses elicited by cancer vaccines, by inhibiting the immunosuppressive cells (i.e., Tregs and MDSCs) and increasing interferon gamma (IFN-γ) secreting CD8+ T cells. Moreover, it induces immunogenic cell death (ICD) in cancer cells, with the release of danger signals able to polarize dendritic cells (DCs) and activate an antitumor T helper 1 (Th1) responses. Consequently, the immunogenicity of tumor cells is modulated, becoming more susceptible to T-cell-mediated lysis (187189). Additionally, chemotherapy increases the tumor mutational burden leading to the release of an increased load of neoantigens into the TME (190195). Several pre-clinical studies, including from our group, have confirmed and demonstrated the immunomodulatory effects of standard and metronomic chemotherapy in potentiating the efficacy of a cancer vaccine (38, 196, 197). However, when the same protocols have been transferred in the clinical setting, partial results and non-lasting effect have been observed (49, 198, 199).The possible reasons for such failure are: (1) the choice of chemotherapy drugs, their correct dose, schedule and combination; and (2) the selection of patients’ cohort. Therefore, additional work is required to fully decode the pharmacological and immunological interactions between peptide-based anticancer vaccines and chemotherapy.

5.2 Radiotherapy and vaccines

Similar to chemotherapy, also radiotherapy (RT) can significantly increase the efficacy of vaccines. Indeed, it induces significant changes in the TME, with the release of both antigens from tumor cells and pro-inflammatory mediators. Overall, these effects increase tumor immune infiltration and trigger the innate as well as adaptive immune system (200, 201). The latter effect drives the systemic antitumor immunity which underlies the so-called “abscopal effect,” consisting in the capacity to eradicate distant metastasis (202206). Overall, immunologically ‘cold’ tumors are converted into ‘hot’ tumors. Moreover, RT is considered an “in situ vaccination”, because induces DNA damage, altered gene transcription and tumor neoantigen expression. Several studies have shown an increased anticancer immune responses when RT is administered both before and after a tumor-specific therapeutic vaccine. In particular, in human papillomavirus (HPV)- driven cancer models it has been shown that the combination of radiotherapy with vaccination strategies based on HPV E7 induces more efficient, DC maturation and peptide- specific T cell responses (207, 208).

5.3 Immune checkpoint inhibitors and vaccines

Immune checkpoint inhibitors (ICIs) are able to unleash the T cell response in the TME (209) and can reprogram the activation of T cells functionally exhausted by the chronic exposure to an antigen (117, 210, 211). Unleashing of antitumor T cells by ICIs represents the optimal strategy to complement anticancer vaccines. Indeed, anti-tumor T cells induced by the cancer vaccines would be more effective in a favorable TME generated by the ICIs. In particular, the immunomodulatory effect of ICI has been evaluated in a preclinical setting, showing the induction of CTL activation and the inhibition of immunosuppressive cells (MDSC and Tregs) (ref). In addition, several clinical trials have assessed cancer vaccines combined with a checkpoint inhibitor, such as anti-CTLA-4, PD-1, or PD-L1 (https://clinicaltrials.gov/ct2/results?term5cancer1vaccines+%261immune+checkpoint+inhibitors&Search=Apply&recrs=a&age_v=&gndr=&type=&rslt).

In details, the most significant results were observed in two clinical trials with T-VEC and sipuleucel-T vaccines, where the combination with ICIs induced higher antitumor activity and objective response rate (ORR) compared to the ICIs alone (212, 213). The combination of SLP vaccine ISA101 and anti-PD-1 immune checkpoint nivolumab was shown to be well tolerated in 24 patients with HPV-16–positive cancer. Moreover, the efficacy of combinatorial strategy appeared superior to that of nivolumab monotherapy (59). Other clinical trials have assessed the combination of ICI and cancer vaccines with contrasting efficacy data. Additional studies are required to identify the most appropriate clinical settings for optimal efficacy.

5.4 IDO Inhibitors

Indoleamine 2,3-dioxygenase (IDO) 1, an enzyme catabolizing tryptophan to kynurenine, generates immunosuppression in TME and his over-expression is correlated with tumor progression (214). In particular, several preclinical studies have shown that over expression of IDO1 correlates with an increase of Tregs and MDSC cells, and upregulation of PD-1 in cytotoxic T cells (215). In different tumor settings (i.e. colon cancer, cervical cancer, melanoma) high level of IDO1 was associated with poorer outcomes (216, 217). For these reasons, therapeutic strategies based on IDO1 inhibitors combined with a cancer vaccine, could be a promising approach. Indeed, small-molecule inhibitors of IDO 1 as well as peptide vaccines derived from IDO were tested in early phase clinical trials in combination with ICIs, demonstrating the safety and promising immune and clinical responses (NCT05155254) (218, 219).

6 Conclusions and future prospective

Over the years, various efforts have been made in developing cancer peptide vaccines, but their effectiveness has been limited. Several reasons are responsible for such failures, including the selection of poor immunogenic target antigens and the strong immunosuppressive TME. Strategies to address and overcome each of such aspects are currently pursued in the field. Derivatives of TAAs are designed (e.g. heteroclitic peptides, homologous non-self antigens) and TSAs are selected to improve the antigen immunogenicity. The latter include both mutation-derived as well as unconventional antigens, which, however, represent a great technological challenge and are currently of limited feasibility on a large scale. Likewise, combination treatments (e.g. vaccines with chemotherapy or radiotherapy or ICIs) are evaluated to counterbalance the immunosuppressive TME (Figure 2). In addition, inefficient delivery systems, patients’ selection and loss of MHC class-I may contribute to failures, but these aspects are beyond the scope of the present review article (144, 220). Consequently, several issues remain to be solved.

FIGURE 2
www.frontiersin.org

Figure 2 Strategies for improving cancer vaccine immunogenicity. Immunogenicity can be improved by (A) identification/generation of TAA-like non-self antigens; (B) design of combinatorial strategies including vaccines and standard and IO therapies; (C) antigen formulation in Th-1 driving adjuvants.

Indeed, although a significant number of early stage clinical trials,based on peptide-based vaccines, are currently recruiting around the World (nr. 64 as of November 2022), a single trial is beyond phase II (NCT03284866) (https://clinicaltrials.gov/ct2/results?term=peptide+cancer+vaccine&recrs=a&age_v=&gndr=&type=&rslt=&Search=Apply). This confirms the limited efficacy of peptide-based cancer vaccines.

Therefore, further studies are necessary to clarify how to identify new efficient strategies able to give to peptide-based cancer vaccines a new horizon.

Author contributions

MT and LB conceived and drafted the review. All authors contributed to the article and approved the submitted version.

Funding

The study was funded by the Italian Ministry of Health through Institutional “Ricerca Corrente” (LB and MT) L2/3 project to LB “Tumor antigen discovery for innovative cancer immunotherapies in HCC: from benchside to bedside (HepAnt)”. L2/13 project to MT “Peptide optimization to improve antigen presentation and anti-tumor response for sarcomas”.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den Eynde B, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science (1991) 254:1643–7. doi: 10.1126/science.1840703

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Henderson RA, Finn OJ. Human tumor antigens are ready to fly. Adv Immunol (1996) 62:217–56. doi: 10.1016/s0065-2776(08)60431-9

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Graziano DF, Finn OJ. Tumor antigens and tumor antigen discovery. Cancer Treat Res (2005) 123:89–111. doi: 10.1007/0-387-27545-2_4

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Vonderheide RH, Hahn WC, Schultze JL, Nadler LM. The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes. Immunity (1999) 10:673–9. doi: 10.1016/s1074-7613(00)80066-7

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Disis ML, Wallace DR, Gooley TA, Dang Y, Slota M, Lu H, et al. Concurrent trastuzumab and HER2/neu-specific vaccination in patients with metastatic breast cancer. J Clin Oncol (2009) 27:4685–92. doi: 10.1200/JCO.2008.20.6789

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Finn OJ, Gantt KR, Lepisto AJ, Pejawar-Gaddy S, Xue J, Beatty PL. Importance of MUC1 and spontaneous mouse tumor models for understanding the immunobiology of human adenocarcinomas. Immunol Res (2011) 50:261–8. doi: 10.1007/s12026-011-8214-1

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Parkhurst MR, Fitzgerald EB, Southwood S, Sette A, Rosenberg SA, Kawakami Y. Identification of a shared HLA-A*0201-restricted T-cell epitope from the melanoma antigen tyrosinase-related protein 2 (TRP2). Cancer Res (1998) 58:4895–901.

PubMed Abstract | Google Scholar

8. Correale P, Walmsley K, Nieroda C, Zaremba S, Zhu M, Schlom J, et al. In vitro generation of human cytotoxic T lymphocytes specific for peptides derived from prostate-specific antigen. J Natl Cancer Inst (1997) 89:293–300. doi: 10.1093/jnci/89.4.293

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Hofmann O, Caballero OL, Stevenson BJ, Chen YT, Cohen T, Chua R, et al. Genome-wide analysis of cancer/testis gene expression. Proc Natl Acad Sci U.S.A. (2008) 105:20422–7. doi: 10.1073/pnas.0810777105

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Hollingsworth RE, Jansen K. Turning the corner on therapeutic cancer vaccines. NPJ Vaccines (2019) 4:7. doi: 10.1038/s41541-019-0103-y

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Vigneron N. Human tumor antigens and cancer immunotherapy. BioMed Res Int (2015) 2015, 948501. doi: 10.1155/2015/948501

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Tagliamonte M, Petrizzo A, Tornesello ML, Buonaguro FM, Buonaguro L. Antigen-specific vaccines for cancer treatment. Hum Vaccin Immunother (2014) 10:3332–46. doi: 10.4161/21645515.2014.973317

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Schumacher TN, Scheper W, Kvistborg P. Cancer neoantigens. Annu Rev Immunol (2019) 37:173–200. doi: 10.1146/annurev-immunol-042617-053402

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Hoebe K, Janssen E, Beutler B. The interface between innate and adaptive immunity. Nat Immunol (2004) 5:971–4. doi: 10.1038/ni1004-971

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Halle S, Halle O, Förster R. Mechanisms and dynamics of T cell-mediated cytotoxicity in Vivo. Trends Immunol (2017) 38:432–43. doi: 10.1016/j.it.2017.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Pulendran B, A. P,S, O'Hagan DT. Emerging concepts in the science of vaccine adjuvants. Nat Rev Drug Discovery (2021) 20:454–75. doi: 10.1038/s41573-021-00163-y

CrossRef Full Text | Google Scholar

17. Melero I, Gaudernack G, Gerritsen W, Huber C, Parmiani G, Scholl S, et al. Therapeutic vaccines for cancer: an overview of clinical trials. Nat Rev Clin Oncol (2014) 11:509–24. doi: 10.1038/nrclinonc.2014.111

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Melief CJ, van Hall T, Arens R, Ossendorp F, van der Burg SH. Therapeutic cancer vaccines. J Clin Invest (2015) 125:3401–12. doi: 10.1172/JCI80009

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Goydos JS, Elder E, Whiteside TL, Finn OJ, Lotze MT. A phase I trial of a synthetic mucin peptide vaccine. Induction of specific immune reactivity in patients with adenocarcinoma. J Surg Res (1996) 63:298–304. doi: 10.1006/jsre.1996.0264

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Apostolopoulos V, Thalhammer T, Tzakos AG, Stojanovska L. Targeting antigens to dendritic cell receptors for vaccine development. J Drug Delivery (2013) 2013, 869718. doi: 10.1155/2013/869718

CrossRef Full Text | Google Scholar

21. Sheng KC, Pouniotis DS, Wright MD, Tang CK, Lazoura E, Pietersz GA, et al. Mannan derivatives induce phenotypic and functional maturation of mouse dendritic cells. Immunology (2006) 118:372–83. doi: 10.1111/j.1365-2567.2006.02384.x

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Apostolopoulos V, Stojanovska L, Gargosky SE. MUC1 (CD227): a multi-tasked molecule. Cell Mol Life Sci (2015) 72:4475–500. doi: 10.1007/s00018-015-2014-z

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Tang CK, Katsara M, Apostolopoulos V. Strategies used for MUC1 immunotherapy: human clinical studies. Expert Rev Vaccines (2008) 7:963–75. doi: 10.1586/14760584.7.7.963

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Tang CK, Sheng KC, Pouniotis D, Esparon S, Son HY, Kim CW, et al. Oxidized and reduced mannan mediated MUC1 DNA immunization induce effective anti-tumor responses. Vaccine (2008) 26:3827–34. doi: 10.1016/j.vaccine.2008.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, et al. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res (2009) 15:5323–37. doi: 10.1158/1078-0432.CCR-09-0737

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Hossain MK, Wall KA. Immunological evaluation of recent MUC1 glycopeptide cancer vaccines. Vaccines (Basel) (2016) 4. doi: 10.3390/vaccines4030025

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Hollingsworth MA, Swanson BJ. Mucins in cancer: protection and control of the cell surface. Nat Rev Cancer (2004) 4:45–60. doi: 10.1038/nrc1251

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Kimura T, Finn OJ. MUC1 immunotherapy is here to stay. Expert Opin Biol Ther (2013) 13:35–49. doi: 10.1517/14712598.2012.725719

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Rajabi H, Kufe D. MUC1-C oncoprotein integrates a program of EMT, epigenetic reprogramming and immune evasion in human carcinomas. Biochim Biophys Acta Rev Cancer (2017) 1868:117–22. doi: 10.1016/j.bbcan.2017.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Bouillez A, Rajabi H, Jin C, Samur M, Tagde A, Alam M, et al. MUC1-C integrates PD-L1 induction with repression of immune effectors in non-small-cell lung cancer. Oncogene (2017) 36:4037–46. doi: 10.1038/onc.2017.47

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Bouillez A, Adeegbe D, Jin C, Hu X, Tagde A, Alam M, et al. MUC1-C promotes the suppressive immune microenvironment in non-small cell lung cancer. Oncoimmunology (2017) 6:e1338998. doi: 10.1080/2162402X.2017.1338998

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med (2010) 363:411–22. doi: 10.1056/NEJMoa1001294

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Gulley JL, Madan RA, Tsang KY, Jochems C, Marte JL, Farsaci B, et al. Immune impact induced by PROSTVAC (PSA-TRICOM), a therapeutic vaccine for prostate cancer. Cancer Immunol Res (2014) 2:133–41. doi: 10.1158/2326-6066.CIR-13-0108

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Arlen PM, Skarupa L, Pazdur M, Seetharam M, Tsang KY, Grosenbach DW, et al. Clinical safety of a viral vector based prostate cancer vaccine strategy. J Urol (2007) 178:1515–20. doi: 10.1016/j.juro.2007.05.117

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Hatziioannou A, Alissafi T, Verginis P. Myeloid-derived suppressor cells and T regulatory cells in tumors: unraveling the dark side of the force. J Leukoc Biol (2017) 102:407–21. doi: 10.1189/jlb.5VMR1116-493R

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Campoli M, Ferrone S. HLA antigen changes in Malignant cells: epigenetic mechanisms and biologic significance. Oncogene (2008) 27:5869–85. doi: 10.1038/onc.2008.273

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Godfrey DI, Le Nours J, Andrews DM, Uldrich AP, Rossjohn J. Unconventional T cell targets for cancer immunotherapy. Immunity (2018) 48:453–73. doi: 10.1016/j.immuni.2018.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Petrizzo A, Mauriello A, Luciano A, Rea D, Barbieri A, Arra C, et al. Inhibition of tumor growth by cancer vaccine combined with metronomic chemotherapy and anti-PD-1 in a pre-clinical setting. Oncotarget (2018) 9:3576–89. doi: 10.18632/oncotarget.23181

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Lee KL, Schlom J, Hamilton DH. Combination therapies utilizing neoepitope-targeted vaccines. Cancer Immunol Immunother (2021) 70:875–85. doi: 10.1007/s00262-020-02729-y

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Kerr MD, McBride DA, Chumber AK, Shah NJ. Combining therapeutic vaccines with chemo- and immunotherapies in the treatment of cancer. Expert Opin Drug Discovery (2021) 16:89–99. doi: 10.1080/17460441.2020.1811673

CrossRef Full Text | Google Scholar

41. Garboczi DN, Ghosh P, Utz U, Fan QR, Biddison WE, Wiley DC. Structure of the complex between human T-cell receptor, viral peptide and HLA-A2. Nature (1996) 384:134–41. doi: 10.1038/384134a0

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Rammensee HG, Friede T, Stevanoviíc S. MHC ligands and peptide motifs: first listing. Immunogenetics (1995) 41:178–228. doi: 10.1007/bf00172063

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Bijker MS, van den Eeden SJ, Franken KL, Melief CJ, van der Burg SH, Offringa R. Superior induction of anti-tumor CTL immunity by extended peptide vaccines involves prolonged, DC-focused antigen presentation. Eur J Immunol (2008) 38:1033–42. doi: 10.1002/eji.200737995

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Sidney J, del Guercio MF, Southwood S, Engelhard VH, Appella E, Rammensee HG, et al. Several HLA alleles share overlapping peptide specificities. J Immunol (1995) 154:247–59. doi: 10.4049/jimmunol.154.1.247

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Southwood S, Sidney J, Kondo A, del Guercio MF, Appella E, Hoffman S, et al. Several common HLA-DR types share largely overlapping peptide binding repertoires. J Immunol (1998) 160:3363–73. doi: 10.4049/jimmunol.160.7.3363

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Diao L, Meibohm B. Pharmacokinetics and pharmacokinetic-pharmacodynamic correlations of therapeutic peptides. Clin Pharmacokinet (2013) 52:855–68. doi: 10.1007/s40262-013-0079-0

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Di L, Obach RS. Addressing the challenges of low clearance in drug research. AAPS J (2015) 17:352–7. doi: 10.1208/s12248-014-9691-7

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Falk K, Rötzschke O, Stevanović S, Jung G, Rammensee HG. Allele-specific motifs revealed by sequencing of self-peptides eluted from MHC molecules. Nature (1991) 351:290–6. doi: 10.1038/351290a0

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Walter S, Weinschenk T, Stenzl A, Zdrojowy R, Pluzanska A, Szczylik C, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med (2012) 18:1254–61. doi: 10.1038/nm.2883

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Feyerabend S, Stevanovic S, Gouttefangeas C, Wernet D, Hennenlotter J, Bedke J, et al. Novel multi-peptide vaccination in Hla-A2+ hormone sensitive patients with biochemical relapse of prostate cancer. Prostate (2009) 69:917–27. doi: 10.1002/pros.20941

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Löffler MW, Gori S, Izzo F, Mayer-Mokler A, Ascierto PA, Königsrainer A, et al. Phase I/II multicenter trial of a novel therapeutic cancer vaccine, hepaVac-101, for hepatocellular carcinoma. Clin Cancer Res (2022) 28:2555–66. doi: 10.1158/1078-0432.Ccr-21-4424

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Hung K, Hayashi R, Lafond-Walker A, Lowenstein C, Pardoll D, Levitsky H. The central role of CD4(+) T cells in the antitumor immune response. J Exp Med (1998) 188:2357–68. doi: 10.1084/jem.188.12.2357

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Matsui S, Ahlers JD, Vortmeyer AO, Terabe M, Tsukui T, Carbone DP, et al. A model for CD8+ CTL tumor immunosurveillance and regulation of tumor escape by CD4 T cells through an effect on quality of CTL. J Immunol (1999) 163:184–93. doi: 10.4049/jimmunol.163.1.184

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Pardoll DM. Inducing autoimmune disease to treat cancer. Proc Natl Acad Sci U.S.A. (1999) 96:5340–2. doi: 10.1073/pnas.96.10.5340

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Ciesielski MJ, Ahluwalia MS, Munich SA, Orton M, Barone T, Chanan-Khan A, et al. Antitumor cytotoxic T-cell response induced by a survivin peptide mimic. Cancer Immunol Immunother (2010) 59:1211–21. doi: 10.1007/s00262-010-0845-x

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Ciesielski MJ, Kozbor D, Castanaro CA, Barone TA, Fenstermaker RA. Therapeutic effect of a T helper cell supported CTL response induced by a survivin peptide vaccine against murine cerebral glioma. Cancer Immunol Immunother (2008) 57:1827–35. doi: 10.1007/s00262-008-0510-9

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-van der Meer DM, Vloon AP, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med (2009) 361:1838–47. doi: 10.1056/NEJMoa0810097

PubMed Abstract | CrossRef Full Text | Google Scholar

58. van Poelgeest MI, Welters MJ, Vermeij R, Stynenbosch LF, Loof NM, Berends-van der Meer DM, et al. Vaccination against oncoproteins of HPV16 for noninvasive vulvar/vaginal lesions: Lesion clearance is related to the strength of the T-cell response. Clin Cancer Res (2016) 22:2342–50. doi: 10.1158/1078-0432.CCR-15-2594

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Massarelli E, William W, Johnson F, Kies M, Ferrarotto R, Guo M, et al. Combining immune checkpoint blockade and tumor-specific vaccine for patients with incurable human papillomavirus 16-related cancer: A phase 2 clinical trial. JAMA Oncol (2019) 5:67–73. doi: 10.1001/jamaoncol.2018.4051

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Melief CJM, Welters MJP, Vergote I, Kroep JR, Kenter GG, Ottevanger PB, et al. Strong vaccine responses during chemotherapy are associated with prolonged cancer survival. Sci Transl Med (2020) 12. doi: 10.1126/scitranslmed.aaz8235

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Pavlick A, Blazquez AB, Meseck M, Lattanzi M, Ott PA, Marron TU, et al. Combined vaccination with NY-ESO-1 protein, poly-ICLC, and montanide improves humoral and cellular immune responses in patients with high-risk melanoma. Cancer Immunol Res (2020) 8:70–80. doi: 10.1158/2326-6066.Cir-19-0545

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Comiskey MC, Dallos MC, Drake CG. Immunotherapy in prostate cancer: Teaching an old dog new tricks. Curr Oncol Rep (2018) 20:75. doi: 10.1007/s11912-018-0712-z

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Buonaguro L, Tagliamonte M. Selecting target antigens for cancer vaccine development. Vaccines (Basel) (2020) 8. doi: 10.3390/vaccines8040615

CrossRef Full Text | Google Scholar

64. Karbach J, Neumann A, Atmaca A, Wahle C, Brand K, von Boehmer L, et al. Efficient in vivo priming by vaccination with recombinant NY-ESO-1 protein and CpG in antigen naive prostate cancer patients. Clin Cancer Res (2011) 17:861–70. doi: 10.1158/1078-0432.Ccr-10-1811

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Buonaguro L, Petrizzo A, Tornesello ML, Buonaguro FM. Translating tumor antigens into cancer vaccines. Clin Vaccine Immunol (2011) 18:23–34. doi: 10.1128/cvi.00286-10

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Roudko V, Bozkus CC, Orfanelli T, McClain CB, Carr C, O'Donnell T, et al. Shared immunogenic poly-epitope frameshift mutations in microsatellite unstable tumors. Cell (2020) 183:1634–1649.e1617. doi: 10.1016/j.cell.2020.11.004

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Smith CC, Selitsky SR, Chai S, Armistead PM, Vincent BG, Serody JS. Alternative tumour-specific antigens. Nat Rev Cancer (2019) 19:465–78. doi: 10.1038/s41568-019-0162-4

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Engelhard VH, Obeng RC, Cummings KL, Petroni GR, Ambakhutwala AL, Chianese-Bullock KA, et al. MHC-restricted phosphopeptide antigens: preclinical validation and first-in-humans clinical trial in participants with high-risk melanoma. J Immunother Cancer (2020) 8. doi: 10.1136/jitc-2019-000262

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Malaker SA, Penny SA, Steadman LG, Myers PT, Loke JC, Raghavan M, et al. Identification of glycopeptides as posttranslationally modified neoantigens in leukemia. Cancer Immunol Res (2017) 5:376–84. doi: 10.1158/2326-6066.Cir-16-0280

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Gubin MM, Artyomov MN, Mardis ER, Schreiber RD. Tumor neoantigens: building a framework for personalized cancer immunotherapy. J Clin Invest (2015) 125:3413–21. doi: 10.1172/jci80008

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Hu Z, Ott PA, Wu CJ. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat Rev Immunol (2018) 18:168–82. doi: 10.1038/nri.2017.131

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Sturm T, Sautter B, Wörner TP, Stevanović S, Rammensee HG, Planz O, et al. Mild acid elution and MHC immunoaffinity chromatography reveal similar albeit not identical profiles of the HLA class I immunopeptidome. J Proteome Res (2021) 20:289–304. doi: 10.1021/acs.jproteome.0c00386

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Bassani-Sternberg M, Barnea E, Beer I, Avivi I, Katz T, Admon A. Soluble plasma HLA peptidome as a potential source for cancer biomarkers. Proc Natl Acad Sci U.S.A. (2010) 107:18769–76. doi: 10.1073/pnas.1008501107

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Kessler AL, Bruno MJ, Buschow SI. The potential of soluble human leukocyte antigen molecules for early cancer detection and therapeutic vaccine design. Vaccines (Basel) (2020) 8. doi: 10.3390/vaccines8040775

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Weinschenk T, Gouttefangeas C, Schirle M, Obermayr F, Walter S, Schoor O, et al. Integrated functional genomics approach for the design of patient-individual antitumor vaccines. Cancer Res (2002) 62:5818–27.

PubMed Abstract | Google Scholar

76. Singh-Jasuja H, Emmerich NP, Rammensee HG. The Tübingen approach: identification, selection, and validation of tumor-associated HLA peptides for cancer therapy. Cancer Immunol Immunother (2004) 53:187–95. doi: 10.1007/s00262-003-0480-x

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Rammensee HG, Weinschenk T, Gouttefangeas C, Stevanović S. Towards patient-specific tumor antigen selection for vaccination. Immunol Rev (2002) 188:164–76. doi: 10.1034/j.1600-065x.2002.18815.x

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Rammensee HG, Falk K, Rötzschke O. Peptides naturally presented by MHC class I molecules. Annu Rev Immunol (1993) 11:213–44. doi: 10.1146/annurev.iy.11.040193.001241

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Kowalewski DJ, Stevanović S. Biochemical large-scale identification of MHC class I ligands. Methods Mol Biol (2013) 960:145–57. doi: 10.1007/978-1-62703-218-6_12

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Nelde A, Kowalewski DJ, Stevanović S. Purification and identification of naturally presented MHC class I and II ligands. Methods Mol Biol (2019) 1988:123–36. doi: 10.1007/978-1-4939-9450-2_10

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Bichmann L, Nelde A, Ghosh M, Heumos L, Mohr C, Peltzer A, et al. MHCquant: Automated and reproducible data analysis for immunopeptidomics. J Proteome Res (2019) 18:3876–84. doi: 10.1021/acs.jproteome.9b00313

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Pak H, Michaux J, Huber F, Chong C, Stevenson BJ, Müller M, et al. Sensitive immunopeptidomics by leveraging available large-scale multi-HLA spectral libraries, data-independent acquisition, and MS/MS prediction. Mol Cell Proteomics (2021) 20:100080. doi: 10.1016/j.mcpro.2021.100080

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Ghosh M, Gauger M, Marcu A, Nelde A, Denk M, Schuster H, et al. Guidance document: Validation of a high-performance liquid chromatography-tandem mass spectrometry immunopeptidomics assay for the identification of HLA class I ligands suitable for pharmaceutical therapies. Mol Cell Proteomics (2020) 19:432–43. doi: 10.1074/mcp.C119.001652

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Erhard F, Dölken L, Schilling B, Schlosser A. Identification of the cryptic HLA-I immunopeptidome. Cancer Immunol Res (2020) 8:1018–26. doi: 10.1158/2326-6066.Cir-19-0886

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Erhard F, Halenius A, Zimmermann C, L'Hernault A, Kowalewski DJ, Weekes MP, et al. Improved Ribo-seq enables identification of cryptic translation events. Nat Methods (2018) 15:363–6. doi: 10.1038/nmeth.4631

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Laumont CM, Vincent K, Hesnard L, Audemard É., Bonneil É., Laverdure JP, et al. Noncoding regions are the main source of targetable tumor-specific antigens. Sci Transl Med (2018) 10. doi: 10.1126/scitranslmed.aau5516

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Creech AL, Ting YS, Goulding SP, Sauld JFK, Barthelme D, Rooney MS, et al. The role of mass spectrometry and proteogenomics in the advancement of HLA epitope prediction. Proteomics (2018) 18:e1700259. doi: 10.1002/pmic.201700259

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Laumont CM, Perreault C. Exploiting non-canonical translation to identify new targets for T cell-based cancer immunotherapy. Cell Mol Life Sci (2018) 75:607–21. doi: 10.1007/s00018-017-2628-4

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Peters B, Nielsen M, Sette A. T cell epitope predictions. Annu Rev Immunol (2020) 38:123–45. doi: 10.1146/annurev-immunol-082119-124838

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Bassani-Sternberg M, Bräunlein E, Klar R, Engleitner T, Sinitcyn P, Audehm S, et al. Direct identification of clinically relevant neoepitopes presented on native human melanoma tissue by mass spectrometry. Nat Commun (2016) 7:13404. doi: 10.1038/ncomms13404

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Löffler MW, Mohr C, Bichmann L, Freudenmann LK, Walzer M, Schroeder CM, et al. Multi-omics discovery of exome-derived neoantigens in hepatocellular carcinoma. Genome Med (2019) 11:28. doi: 10.1186/s13073-019-0636-8

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Mauriello A, Zeuli R, Cavalluzzo B, Petrizzo A, Tornesello ML, Buonaguro FM, et al. High somatic mutation and neoantigen burden do not correlate with decreased progression-free survival in HCC patients not undergoing immunotherapy. Cancers (Basel) (2019) 11. doi: 10.3390/cancers11121824

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Petrizzo A, Tagliamonte M, Mauriello A, Costa V, Aprile M, Esposito R, et al. Unique true predicted neoantigens (TPNAs) correlates with anti-tumor immune control in HCC patients. J Transl Med (2018) 16:286. doi: 10.1186/s12967-018-1662-9

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Wells DK, van Buuren MM, Dang KK, Hubbard-Lucey VM, Sheehan KCF, Campbell KM, et al. Key parameters of tumor epitope immunogenicity revealed through a consortium approach improve neoantigen prediction. Cell (2020) 183:818–834 e813. doi: 10.1016/j.cell.2020.09.015

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Raoufi E, Hemmati M, Eftekhari S, Khaksaran K, Mahmodi Z, Farajollahi MM, et al. Epitope prediction by novel immunoinformatics approach: A state-of-the-art review. Int J Pept Res Ther (2020) 26:1155–63. doi: 10.1007/s10989-019-09918-z

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Atanasova M, Patronov A, Dimitrov I, Flower DR, Doytchinova I. EpiDOCK: a molecular docking-based tool for MHC class II binding prediction. Protein Eng Des Sel (2013) 26:631–4. doi: 10.1093/protein/gzt018

PubMed Abstract | CrossRef Full Text | Google Scholar

97. He Y, Xiang Z, Mobley HL. Vaxign: the first web-based vaccine design program for reverse vaccinology and applications for vaccine development. J BioMed Biotechnol (2010) 2010:297505. doi: 10.1155/2010/297505

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Molero-Abraham M, Lafuente EM, Flower DR, Reche PA. Selection of conserved epitopes from hepatitis C virus for pan-populational stimulation of T-cell responses. Clin Dev Immunol (2013) 2013:601943. doi: 10.1155/2013/601943

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Doytchinova IA, Guan P, Flower DR. EpiJen: a server for multistep T cell epitope prediction. BMC Bioinf (2006) 7:131. doi: 10.1186/1471-2105-7-131

CrossRef Full Text | Google Scholar

100. Singh H, Raghava GP. ProPred: prediction of HLA-DR binding sites. Bioinformatics (2001) 17:1236–7. doi: 10.1093/bioinformatics/17.12.1236

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Larsen MV, Lundegaard C, Lamberth K, Buus S, Brunak S, Lund O, et al. An integrative approach to CTL epitope prediction: a combined algorithm integrating MHC class I binding, TAP transport efficiency, and proteasomal cleavage predictions. Eur J Immunol (2005) 35:2295–303. doi: 10.1002/eji.200425811

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Zhang GL, Ansari HR, Bradley P, Cawley GC, Hertz T, Hu X, et al. Machine learning competition in immunology - Prediction of HLA class I binding peptides. J Immunol Methods (2011) 374:1–4. doi: 10.1016/j.jim.2011.09.010

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Dhanda SK, Gupta S, Vir P, Raghava GP. Prediction of IL4 inducing peptides. Clin Dev Immunol (2013) 2013:263952. doi: 10.1155/2013/263952

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Bhasin M, Raghava GP. SVM based method for predicting HLA-DRB1*0401 binding peptides in an antigen sequence. Bioinformatics (2004) 20:421–3. doi: 10.1093/bioinformatics/btg424

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Wang P, Sidney J, Dow C, Mothe B, Sette A, Peters B. A systematic assessment of MHC class II peptide binding predictions and evaluation of a consensus approach. PloS Comput Biol (2008) 4:e1000048. doi: 10.1371/journal.pcbi.1000048

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Reynisson B, Alvarez B, Paul S, Peters B, Nielsen M. NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by concurrent motif deconvolution and integration of MS MHC eluted ligand data. Nucleic Acids Res (2020) 48:W449–w454. doi: 10.1093/nar/gkaa379

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Sarkizova S, Klaeger S, Le PM, Li LW, Oliveira G, Keshishian H, et al. A large peptidome dataset improves HLA class I epitope prediction across most of the human population. Nat Biotechnol (2020) 38:199–209. doi: 10.1038/s41587-019-0322-9

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Shao XM, Bhattacharya R, Huang J, Sivakumar IKA, Tokheim C, Zheng L, et al. High-throughput prediction of MHC class I and II neoantigens with MHCnuggets. Cancer Immunol Res (2020) 8:396–408. doi: 10.1158/2326-6066.Cir-19-0464

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Gfeller D, Bassani-Sternberg M, Schmidt J, Luescher IF. Current tools for predicting cancer-specific T cell immunity. Oncoimmunology (2016) 5:e1177691. doi: 10.1080/2162402x.2016.1177691

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Juncker AS, Larsen MV, Weinhold N, Nielsen M, Brunak S, Lund O. Systematic characterisation of cellular localisation and expression profiles of proteins containing MHC ligands. PloS One (2009) 4:e7448. doi: 10.1371/journal.pone.0007448

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Hickman HD, Luis AD, Buchli R, Few SR, Sathiamurthy M, VanGundy RS, et al. Toward a definition of self: proteomic evaluation of the class I peptide repertoire. J Immunol (2004) 172:2944–52. doi: 10.4049/jimmunol.172.5.2944

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Milner E, Barnea E, Beer I, Admon A. The turnover kinetics of major histocompatibility complex peptides of human cancer cells. Mol Cell Proteomics (2006) 5:357–65. doi: 10.1074/mcp.M500241-MCP200

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Abelin JG, Keskin DB, Sarkizova S, Hartigan CR, Zhang W, Sidney J, et al. Mass spectrometry profiling of HLA-associated peptidomes in mono-allelic cells enables more accurate epitope prediction. Immunity (2017) 46:315–26. doi: 10.1016/j.immuni.2017.02.007

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Fortier MH, Caron E, Hardy MP, Voisin G, Lemieux S, Perreault C, et al. The MHC class I peptide repertoire is molded by the transcriptome. J Exp Med (2008) 205:595–610. doi: 10.1084/jem.20071985

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Bassani-Sternberg M, Pletscher-Frankild S, Jensen LJ, Mann M. Mass spectrometry of human leukocyte antigen class I peptidomes reveals strong effects of protein abundance and turnover on antigen presentation. Mol Cell Proteomics (2015) 14:658–73. doi: 10.1074/mcp.M114.042812

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Koşaloğlu-Yalçın Z, Lee J, Greenbaum J, Schoenberger SP, Miller A, Kim YJ, et al. Combined assessment of MHC binding and antigen abundance improves T cell epitope predictions. iScience (2022) 25:103850. doi: 10.1016/j.isci.2022.103850

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Toes RE, Blom RJ, Offringa R, Kast WM, Melief CJ. Enhanced tumor outgrowth after peptide vaccination. Functional deletion of tumor-specific CTL induced by peptide vaccination can lead to the inability to reject tumors. J Immunol (1996) 156:3911–8. doi: 10.4049/jimmunol.156.10.3911

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Tuettenberg A, Becker C, Huter E, Knop J, Enk AH, Jonuleit H. Induction of strong and persistent MelanA/MART-1-specific immune responses by adjuvant dendritic cell-based vaccination of stage II melanoma patients. Int J Cancer (2006) 118:2617–27. doi: 10.1002/ijc.21679

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Bonam SR, Partidos CD, Halmuthur SKM, Muller S. An overview of novel adjuvants designed for improving vaccine efficacy. Trends Pharmacol Sci (2017) 38:771–93. doi: 10.1016/j.tips.2017.06.002

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Dubensky TW Jr., Reed SG. Adjuvants for cancer vaccines. Semin Immunol (2010) 22:155–61. doi: 10.1016/j.smim.2010.04.007

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Temizoz B, Kuroda E, Ishii KJ. Combination and inducible adjuvants targeting nucleic acid sensors. Curr Opin Pharmacol (2018) 41:104–13. doi: 10.1016/j.coph.2018.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Hogenesch H. Mechanism of immunopotentiation and safety of aluminum adjuvants. Front Immunol (2012) 3:406. doi: 10.3389/fimmu.2012.00406

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Mosca F, Tritto E, Muzzi A, Monaci E, Bagnoli F, Iavarone C, et al. Molecular and cellular signatures of human vaccine adjuvants. Proc Natl Acad Sci U.S.A. (2008) 105:10501–6. doi: 10.1073/pnas.0804699105

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Cuzzubbo S, Mangsbo S, Nagarajan D, Habra K, Pockley AG, McArdle SEB. Cancer vaccines: Adjuvant potency, importance of age, lifestyle, and treatments. Front Immunol (2020) 11:615240. doi: 10.3389/fimmu.2020.615240

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Smith M, García-Martínez E, Pitter MR, Fucikova J, Spisek R, Zitvogel L, et al. Trial Watch: Toll-like receptor agonists in cancer immunotherapy. Oncoimmunology (2018) 7:e1526250. doi: 10.1080/2162402x.2018.1526250

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Gay NJ, Gangloff M. Structure and function of Toll receptors and their ligands. Annu Rev Biochem (2007) 76:141–65. doi: 10.1146/annurev.biochem.76.060305.151318

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immunol (2003) 21:335–76. doi: 10.1146/annurev.immunol.21.120601.141126

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Circelli L, Petrizzo A, Tagliamonte M, Heidenreich R, Tornesello ML, Buonaguro FM, et al. Immunological effects of a novel RNA-based adjuvant in liver cancer patients. Cancer Immunol Immunother (2017) 66:103–12. doi: 10.1007/s00262-016-1923-5

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Akira S, Takeda K, Kaisho T. Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol (2001) 2:675–80. doi: 10.1038/90609

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Van Hoeven N, Fox CB, Granger B, Evers T, Joshi SW, Nana GI, et al. A formulated TLR7/8 agonist is a flexible, highly potent and effective adjuvant for pandemic influenza vaccines. Sci Rep (2017) 7:46426. doi: 10.1038/srep46426

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Smith AJ, Li Y, Bazin HG, St-Jean JR, Larocque D, Evans JT, et al. Evaluation of novel synthetic TLR7/8 agonists as vaccine adjuvants. Vaccine (2016) 34:4304–12. doi: 10.1016/j.vaccine.2016.06.080

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Vollmer J, Krieg AM. Immunotherapeutic applications of CpG oligodeoxynucleotide TLR9 agonists. Adv Drug Delivery Rev (2009) 61:195–204. doi: 10.1016/j.addr.2008.12.008

CrossRef Full Text | Google Scholar

133. Bode C, Zhao G, Steinhagen F, Kinjo T, Klinman DM. CpG DNA as a vaccine adjuvant. Expert Rev Vaccines (2011) 10:499–511. doi: 10.1586/erv.10.174

PubMed Abstract | CrossRef Full Text | Google Scholar

134. Martins KA, Bavari S, Salazar AM. Vaccine adjuvant uses of poly-IC and derivatives. Expert Rev Vaccines (2015) 14:447–59. doi: 10.1586/14760584.2015.966085

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Kyi C, Roudko V, Sabado R, Saenger Y, Loging W, Mandeli J, et al. Therapeutic immune modulation against solid cancers with intratumoral poly-ICLC: A pilot trial. Clin Cancer Res (2018) 24:4937–48. doi: 10.1158/1078-0432.Ccr-17-1866

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Heidenreich R, Jasny E, Kowalczyk A, Lutz J, Probst J, Baumhof P, et al. A novel RNA-based adjuvant combines strong immunostimulatory capacities with a favorable safety profile. Int J Cancer (2015) 137:372–84. doi: 10.1002/ijc.29402

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Krutzik SR, Tan B, Li H, Ochoa MT, Liu PT, Sharfstein SE, et al. TLR activation triggers the rapid differentiation of monocytes into macrophages and dendritic cells. Nat Med (2005) 11:653–60. doi: 10.1038/nm1246

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Jackson S, Lentino J, Kopp J, Murray L, Ellison W, Rhee M, et al. Immunogenicity of a two-dose investigational hepatitis B vaccine, HBsAg-1018, using a toll-like receptor 9 agonist adjuvant compared with a licensed hepatitis B vaccine in adults. Vaccine (2018) 36:668–74. doi: 10.1016/j.vaccine.2017.12.038

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Makadia HK, Siegel SJ. Poly lactic-co-glycolic acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers (Basel) (2011) 3:1377–97. doi: 10.3390/polym3031377

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Stark B, Andreae F, Mosgoeller W, Edetsberger M, Gaubitzer E, Koehler G, et al. Liposomal vasoactive intestinal peptide for lung application: protection from proteolytic degradation. Eur J Pharm Biopharm (2008) 70:153–64. doi: 10.1016/j.ejpb.2008.04.015

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Arbelaez CA, Estrada J, Gessner MA, Glaus C, Morales AB, Mohn D, et al. A nanoparticle vaccine that targets neoantigen peptides to lymphoid tissues elicits robust antitumor T cell responses. NPJ Vaccines (2020) 5:106. doi: 10.1038/s41541-020-00253-9

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Chatin B, Mevel M, Devalliere J, Dallet L, Haudebourg T, Peuziat P, et al. Liposome-based formulation for intracellular delivery of functional proteins. Mol Ther Nucleic Acids (2015) 4:e244. doi: 10.1038/mtna.2015.17

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Varypataki EM, Silva AL, Barnier-Quer C, Collin N, Ossendorp F, Jiskoot W. Synthetic long peptide-based vaccine formulations for induction of cell mediated immunity: A comparative study of cationic liposomes and PLGA nanoparticles. J Control Release (2016) 226:98–106. doi: 10.1016/j.jconrel.2016.02.018

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Petrizzo A, Conte C, Tagliamonte M, Napolitano M, Bifulco K, Carriero V, et al. Functional characterization of biodegradable nanoparticles as antigen delivery system. J Exp Clin Cancer Res (2015) 34:114. doi: 10.1186/s13046-015-0231-9

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Biswas S, Torchilin VP. Nanopreparations for organelle-specific delivery in cancer. Adv Drug Delivery Rev (2014) 66:26–41. doi: 10.1016/j.addr.2013.11.004

CrossRef Full Text | Google Scholar

146. Koshy ST, Mooney DJ. Biomaterials for enhancing anti-cancer immunity. Curr Opin Biotechnol (2016) 40:1–8. doi: 10.1016/j.copbio.2016.02.001

PubMed Abstract | CrossRef Full Text | Google Scholar

147. Fujita Y, Taguchi H. Current status of multiple antigen-presenting peptide vaccine systems: Application of organic and inorganic nanoparticles. Chem Cent J (2011) 5:48. doi: 10.1186/1752-153X-5-48

PubMed Abstract | CrossRef Full Text | Google Scholar

148. Grippin AJ, Sayour EJ, Mitchell DA. Translational nanoparticle engineering for cancer vaccines. Oncoimmunology (2017) 6:e1290036. doi: 10.1080/2162402X.2017.1290036

PubMed Abstract | CrossRef Full Text | Google Scholar

149. Lopez-Campos F, Candini D, Carrasco E, Berenguer Frances MA. Nanoparticles applied to cancer immunoregulation. Rep Pract Oncol Radiother (2019) 24:47–55. doi: 10.1016/j.rpor.2018.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

150. Gao S, Yang D, Fang Y, Lin X, Jin X, Wang Q, et al. Engineering nanoparticles for targeted remodeling of the tumor microenvironment to improve cancer immunotherapy. Theranostics (2019) 9:126–51. doi: 10.7150/thno.29431

PubMed Abstract | CrossRef Full Text | Google Scholar

151. Butts C, Maksymiuk A, Goss G, Soulieres D, Marshall E, Cormier Y, et al. Updated survival analysis in patients with stage IIIB or IV non-small-cell lung cancer receiving BLP25 liposome vaccine (L-BLP25): phase IIB randomized, multicenter, open-label trial. J Cancer Res Clin Oncol (2011) 137:1337–42. doi: 10.1007/s00432-011-1003-3

PubMed Abstract | CrossRef Full Text | Google Scholar

152. Butts C, Murray RN, Smith CJ, Ellis PM, Jasas K, Maksymiuk A, et al. A multicenter open-label study to assess the safety of a new formulation of BLP25 liposome vaccine in patients with unresectable stage III non-small-cell lung cancer. Clin Lung Cancer (2010) 11:391–5. doi: 10.3816/CLC.2010.n.101

PubMed Abstract | CrossRef Full Text | Google Scholar

153. McQuade JL, Homsi J, Torres-Cabala CA, Bassett R, Popuri RM, James ML, et al. A phase II trial of recombinant MAGE-A3 protein with immunostimulant AS15 in combination with high-dose Interleukin-2 (HDIL2) induction therapy in metastatic melanoma. BMC Cancer (2018) 18:1274. doi: 10.1186/s12885-018-5193-9

PubMed Abstract | CrossRef Full Text | Google Scholar

154. Karkada M, Berinstein NL, Mansour M. Therapeutic vaccines and cancer: focus on DPX-0907. Biologics (2014) 8:27–38. doi: 10.2147/BTT.S55196

PubMed Abstract | CrossRef Full Text | Google Scholar

155. Klein O, Davis ID, McArthur GA, Chen L, Haydon A, Parente P, et al. Low-dose cyclophosphamide enhances antigen-specific CD4(+) T cell responses to NY-ESO-1/ISCOMATRIX vaccine in patients with advanced melanoma. Cancer Immunol Immunother (2015) 64:507–18. doi: 10.1007/s00262-015-1656-x

PubMed Abstract | CrossRef Full Text | Google Scholar

156. Kitano S, Kageyama S, Nagata Y, Miyahara Y, Hiasa A, Naota H, et al. HER2-specific T-cell immune responses in patients vaccinated with truncated HER2 protein complexed with nanogels of cholesteryl pullulan. Clin Cancer Res (2006) 12:7397–405. doi: 10.1158/1078-0432.CCR-06-1546

PubMed Abstract | CrossRef Full Text | Google Scholar

157. Harari A, Graciotti M, Bassani-Sternberg M, Kandalaft LE. Antitumour dendritic cell vaccination in a priming and boosting approach. Nat Rev Drug Discovery (2020) 19:635–52. doi: 10.1038/s41573-020-0074-8

CrossRef Full Text | Google Scholar

158. Zitvogel L, Regnault A, Lozier A, Wolfers J, Flament C, Tenza D, et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med (1998) 4:594–600. doi: 10.1038/nm0598-594

PubMed Abstract | CrossRef Full Text | Google Scholar

159. Seo N, Akiyoshi K, Shiku H. Exosome-mediated regulation of tumor immunology. Cancer Sci (2018) 109:2998–3004. doi: 10.1111/cas.13735

PubMed Abstract | CrossRef Full Text | Google Scholar

160. Munich S, Sobo-Vujanovic A, Buchser WJ, Beer-Stolz D, Vujanovic NL. Dendritic cell exosomes directly kill tumor cells and activate natural killer cells via TNF superfamily ligands. Oncoimmunology (2012) 1:1074–83. doi: 10.4161/onci.20897

PubMed Abstract | CrossRef Full Text | Google Scholar

161. Pitt JM, Andre F, Amigorena S, Soria JC, Eggermont A, Kroemer G, et al. Dendritic cell-derived exosomes for cancer therapy. J Clin Invest (2016) 126:1224–32. doi: 10.1172/JCI81137

PubMed Abstract | CrossRef Full Text | Google Scholar

162. Andre F, Chaput N, Schartz NE, Flament C, Aubert N, Bernard J, et al. Exosomes as potent cell-free peptide-based vaccine. I. Dendritic cell-derived exosomes transfer functional MHC class I/peptide complexes to dendritic cells. J Immunol (2004) 172:2126–36. doi: 10.4049/jimmunol.172.4.2126

PubMed Abstract | CrossRef Full Text | Google Scholar

163. Viaud S, Thery C, Ploix S, Tursz T, Lapierre V, Lantz O, et al. Dendritic cell-derived exosomes for cancer immunotherapy: what's next? Cancer Res (2010) 70:1281–5. doi: 10.1158/0008-5472.CAN-09-3276

PubMed Abstract | CrossRef Full Text | Google Scholar

164. Wolfers J, Lozier A, Raposo G, Regnault A, Thery C, Masurier C, et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat Med (2001) 7:297–303. doi: 10.1038/85438

PubMed Abstract | CrossRef Full Text | Google Scholar

165. Stuge TB, Holmes SP, Saharan S, Tuettenberg A, Roederer M, Weber JS, et al. Diversity and recognition efficiency of T cell responses to cancer. PloS Med (2004) 1:e28. doi: 10.1371/journal.pmed.0010028

PubMed Abstract | CrossRef Full Text | Google Scholar

166. Boon T, Van Pel A. T cell-recognized antigenic peptides derived from the cellular genome are not protein degradation products but can be generated directly by transcription and translation of short subgenic regions. A hypothesis Immunogenetics (1989) 29:75–9. doi: 10.1007/bf00395854

CrossRef Full Text | Google Scholar

167. Fikes JD, Sette A. Design of multi-epitope, analogue-based cancer vaccines. Expert Opin Biol Ther (2003) 3:985–93. doi: 10.1517/14712598.3.6.985

PubMed Abstract | CrossRef Full Text | Google Scholar

168. Kanaseki T, Torigoe T. Proteogenomics: advances in cancer antigen research. Immunol Med (2019) 42:65–70. doi: 10.1080/25785826.2019.1640500

PubMed Abstract | CrossRef Full Text | Google Scholar

169. Carrabba MG, Castelli C, Maeurer MJ, Squarcina P, Cova A, Pilla L, et al. Suboptimal activation of CD8(+) T cells by melanoma-derived altered peptide ligands: role of Melan-A/MART-1 optimized analogues. Cancer Res (2003) 63:1560–7.

PubMed Abstract | Google Scholar

170. Parkhurst MR, Salgaller ML, Southwood S, Robbins PF, Sette A, Rosenberg SA, et al. Improved induction of melanoma-reactive CTL with peptides from the melanoma antigen gp100 modified at HLA-A*0201-binding residues. J Immunol (1996) 157:2539–48. doi: 10.4049/jimmunol.157.6.2539

PubMed Abstract | CrossRef Full Text | Google Scholar

171. Madura F, Rizkallah PJ, Holland CJ, Fuller A, Bulek A, Godkin AJ, et al. Structural basis for ineffective T-cell responses to MHC anchor residue-improved "heteroclitic" peptides. Eur J Immunol (2015) 45:584–91. doi: 10.1002/eji.201445114

PubMed Abstract | CrossRef Full Text | Google Scholar

172. Cavalluzzo B, Ragone C, Mauriello A, Petrizzo A, Manolio C, Caporale A, et al. Identification and characterization of heteroclitic peptides in TCR-binding positions with improved HLA-binding efficacy. J Transl Med (2021) 19:89. doi: 10.1186/s12967-021-02757-x

PubMed Abstract | CrossRef Full Text | Google Scholar

173. Tagliamonte M, Mauriello A, Cavalluzzo B, Ragone C, Manolio C, Luciano A, et al. MHC-optimized peptide scaffold for improved antigen presentation and anti-tumor response. Front Immunol (2021) 12:769799. doi: 10.3389/fimmu.2021.769799

PubMed Abstract | CrossRef Full Text | Google Scholar

174. Wooldridge L, Ekeruche-Makinde J, van den Berg HA, Skowera A, Miles JJ, Tan MP, et al. A single autoimmune T cell receptor recognizes more than a million different peptides. J Biol Chem (2012) 287:1168–77. doi: 10.1074/jbc.M111.289488

PubMed Abstract | CrossRef Full Text | Google Scholar

175. Sewell AK. Why must T cells be cross-reactive? Nat Rev Immunol (2012) 12:669–77. doi: 10.1038/nri3279

PubMed Abstract | CrossRef Full Text | Google Scholar

176. Ragone C, Manolio C, Cavalluzzo B, Mauriello A, Tornesello ML, Buonaguro FM, et al. Identification and validation of viral antigens sharing sequence and structural homology with tumor-associated antigens (TAAs). J Immunother Cancer (2021) 9. doi: 10.1136/jitc-2021-002694

PubMed Abstract | CrossRef Full Text | Google Scholar

177. Vujanovic L, Shi J, Kirkwood JM, Storkus WJ, Butterfield LH. Molecular mimicry of MAGE-A6 and Mycoplasma penetrans HF-2 epitopes in the induction of antitumor CD8(+) T-cell responses. Oncoimmunology (2014) 3:e954501. doi: 10.4161/21624011.2014.954501

PubMed Abstract | CrossRef Full Text | Google Scholar

178. Loftus DJ, Castelli C, Clay TM, Squarcina P, Marincola FM, Nishimura MI, et al. Identification of epitope mimics recognized by CTL reactive to the melanoma/melanocyte-derived peptide MART-1(27-35). J Exp Med (1996) 184:647–57. doi: 10.1084/jem.184.2.647

PubMed Abstract | CrossRef Full Text | Google Scholar

179. Chiaro J, Kasanen HHE, Whalley T, Capasso C, Grönholm M, Feola S, et al. Viral molecular mimicry influences the antitumor immune response in murine and human melanoma. Cancer Immunol Res (2021) 9:981–93. doi: 10.1158/2326-6066.Cir-20-0814

PubMed Abstract | CrossRef Full Text | Google Scholar

180. Tagliamonte M, Cavalluzzo B, Mauriello A, Ragone C, Buonaguro FM, Tornesello ML, et al. Molecular mimicry and cancer vaccine development. Mol Cancer (2023) 22:75. doi: 10.1186/s12943-023-01776-0

PubMed Abstract | CrossRef Full Text | Google Scholar

181. Buonaguro L, Cavalluzzo B, Mauriello A, Ragone C, Tornesello AL, Buonaguro FM, et al. Microorganisms-derived antigens for preventive anti-cancer vaccines. Mol Aspects Med (2023) 92:101192. doi: 10.1016/j.mam.2023.101192

PubMed Abstract | CrossRef Full Text | Google Scholar

182. Ragone C, Manolio C, Mauriello A, Cavalluzzo B, Buonaguro FM, Tornesello ML, et al. Molecular mimicry between tumor associated antigens and microbiota-derived epitopes. J Transl Med (2022) 20:316. doi: 10.1186/s12967-022-03512-6

PubMed Abstract | CrossRef Full Text | Google Scholar

183. Buonaguro L, Cerullo V. Pathogens: Our allies against cancer? Mol Ther (2021) 29:10–2. doi: 10.1016/j.ymthe.2020.12.005

PubMed Abstract | CrossRef Full Text | Google Scholar

184. van der Burg SH, Arens R, Ossendorp F, van Hall T, Melief CJ. Vaccines for established cancer: overcoming the challenges posed by immune evasion. Nat Rev Cancer (2016) 16:219–33. doi: 10.1038/nrc.2016.16

PubMed Abstract | CrossRef Full Text | Google Scholar

185. Saxena M, van der Burg SH, Melief CJM, Bhardwaj N. Therapeutic cancer vaccines. Nat Rev Cancer (2021) 21:360–78. doi: 10.1038/s41568-021-00346-0

PubMed Abstract | CrossRef Full Text | Google Scholar

186. Tagliamonte M, Petrizzo A, Mauriello A, Tornesello ML, Buonaguro FM, Buonaguro L. Potentiating cancer vaccine efficacy in liver cancer. Oncoimmunology (2018) 7:e1488564. doi: 10.1080/2162402X.2018.1488564

PubMed Abstract | CrossRef Full Text | Google Scholar

187. Chakraborty M, Abrams SI, Coleman CN, Camphausen K, Schlom J, Hodge JW. External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing. Cancer Res (2004) 64:4328–37. doi: 10.1158/0008-5472.Can-04-0073

PubMed Abstract | CrossRef Full Text | Google Scholar

188. Gameiro SR, Malamas AS, Bernstein MB, Tsang KY, Vassantachart A, Sahoo N, et al. Tumor cells surviving exposure to proton or photon radiation share a common immunogenic modulation signature, rendering them more sensitive to T cell-mediated killing. Int J Radiat Oncol Biol Phys (2016) 95:120–30. doi: 10.1016/j.ijrobp.2016.02.022

PubMed Abstract | CrossRef Full Text | Google Scholar

189. Hodge JW, Garnett CT, Farsaci B, Palena C, Tsang KY, Ferrone S, et al. Chemotherapy-induced immunogenic modulation of tumor cells enhances killing by cytotoxic T lymphocytes and is distinct from immunogenic cell death. Int J Cancer (2013) 133:624–36. doi: 10.1002/ijc.28070

PubMed Abstract | CrossRef Full Text | Google Scholar

190. Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell (2015) 28:690–714. doi: 10.1016/j.ccell.2015.10.012

PubMed Abstract | CrossRef Full Text | Google Scholar

191. Ghiringhelli F, Menard C, Puig PE, Ladoire S, Roux S, Martin F, et al. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol Immunother (2007) 56:641–8. doi: 10.1007/s00262-006-0225-8

PubMed Abstract | CrossRef Full Text | Google Scholar

192. Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat Med (2009) 15:1170–8. doi: 10.1038/nm.2028

PubMed Abstract | CrossRef Full Text | Google Scholar

193. Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med (2007) 13:1050–9. doi: 10.1038/nm1622

PubMed Abstract | CrossRef Full Text | Google Scholar

194. Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res (2005) 11:6713–21. doi: 10.1158/1078-0432.Ccr-05-0883

PubMed Abstract | CrossRef Full Text | Google Scholar

195. Garnett CT, Schlom J, Hodge JW. Combination of docetaxel and recombinant vaccine enhances T-cell responses and antitumor activity: effects of docetaxel on immune enhancement. Clin Cancer Res (2008) 14:3536–44. doi: 10.1158/1078-0432.Ccr-07-4025

PubMed Abstract | CrossRef Full Text | Google Scholar

196. Tagliamonte M, Petrizzo A, Napolitano M, Luciano A, Rea D, Barbieri A, et al. A novel multi-drug metronomic chemotherapy significantly delays tumor growth in mice. J Transl Med (2016) 14:58. doi: 10.1186/s12967-016-0812-1

PubMed Abstract | CrossRef Full Text | Google Scholar

197. Tagliamonte M, Petrizzo A, Napolitano M, Luciano A, Arra C, Maiolino P, et al. Novel metronomic chemotherapy and cancer vaccine combinatorial strategy for hepatocellular carcinoma in a mouse model. Cancer Immunol Immunother (2015) 64:1305–14. doi: 10.1007/s00262-015-1698-0

PubMed Abstract | CrossRef Full Text | Google Scholar

198. Madondo MT, Quinn M, Plebanski M. Low dose cyclophosphamide: Mechanisms of T cell modulation. Cancer Treat Rev (2016) 42:3–9. doi: 10.1016/j.ctrv.2015.11.005

PubMed Abstract | CrossRef Full Text | Google Scholar

199. Rampling R, Peoples S, Mulholland PJ, James A, Al-Salihi O, Twelves CJ, et al. A cancer research UK first time in human phase I trial of IMA950 (Novel multipeptide therapeutic vaccine) in patients with newly diagnosed glioblastoma. Clin Cancer Res (2016) 22:4776–85. doi: 10.1158/1078-0432.Ccr-16-0506

PubMed Abstract | CrossRef Full Text | Google Scholar

200. Demaria S, Golden EB, Formenti SC. Role of local radiation therapy in cancer immunotherapy. JAMA Oncol (2015) 1:1325–32. doi: 10.1001/jamaoncol.2015.2756

PubMed Abstract | CrossRef Full Text | Google Scholar

201. Dillon MT, Bergerhoff KF, Pedersen M, Whittock H, Crespo-Rodriguez E, Patin EC, et al. ATR inhibition potentiates the radiation-induced inflammatory tumor microenvironment. Clin Cancer Res (2019) 25:3392–403. doi: 10.1158/1078-0432.Ccr-18-1821

PubMed Abstract | CrossRef Full Text | Google Scholar

202. Weichselbaum RR, Liang H, Deng L, Fu YX. Radiotherapy and immunotherapy: a beneficial liaison? Nat Rev Clin Oncol (2017) 14:365–79. doi: 10.1038/nrclinonc.2016.211

PubMed Abstract | CrossRef Full Text | Google Scholar

203. Stamell EF, Wolchok JD, Gnjatic S, Lee NY, Brownell I. The abscopal effect associated with a systemic anti-melanoma immune response. Int J Radiat Oncol Biol Phys (2013) 85:293–5. doi: 10.1016/j.ijrobp.2012.03.017

PubMed Abstract | CrossRef Full Text | Google Scholar

204. Golden EB, Demaria S, Schiff PB, Chachoua A, Formenti SC. An abscopal response to radiation and ipilimumab in a patient with metastatic non-small cell lung cancer. Cancer Immunol Res (2013) 1:365–72. doi: 10.1158/2326-6066.Cir-13-0115

PubMed Abstract | CrossRef Full Text | Google Scholar

205. Chandra RA, Wilhite TJ, Balboni TA, Alexander BM, Spektor A, Ott PA, et al. A systematic evaluation of abscopal responses following radiotherapy in patients with metastatic melanoma treated with ipilimumab. Oncoimmunology (2015) 4:e1046028. doi: 10.1080/2162402x.2015.1046028

PubMed Abstract | CrossRef Full Text | Google Scholar

206. Kulzer L, Rubner Y, Deloch L, Allgäuer A, Frey B, Fietkau R, et al. Norm- and hypo-fractionated radiotherapy is capable of activating human dendritic cells. J Immunotoxicol (2014) 11:328–36. doi: 10.3109/1547691x.2014.880533

PubMed Abstract | CrossRef Full Text | Google Scholar

207. Mondini M, Nizard M, Tran T, Mauge L, Loi M, Clémenson C, et al. Synergy of radiotherapy and a cancer vaccine for the treatment of HPV-associated head and neck cancer. Mol Cancer Ther (2015) 14:1336–45. doi: 10.1158/1535-7163.Mct-14-1015

PubMed Abstract | CrossRef Full Text | Google Scholar

208. Chang MC, Chen YL, Lin HW, Chiang YC, Chang CF, Hsieh SF, et al. Irradiation enhances abscopal anti-tumor effects of antigen-specific immunotherapy through regulating tumor microenvironment. Mol Ther (2018) 26:404–19. doi: 10.1016/j.ymthe.2017.11.011

PubMed Abstract | CrossRef Full Text | Google Scholar

209. Sharma P, Goswami S, Raychaudhuri D, Siddiqui BA, Singh P, Nagarajan A, et al. Immune checkpoint therapy-current perspectives and future directions. Cell (2023) 186:1652–69. doi: 10.1016/j.cell.2023.03.006

PubMed Abstract | CrossRef Full Text | Google Scholar

210. Nasti TH, Eberhardt CS. Vaccination against Cancer or Infectious Agents during Checkpoint Inhibitor Therapy. Vaccines (Basel) (2021) 9. doi: 10.3390/vaccines9121396

PubMed Abstract | CrossRef Full Text | Google Scholar

211. Aichele P, Brduscha-Riem K, Zinkernagel RM, Hengartner H, Pircher H. T cell priming versus T cell tolerance induced by synthetic peptides. J Exp Med (1995) 182:261–6. doi: 10.1084/jem.182.1.261

PubMed Abstract | CrossRef Full Text | Google Scholar

212. Puzanov I, Milhem MM, Minor D, Hamid O, Li A, Chen L, et al. Talimogene laherparepvec in combination with ipilimumab in previously untreated, unresectable stage IIIB-IV melanoma. J Clin Oncol (2016) 34:2619–26. doi: 10.1200/JCO.2016.67.1529

PubMed Abstract | CrossRef Full Text | Google Scholar

213. Scholz M, Yep S, Chancey M, Kelly C, Chau K, Turner J, et al. Phase I clinical trial of sipuleucel-T combined with escalating doses of ipilimumab in progressive metastatic castrate-resistant prostate cancer. Immunotargets Ther (2017) 6:11–6. doi: 10.2147/ITT.S122497

PubMed Abstract | CrossRef Full Text | Google Scholar

214. Brandacher G, Perathoner A, Ladurner R, Schneeberger S, Obrist P, Winkler C, et al. Prognostic value of indoleamine 2,3-dioxygenase expression in colorectal cancer: effect on tumor-infiltrating T cells. Clin Cancer Res (2006) 12:1144–51. doi: 10.1158/1078-0432.CCR-05-1966

PubMed Abstract | CrossRef Full Text | Google Scholar

215. Ino K, Yamamoto E, Shibata K, Kajiyama H, Yoshida N, Terauchi M, et al. Inverse correlation between tumoral indoleamine 2,3-dioxygenase expression and tumor-infiltrating lymphocytes in endometrial cancer: its association with disease progression and survival. Clin Cancer Res (2008) 14:2310–7. doi: 10.1158/1078-0432.CCR-07-4144

PubMed Abstract | CrossRef Full Text | Google Scholar

216. Nakamura T, Shima T, Saeki A, Hidaka T, Nakashima A, Takikawa O, et al. Expression of indoleamine 2, 3-dioxygenase and the recruitment of Foxp3-expressing regulatory T cells in the development and progression of uterine cervical cancer. Cancer Sci (2007) 98:874–81. doi: 10.1111/j.1349-7006.2007.00470.x

PubMed Abstract | CrossRef Full Text | Google Scholar

217. Meireson A, Ferdinande L, Haspeslagh M, Hennart B, Allorge D, Ost P, et al. Clinical relevance of serum kyn/trp ratio and basal and IFNgamma-upregulated IDO1 expression in peripheral monocytes in early stage melanoma. Front Immunol (2021) 12:736498. doi: 10.3389/fimmu.2021.736498

PubMed Abstract | CrossRef Full Text | Google Scholar

218. Bjoern J, Iversen TZ, Nitschke NJ, Andersen MH, Svane IM. Safety, immune and clinical responses in metastatic melanoma patients vaccinated with a long peptide derived from indoleamine 2,3-dioxygenase in combination with ipilimumab. Cytotherapy (2016) 18:1043–55. doi: 10.1016/j.jcyt.2016.05.010

PubMed Abstract | CrossRef Full Text | Google Scholar

219. Kjeldsen JW, Lorentzen CL, Martinenaite E, Ellebaek E, Donia M, Holmstroem RB, et al. A phase 1/2 trial of an immune-modulatory vaccine against IDO/PD-L1 in combination with nivolumab in metastatic melanoma. Nat Med (2021) 27:2212–23. doi: 10.1038/s41591-021-01544-x

PubMed Abstract | CrossRef Full Text | Google Scholar

220. DhatChinamoorthy K, Colbert JD, Rock KL. Cancer immune evasion through loss of MHC class I antigen presentation. Front Immunol (2021) 12:636568. doi: 10.3389/fimmu.2021.636568

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: peptides, TAA, TME, molecular mimicry, immunopeptidome, combinatorial strategies, adjuvant, MHC

Citation: Buonaguro L and Tagliamonte M (2023) Peptide-based vaccine for cancer therapies. Front. Immunol. 14:1210044. doi: 10.3389/fimmu.2023.1210044

Received: 21 April 2023; Accepted: 31 July 2023;
Published: 16 August 2023.

Edited by:

Cornelis Joseph Melief, Leiden University, Netherlands

Reviewed by:

Kwong Tsang, Precision Biologics, Inc., United States
Judith M. Ramage, University of Nottingham, United Kingdom

Copyright © 2023 Buonaguro and Tagliamonte. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Maria Tagliamonte, m.tagliamonte@istitutotumori.na.it

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.