Next Article in Journal
Glycine soja Leaf and Stem Extract Ameliorates Atopic Dermatitis-like Skin Inflammation by Inhibiting JAK/STAT Signaling
Previous Article in Journal
NetLnc: A Network-Based Computational Framework to Identify Immune Checkpoint-Related lncRNAs for Immunotherapy Response in Melanoma
Previous Article in Special Issue
Expression of the RPSA-Containing and 67EBP Laminin Receptors in Relation to the Debatable Nature of the 67 kDa Laminin Receptor 67LR in Colorectal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Signature Gene Mutations in Colorectal Cancer: Potential Neoantigens for Cancer Vaccines

1
Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea
2
Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(10), 4559; https://doi.org/10.3390/ijms26104559
Submission received: 8 April 2025 / Revised: 7 May 2025 / Accepted: 8 May 2025 / Published: 9 May 2025

Abstract

:
Colorectal cancer (CRC), the third most common cancer worldwide, is one of the deadliest cancers. CRC is known as a cold tumor, characterized by a low immune response that makes it difficult for immune cells to infiltrate and exhibits strong resistance to immunotherapy with checkpoint inhibition. This restricted response is largely attributed to signature gene mutations including mismatch repair (MMR) genes, KRAS, BRAF, APC, and TP53, which are also the main oncogenes in CRC. Mutated signature genes continuously upregulate abnormal signaling pathways, leading to excessive proliferation, cancer progression, and metastasis. Furthermore, it reorganizes the tumor microenvironment (TME) by recruiting immunosuppressive cells. However, the mutation can produce neoantigens that can provoke an immune response, making it a potential target for immunotherapy. In particular, cancer vaccines that leverage the strong neoantigenic properties of these mutations are considered promising for overcoming immune resistance and eliciting anti-tumor responses. In this review, we will describe signature gene mutations in CRC and focus on cancer vaccines targeting these mutations as potential therapies for CRC.

Graphical Abstract

1. Introduction

Colorectal cancer (CRC) is one of the most deadly carcinomas, with 1.9 million incidence cases and 0.9 million deaths worldwide in 2020 [1]. Colorectal cancer can be classified based on the degree of microsatellite instability into three categories: microsatellite stable (MSS), microsatellite instability-high (MSI-H), and microsatellite instability-low (MSI-L) [2]. MSI-H is frequently caused by the loss of function of the DNA mismatch repair (MMR) genes, and the B-Raf proto-oncogene, serine/threonine kinase (BRAF) V600E gene mutation is also frequently observed [2]. While MSS CRC generally has normal DNA mismatch repair gene function, genetic mutations such as adenomatous polyposis coli (APC), Kirsten rat sarcoma viral oncogene homolog (KRAS), and tumor protein p53 (TP53) are commonly observed [3]. The genetic heterogeneity of CRC, driven by the accumulation of key gene mutations, regulates essential signaling pathways that play distinct roles in tumorigenesis [4,5]. Mismatch repair deficiency (MMR-D or dMMR) is a condition in which dysfunction in at least one of the four MMR genes impairs the recognition and repair of DNA mismatches during replication. This leads to increased mutagenesis in DNA and causes microsatellite instability (MSI) [6]. Mutations in the KRAS gene, present in approximately 40–50% of all CRC cases, lead to excessive activation of the RAS/RAF/MEK/ERK signaling pathway, promoting cell proliferation and inhibiting apoptosis [7]. Similarly, BRAF mutations also contributed to cell proliferation, differentiation, and survival through abnormal activation of the mitogen-activated protein kinase (MAPK) signaling pathway [8]. Somatic mutations in APC gene, a key regulator of the Wnt/β-catenin pathway, occur in approximately 70–80% of CRC cases and contribute to tumor progression [9,10]. Additionally, p53 mutations are observed in 43% of CRC cases, leading to the loss of wild-type p53 function and promoting tumor growth and metastasis [11]. Collectively, genetic mutations are associated with the formation of an immunosuppressive tumor microenvironment (TME), making them crucial targets for cancer therapy [12]. In recent years, CRC has been treated using various methods including immune checkpoint inhibitors (ICIs). However, CRC exhibited the characteristics of a cold tumor due to its immunosuppressive TME, limiting its ability to induce an effective immune response in patients [2,13].
TME consists of the surrounding environment of the tumor, including immune cells such as T and B lymphocytes, mesenchymal stromal cells, the extracellular matrix (ECM), and secreted molecules like growth factors, cytokines, and chemokines [14]. TME interacts with tumor cells to shape tumor heterogeneity. Depending on the activity of immune cells within the TME, tumor immunophenotypes are classified into two types: hot and cold [15,16]. A hot tumor, also known as an immune-inflamed tumor, exhibits a high immune response due to the presence of tumor-infiltrating lymphocytes (TILs). Conversely, a cold tumor, also referred to as an immune-excluded tumor, has few T lymphocytes, resulting in a low immune response [17]. In cold tumors, immunotherapy is less effective, necessitating the development of effective strategies to overcome the immunosuppressive [18]. Previous studies have shown that inhibiting dysregulated signaling pathways can relieve an immunosuppressive environment [19]. Therefore, new strategies targeting these mutations are needed to enhance the efficacy of CRC treatment.
Cancer vaccines utilize an ideal antigen that is highly immunogenic and exclusively expressed in cancer cells for immunotherapy [20]. Cancer vaccines employ neoantigens derived from tumor-associated antigens (TAAs) or tumor-specific antigens (TSAs) to induce robust activation of immune cells, including T lymphocytes, and to establish immunological memory by presenting antigens to antigen-presenting cells (APCs) [21,22,23]. In CRC, signature gene mutations have the potential to serve as neoantigens due to their tumor specificity and are emerging as promising targets for cancer vaccines [24,25]. Currently, in various clinical trials, cancer vaccines are improving patient prognosis while minimizing lesion development through the effective induction of an immune response [26]. However, cancer vaccines still face limitations in immunotherapy due to factors such as the heterogeneity of the TME, the presence of immunosuppressive cells, weak immunogenicity, and variations in patients’ immune systems depending on their human leukocyte antigen (HLA) type [27,28]. To overcome these limitations and improve the therapeutic efficacy of cancer vaccines, research targeting signature gene mutations is necessary and essential for further advancement.
This review aims to discuss the potential of signature gene mutations in CRC as promising targets for neoantigen-based cancer vaccines. Moreover, by presenting some cases of signature gene mutations targeted at cancer vaccines, we highlight the future prospects of cancer vaccine development.

2. Signaling Pathways and Characteristics of Signature Gene Mutations in CRC

The signature gene mutations (MMR, KRAS, BRAF, APC, and TP53) in CRC are well known for playing significant roles in proliferation, invasion, migration, and metastasis [29]. The signaling pathways mediated by signature gene mutations are intricately interconnected, promoting CRC progression [30]. However, from an immunotherapeutic perspective, the mutated sequences of specific genes can serve as promising targets for cancer vaccines by providing neoantigens for effective anti-cancer strategies [31]. In this section, we highlight the signaling cascades of signature genes in CRC (Figure 1).

2.1. MMR in CRC

The MMR system is highly conserved and functions to excise base-base mismatch, thereby correcting errors that occur during DNA replication [32,33]. Its role is closely linked to the DNA damage response, which activates cell cycle arrest and apoptosis to prevent tumorigenesis caused by DNA mismatches [33]. In other words, MMR genes are crucial for maintaining the fidelity of DNA replication, so the inactivation of MMR genes inevitably leads to the spontaneous acquisition of a mutated phenotype in normal cells [33]. Mutations in MMR genes cause frequent somatic mutations, particularly in microsatellite regions, leading to MSI, which is associated with a high tumor mutation burden (TMB). In contrast, MSS exhibits the opposite characteristics [6]. MSI is associated with mutations in several MMR genes, including mutL homolog1 (MLH1), must homolog2 (MSH2), must homolog6 (MSH6), postmeiotic segregation increased2 (PMS2), or in the epithelial cell adhesion molecule (EPCAM) [32,34]. MSI CRC is characterized by hypermethylation of the MLH1 promoter, leading to its inactivation, and is frequently accompanied by the BRAF V600E mutation [35]. While MMR and MSI are not synonymous, their close interconnection carries important diagnostic and clinical implications. MMR gene analysis can be used as a diagnostic approach for evaluating MSI [36]. Unlike the MSS CRC, which exhibits low levels of TILs, MSI-H CRC is highly infiltrated with CD8+ and CD4+ T cells. Therefore, immune checkpoint inhibitors including nivolumab and ipilimumab can be used to treat MSI CRC and have shown effective responses [37]. Although they exhibit positive responses to ICIs, immune evasion mechanisms—such as the upregulation of Wnt/β-catenin signaling, Foxp3⁺ regulatory T cells (Tregs), and immune checkpoint molecules—can attenuate or interfere with their therapeutic efficacy [38]. Recently, to enhance the efficacy of ICIs for MSI-H CRC, combination therapy with cancer vaccines targeting shared neoantigens has been shown to be safe and to exhibit strong anti-tumor effects [34].

2.2. KRAS in CRC

Mutations in the RAS family of genes are frequently found in various malignancies, including CRC. RAS proteins act as key regulators of tumor cell proliferation, migration, and differentiation [39]. The RAS gene encodes three homologous RAS proteins: HRAS, NRAS, and KRAS [40]. In CRC, KRAS mutations are detected in more than 40% of cases [41]. These mutations, which commonly occur at codons 12, 13, and 61, lead to amino acid substitutions that result in structurally altered KRAS proteins, with codon 12 mutations being the most prevalent [42]. G12D (glycine 12 to aspartic acid) and G12V (glycine 12 to valine) are the most common subtypes of KRAS mutations in CRC, due to their low GTP hydrolysis rate compared to other mutations. In particular, G12D mutations occur in approximately 50% of cases [40,43]. KRAS mutations are influenced by the protein’s ability to hydrolyze GTP, and when nucleotide substitution is enhanced, downstream signaling pathways become activated [7]. Normally, KRAS toggles between ON and OFF states through its intrinsic GTPase activity. When GDP is exchanged for GTP, KRAS switches to the ON state and activates the MAPK/PI3K pathway [41]. The MAPK pathway involves sequential phosphorylation of downstream kinases—RAF, MEK, and ERK—which activate transcription factors, leading to cellular proliferation, migration and survival [44]. Mutant KRAS proteins with defective GTPase activity remain locked in the “ON” state, persistently activating downstream signaling and driving uncontrolled cell proliferation, a hallmark of CRC [42,45]. Also, mutations in the KRAS gene enhance cancer cell adaptation to metabolic stress. Mutant KRAS allows cancer cells to survive and proliferate under glucose-deprived conditions, unlike normal cells or KRAS wild-type cancer cells [46].
KRAS has long been regarded as an undruggable target due to its structural and biochemical properties. However, recent studies have shown that various immunotherapeutic approaches exhibit promising efficacy in targeting KRAS-driven cancers [47]. Notably, it has been suggested that combining immunotherapy with allele-specific KRAS inhibitors can improve anti-tumor efficacy in KRAS mutant cancers [48]. Other promising immunotherapy strategies, including adoptive T cell transfer targeting mutant KRAS and KRAS-targeted mRNA vaccines, are designed to stimulate tumor-specific T cell responses [49]. The structural changes caused by these mutations lead to the formation of unique mutant peptides not present in normal tissues, making them an attractive therapeutic target [50]. These mutant peptides can be presented on the surface of tumor cells by major histocompatibility complex (MHC) molecules, allowing their recognition by T cells as neoantigens [51]. Unlike TAAs, which may also be expressed in normal tissues, KRAS-derived neoantigens are tumor-specific, reducing the risk of off-target effects and making them highly attractive candidates for cancer immunotherapy [52]. Studies reporting rapid tumor regression in CRC patients treated with KRAS G12D-specific T cells highlight the potential of KRAS mutations as promising neoantigen targets for immunotherapy [2].

2.3. BRAF in CRC

BRAF is a serine/threonine kinase that plays a critical role in the MAPK signaling pathway alongside KRAS, regulating various cellular processes such as growth, differentiation, and survival [53]. As a member of the RAF kinase family, BRAF functions downstream of KRAS, where it activates MEK1/2, which in turn activates ERK1/2 to transduce cellular signals [54]. Interestingly, BRAF and KRAS mutations are mutually exclusive, meaning that they typically do not occur together in the same tumor [55]. While both mutations result in activation of the MAPK pathway, they operate through distinct mechanisms. BRAF kinase functions independently of RAS-GTP binding and RAF dimerization, leading to continuous activation of the signaling pathway in the presence of specific mutations [56]. Wild-type BRAF forms a negative feedback loop upstream in response to excessive ERK activation. In contrast, hyperactivation of the MAPK pathway due to genetic alterations contributes to CRC progression [57]. In CRC, 8–12% of patients with BRAF alterations carry the V600E mutation, which involves a substitution of valine (V) with glutamic acid (E) at position 600 in the BRAF protein [55,58]. The V600E mutation causes BRAF to become autonomously active by bypassing the normal regulatory mechanisms that control its activation [59]. This persistent activation of the MAPK signaling pathway leads to uncontrolled cell proliferation, resistance to apoptosis, and tumor progression—all of which are hallmarks of malignancy [53]. Additionally, the influence of BRAF mutations extends to the immune environment in CRC. The mutation not only drives tumor cell proliferation but also induces immune modulation, contributing to an immune evasion phenotype [60]. In MSI-H CRC, it can lead to an upregulation of immune checkpoint proteins, such as programmed cell death ligand 1 (PD-L1), which inhibits T cell-mediated immune responses. Although the altered immune environment is characterized by increased infiltration of neutrophils and M1 macrophages, reducing the presence of naïve CD4+ T cells, resting dendritic cells, and plasma cells are typically involved in immune tolerance [61]. These immune changes make BRAF-mutant CRC particularly challenging to treat, as the immunosuppressive TME hinders the effectiveness of many therapeutic strategies.
Recent studies have revealed that targeting the BRAF V600E mutation shows promise in clinical settings. Agents such as vemurafenib and encorafenib have demonstrated effectiveness in selectively inhibiting mutant BRAF, leading to tumor shrinkage in some cases of CRC [62,63]. However, even when BRAF is suppressed, the MAPK pathway is often reactivated through bypass mechanisms, necessitating combination therapy to overcome resistance. Ex vivo studies of T lymphocytes targeting mutant forms of BRAF or associated RAS mutations have also shown significant results [64]. This approach not only allows for the direct targeting of tumor cells but also modulates the immune TME, supporting other therapeutic strategies for anti-tumor activity [55]. Another strategy involves using TAAs to stimulate T cell responses through vaccination, offering hope for future therapeutic paradigms [65].

2.4. APC in CRC

The APC gene is a tumor suppressor gene that encodes a multidomain protein. APC plays a crucial role in regulating various cellular processes through interactions with multiple binding partners. These processes include chromosome segregation, cell migration, apoptosis, adhesion, proliferation, and differentiation. The diverse functions of APC highlight its significance in maintaining cellular homeostasis and preventing tumorigenesis [66]. Moreover, APC plays a crucial role in the activity of the Wnt/β-catenin signaling complex, interacting with components such as GSK3β, Disheveled, and Axin, and functioning as a key regulator of the Wnt signaling pathway. It also modulates the cell cycle and maintains chromosomal stability [67]. In the Wnt/β-catenin signaling pathway, which regulates at least 80 target genes, normal expression of the APC gene ensures proper degradation of β-catenin through phosphorylation and ubiquitination, thereby suppressing tumor formation. Maintaining low levels of β-catenin in the nucleus is important for the negative feedback regulation within the Wnt/β-catenin pathway. However, when APC functions abnormally, the accumulation of β-catenin triggers the transcription of Wnt target genes associated with cancer cell proliferation, survival, and metastasis, thereby creating an immunosuppressive TME [68,69,70]. It has been revealed that APC-mutated CRC exhibits elevated expression levels of vascular endothelial growth factor A (VEGFA) and marker of proliferation Ki67 (MKI67)along with reduced proportions of CD3⁺ and CD68⁺ cells. These findings indicate that APC mutations are associated with tumor invasion and proliferation [71]. In the early stages of CRC development, approximately 80% of CRC harbor inactivating mutations in the APC gene. APC mutations are prevalent across all stages of CRC progression [72]. These mutations initiate 80–85% of sporadic CRC cases, with the exception of MSI CRC status, which results from MMR deficiency [73]. Given the high prevalence of APC gene mutations, they hold potential as predictive biomarkers in CRC.

2.5. TP53 in CRC

The TP53 gene, a tumor suppressor gene, plays a critical role in preventing cancer development. It encodes the p53 protein, a transcription factor that regulates the cell cycle, DNA repair, senescence, and apoptosis, thereby preventing abnormal cell proliferation and division [66,74]. Wild-type p53 functions as a direct activator of the WAD-1 gene, which is induced to suppress tumor cells within the signaling pathway [66]. Programmed cell death pathways (PCD) are essential for the body to maintain homeostasis. TP53 is involved in regulating various types of PCD pathways, including apoptosis, autophagy, pyroptosis, and ferroptosis, as well as pathways related to reactive oxygen species (ROS) generation [74]. Under normal conditions, the p53 protein is continuously degraded by MDM2, maintaining low intracellular levels. However, when cellular stress occurs, such as DNA damage due to environmental stimuli or spontaneous replication errors, or when oncogenes and ROS are activated, p53 degradation by MDM2 is inhibited to maintain homeostasis. As a result, p53 protein accumulates, leading to elevated p53 levels [75,76]. As p53 levels increase, it promotes cell death by upregulating apoptosis-related proteins such as PUMA and NOXA [74,77].
The TP53 gene is one of the most frequently mutated genes in human malignancies. TP53 mutations are commonly observed in CRC [75,78]. TP53 mutations contribute to the progression of adenomas into malignant tumors in colorectal cancer by impairing apoptotic pathways, thereby promoting cellular proliferation and uncontrolled cell cycle progression [66]. More than 20% of TP53-mutated CRC harbor missense mutations at positions R175 or R273. These two mutational hotspots are known to promote tumor metastasis and reduce survival rates in CRC, suggesting that these specific TP53 mutations may play a significant role in CRC progression and patient outcomes [79]. The R175H mutation induces protein misfolding, resulting in structural alterations in the p53 DNA-binding domain. TP53 mutations harboring the R175H mutation enhance tumorigenic potential and increase the expression of drug resistance genes, leading to a poor prognosis for CRC patients [78,80]. Additionally, CRC tumors with R273 mutations are more likely to progress to metastatic diseases which are associated with lower survival rates. The R273H mutation regulates a specific transcriptional program that activates oncogenic signaling pathways and promotes the development of more aggressive tumors [79,81]. TP53 mutations, including those at the R273 position, decrease MHC-I expression and increase the secretion of immunosuppressive cytokines, thereby promoting the activation of CAFs. The activation of CAFs leads to the creation of a cold tumor environment, which acts as a mechanism to enhance immune evasion. Furthermore, overexpression of TP53 mutations increases PD-L1 expression levels by activating AKT, thereby inhibiting T cell activation [82]. Notably, when accompanied by KRAS mutations, a more potent immunosuppressive TME is established, ultimately resulting in immune escape [82,83].

3. Immunological Function of TME in CRC

CRC is a malignancy characterized by a complex TME, in which cellular and non-cellular components interact to facilitate immune evasion, tumor progression, and therapeutic resistance [84]. Cellular components (e.g., endothelial cells, CAFs, T and B lymphocytes, APCs, and mast cells) and non-cellular components, such as ECM, cytokines, growth factors, and vascular structures, are intricately interconnected, contributing to immunosuppression [85,86,87]. Furthermore, cancer cells actively remodel the TME by recruiting immunosuppressive mediators such as myeloid-derived suppressor cells (MDSCs), Tregs, tumor-associated macrophages (TAMs), and CAFs, while simultaneously modulating their own immunogenicity through signature gene mutations [88,89]. For these reasons, CRC is classified as a “cold tumor,” exhibiting poor responsiveness to immunotherapy [90]. As cold tumors, TME in CRC suppresses infiltrating T cells, NK cells, and dendritic cells, leading to a non-inflamed phenotype [17,83,91].
MDSCs secrete various molecules, including nitric oxide (NO), ROS, TGF-β, and cytokines such as IL-1 and IL-6 [83]. These molecules promote an acidic environment through lactic acid–induced hypoxia-inducible factor-1 alpha (HIF-1α), leading to the expression of immune checkpoint molecules such as PD-L1 [92,93]. In particular, the accumulation of ROS and NO promotes tumorigenesis while suppressing anti-tumor immune responses [93,94]. TAMs also contribute to immune evasion under hypoxic conditions alongside MDSCs [17]. TAMs, in cooperation with Tregs, transform the TME into an anti-inflammatory state by secreting chemokines such as CC Motif Chemokine Ligand 2 (CCL2), CC Motif Chemokine Ligand 5 (CCL5), VEGF, and Transforming growth factor beta (TGF-β), thereby promoting the growth and progression of advanced CRC [95]. Specifically, KRAS mutations influence the polarization of macrophages toward a TAM-like phenotype, characterized by increased expression of immunosuppressive cytokines such as IL-10 and TGF-β [96]. Tregs secrete TGF-β, IL-10, and IL-35, which induce T cell exhaustion and suppress antigen presentation by APCs [97]. In cancers harboring both KRAS and TP53 mutations, an increased infiltration of Tregs has been observed [98]. CAFs, as a key component of the tumor stroma responsible for synthesizing and maintaining the ECM, play critical roles in tumor initiation and progression [99]. CAFs can be broadly classified into two subtypes: inflammatory CAFs (iCAFs) and myofibroblastic CAFs (myoCAFs). iCAFs are characterized by elevated secretion of IL-6, whereas myoCAFs predominantly secrete TGF-β. Notably, CRCs primarily exhibit the iCAF phenotype, leading to increased IL-6 production and enhanced MDSC recruitment. This process is further exacerbated by KRAS mutations, which promote CXCL1 transcription and enhance MDSC mobilization into the tumor microenvironment [100,101]. These mechanisms provide a foundation for cancer cells to evade immune surveillance and develop resistance to immunotherapy. Comprehensively, converting cold tumors into hot tumors is a central strategy in cancer immunotherapy, as it can enhance the effectiveness and response rate of treatment (Figure 2).

4. Applications of Cancer Vaccines Targeting Signature Gene Mutations in CRC

As mentioned above, CRC is characterized by frequent mutations in specific genes, including MMR, KRAS, BRAF, APC, and TP53, which contribute to tumor progression and immune evasion. Given their crucial roles in shaping the TME, these signature gene mutations are regarded as promising targets for immunotherapy. Cancer vaccines offer an alternative immunotherapeutic approach by priming and expanding tumor-specific T cells before tumor-mediated immune suppression becomes dominant. For this reason, cancer vaccines have emerged as a promising strategy by inducing tumor-specific immune responses and providing a potential approach for CRC treatment [20,102]. Moreover, cancer vaccines can be combined with ICIs to enhance their efficacy by increasing T-cell infiltration in MSS CRC, thereby overcoming one of the major resistance mechanisms to ICI therapy [103]. The main mechanism of cancer vaccines is to introduce selected antigens into APCs to elicit immune responses [21]. Cancer vaccines can be classified into cell-based, peptide-based, nucleic acid-based, and viral vector-based types, depending on the method of APC activation.

4.1. Types of Cancer Vaccines

4.1.1. Cell-Based Vaccine

DCs, as key antigen-presenting cells in peripheral tissues, are responsible for capturing, processing, and presenting antigenic peptides, including TAAs and TSAs. They are pivotal in initiating and regulating CD8⁺ and CD4⁺ T cell responses, particularly in antitumor immunity [104]. Cell-based cancer vaccines, particularly DC vaccines, harness a patient’s dendritic cells to elicit specific immune responses that selectively eliminate target cells. These cells are activated ex vivo by loading them with autologous antigens, after which they are reintroduced into the body [105,106]. Another approach to cellular vaccination involves using whole-cell preparations derived from tumor cells. These cells are either inactivated or genetically modified to prevent pathogenicity. They are recognized by various immune cells, including dendritic cells, macrophages, and NK cells, thereby eliciting broad, non-specific immune responses [27]. These vaccines can serve as safe adjuvants to enhance the efficacy of other anticancer therapies, such as chemotherapy and ICIs [107]. A representative DC-based cancer vaccine is Provenge (sipuleucel-T), the first FDA-approved therapeutic cancer vaccine for prostate cancer [108]. In CRC, GVAX, composed of CRC cells and used in an adjuvant setting, has demonstrated safety and the ability to enhance antitumor immunity [109]. However, a major limitation of cell-based vaccines is the potential for HLA mismatch, which can compromise vaccine efficacy. Therefore, the use of personalized elements such as neoantigens is essential to maximize therapeutic effectiveness.

4.1.2. Peptide-Based Vaccine

Peptide-based cancer vaccines use synthetic peptides to stimulate the immune system. These peptides typically consist of short amino acid sequences containing specific antigenic epitopes [110]. Peptide length influences the type of immune response elicited. Short peptides (8–11 amino acids) are presented by MHC-I molecules and recognized by CD8⁺ T cells, leading to the generation of cytotoxic T lymphocytes (CTLs). In contrast, long peptides (11–30 amino acids) are presented by MHC-II molecules and recognized by CD4⁺ T cells, inducing helper T cell responses [111,112]. The selection of appropriate tumor antigens is critical in designing peptide vaccines. Targeting neoantigens—abnormally overexpressed or mutated proteins specific to cancer cells—is essential for effective cancer vaccine development [112]. Peptide-based vaccines alone are often insufficient due to their low immunogenicity. Therefore, potent adjuvants or immune stimulants are required to induce robust immune responses [113]. Despite certain limitations, peptide-based vaccines remain an attractive therapeutic option due to their ease of synthesis and low production cost [111]. Peptide-based vaccines are currently being evaluated in clinical trials, and in CRC, ELI-002 targeting KRAS mutations has demonstrated safety and promising therapeutic efficacy [114,115]. NCT04117087 is a Phase I study investigating a synthetic long peptide (SLP) KRAS vaccine in combination with dual checkpoint blockade (ipilimumab and nivolumab). The trial aims to enhance immune responses in cold tumors, including MSS CRC. Overall, induction of KRAS-specific T cell responses has been associated with improved survival [116,117]. These findings support peptide-based cancer vaccines as a promising strategy for CRC treatment.

4.1.3. Nucleic Acid-Based Vaccine

Nucleic acid-based cancer vaccines deliver plasmid DNA (pDNA) or mRNA into APCs to activate the immune system [118]. Nucleic acid-based vaccines offer unique advantages, including rapid development, efficient manufacturing, and ease of customization [119]. Furthermore, nucleic acid vaccines have demonstrated favorable safety profiles compared to traditional vaccines and viral vector platforms, which rely on live-attenuated or replication-deficient viruses. Briefly, nucleic acid-based vaccines are more efficient, easier to produce, and readily modifiable than other vaccine types [120]. mRNA vaccines have emerged as a promising therapeutic platform since the COVID-19 pandemic. They allow flexible sequence design and enable the encoding of tumor antigens, thereby inducing both innate and adaptive immune responses. Additionally, unlike DNA vaccines, mRNA vaccines eliminate the risk of host genome integration [120]. Due to the instability of mRNA vaccines, an optimized delivery system is essential for protection against degradation, with lipid nanoparticles (LNPs) serving as a key formulation for effective mRNA delivery [121]. Despite remaining challenges such as potential side effects and limited efficacy, various mRNA-based cancer vaccines are currently undergoing clinical trials [119,122]. BNT122 (NCT04486378), an mRNA-based vaccine, is being investigated for the treatment of stage II/III CRC. This study demonstrates that BNT122 induces neoantigen-specific T cell responses, leading to prolonged disease-free survival [123]. In addition, mRNA-5671 (NCT03948763), targeting KRAS mutations, is undergoing a Phase I trial in combination with pembrolizumab, a PD-1 inhibitor [119]. These studies underscore the growing potential of mRNA-based vaccines as a novel approach to cancer immunotherapy.

4.1.4. Viral Vector-Based Vaccine

Viruses inherently possess immunogenic properties, and their genomes can be engineered to deliver transgenes for expression in host cells, including immune cells [124]. Therefore, they can elicit strong and long-lasting immune responses without the need for adjuvants. Various viruses are utilized as recombinant viral vectors, including adenoviruses, poxviruses, herpesviruses, and lentiviruses [125]. These viral vectors are human-compatible, non-immunogenic, and non-pathogenic, and offer efficient gene delivery compared to other vaccine platforms [126,127]. While conventional viral vector vaccines aim to stimulate immune responses via APCs, oncolytic virus therapy represents an alternative viral approach that directly targets and destroys cancer cells [127]. It aims to identify, infect, and lyse cancer cells or cells within the TME to inhibit tumor progression and activate antitumor immune responses [128]. Nevertheless, oncolytic viruses are potentially strong therapies for cancer treatment, they cannot be enough to eliminate all the cancer cells because of the heterogeneity and complicated structure of tumors [129]. Thus, further research is required to advance both viral vector-based vaccines and oncolytic virus therapies. The first approved oncolytic virus drug, T-VEC, is based on a genetically modified herpes simplex virus type 1 encoding granulocyte-macrophage colony-stimulating factor (GM-CSF) and is used to treat unresectable melanoma [130]. In CRC, Pexa-Vec, an oncolytic virus therapy, is undergoing Phase I/II clinical trials in combination with ICIs. This combination has shown promising activity in advanced MSS CRC [131,132]. NCT03953235 is a viral vector-based vaccine composed of chimpanzee adenovirus (ChAd68) and self-amplifying mRNA (samRNA). This shared neoantigen vaccine, when combined with ICIs, has been shown to extend median progression-free survival and overall survival [133]. Consequently, ongoing research on both viral vector vaccines and oncolytic virus therapies is essential to address current challenges in cancer treatment.

4.2. Advantages and Challenges of Cancer Vaccines Targeting Signature Genes

Various cancer vaccines targeting signature mutations in CRC are being developed using diverse platforms each with distinct mechanisms (Table 1). In this section, we summarize vaccine candidates currently being studied and discuss their advantages and strategies to overcome challenges. GVAX, cell-based vaccines have been designed to stimulate broad immune activation by secreting GM-CSF [134]. Despite eliciting strong immune responses, their efficacy as monotherapies has been constrained by the immunosuppressive TME and insufficient antigen specificity. Similarly, TG01, a peptide vaccine designed to induce DTH and T-cell proliferation, has achieved immune activation but only modest clinical benefits when used alone [135]. These suggest that combination strategies, such as pairing with ICIs or T-cell modulators, may be critical to fully amplify the therapeutic potential of these vaccines. To enhance immune responses, some platforms focused on boosting antigen-specific T-cell activation. ELI-002, a lipopeptide-based vaccine utilizing AMP-CpG technology, has demonstrated that [136], although sustaining durable memory responses remains a challenge. Long peptide-based vaccines targeting KRAS neoantigens have shown the ability to broaden T-cell diversity by activating both CD4⁺ and CD8⁺ subsets. Ongoing efforts to optimize antigen selection, epitope processing, and delivery methods are expected to further improve the clinical efficacy of these peptide-based platforms. E-39 has shown immune engagement via cytolysis with TALs [137], and thus enhancing the persistence and functionality of activated T cells may strengthen the durable therapeutic outcomes. More recent advancements have aimed to fundamentally address tumor heterogeneity and recurrence at their origin. The KISIMA-Ascl2 vaccine, targets ISC-related antigens, and is promising to combat tumor stemness and therapy resistance [138]. This approach holds significant potential for achieving long-term disease control even though it is still in the early clinical stages. Moreover, mRNA-based vaccines such as BNT122 and mRNA-5681 have emerged as highly adaptable tools for inducing tumor-specific cytotoxic T-cell responses and antigen expression [139,140]. Vector-based strategies, GRT-R904 employs a prime-boost approach [133], and oncolytic virus-based vaccines like Pexa-Vec (JX-594) directly lyse tumor cells and stimulate systemic immunity [131], further diversify the vaccine landscape. Nevertheless, overcoming hurdles such as vector-specific immune clearance and sustaining robust immune memory remains essential to promote their effectiveness. Finally, Nous-209 offers an alternative strategy by inducing FSP-specific T-cell responses promise in MSI-H CRC [141], although broader validation in MSS CRC remains an important next step. Taken together, these show significant progress in CRC immunotherapy, each addressing unique aspects of tumor immunity. To maximize their therapeutic potential, refining antigen targeting, immune activation, and combination strategies is pivotal. With ongoing study and clinical validation, these vaccines offer strong potential for durable, effective treatments against colorectal cancer.

4.3. Development and Limitations of Signature Gene-Targeted Vaccines in CRC

KRAS and BRAF mutations have remained the primary focus of vaccine development in clinical trials, as noted above [145]. These mutations generate strong tumor-specific neoantigens, making them attractive targets for cancer vaccine development. In particular, KRAS mutations such as G12D, G12V, and G13D produce neoantigens recognizable by the immune system, eliciting CD8⁺ T cell responses. Similarly, the BRAF V600E mutation generates a well-characterized neoantigen that has been actively explored in immunotherapy strategies [48,146]. TP53 mutations are primarily associated with loss-of-function, which means that they fail to produce stable and immunogenic neoantigens suitable for vaccine targeting; however, certain gain-of-function phenotypes can elicit robust cytotoxic and T helper cell-mediated immune responses [31]. Simultaneously targeting KRAS and TP53 mutations using long-peptide vaccination induced strong cytotoxic and helper T cell responses [147]. In addition, various vaccine platforms have been employed to target mutant TP53, aiming to enhance immune responses by increasing CD8⁺ T cell activity [148]. Since mutant p53-derived peptide neoantigens are commonly present in various cancer types, they represent a promising target for cancer vaccine development [149]. APC mutations have been proposed as potential sources of tumor-specific neoantigens for cancer vaccine development [150]. However, a major challenge is that APC mutations primarily result in loss-of-function, which limits their capacity to generate directly immunogenic neoantigens [151,152,153]. Consequently, recent research has expanded the concept of neoantigens beyond peptides derived solely from gain-of-function mutations [154]. Loss-of-function mutations may indirectly give rise to neoantigens by causing the aberrant expression of associated signaling proteins. This shift in perspective has led to the identification of overexpressed signaling components as promising vaccine targets in CRC [155]. A recent study has evaluated the efficacy of the KISIMA-Ascl2 vaccine, which targets the self-antigen Ascl2, in an APC-mutant mouse model (Apc+/Min-FCCC mice) [138]. Ascl2 is a key regulator in the Wnt/β-catenin pathway and is overexpressed due to APC mutations [156]. Following vaccination, both T-cell and antibody responses were significantly enhanced in mice. Notably, the immune response was further amplified and exhibited a stronger tumor-suppressive effect when combined with anti-PD-1 antibodies. These results demonstrated that the KISIMA-Ascl2 vaccine effectively suppressed colonic adenoma formation induced by APC mutations, while also confirming its immunogenicity and safety [138]. Similarly, MMR gene mutations are difficult to directly target with vaccines; however, they can generate frameshift-derived neoantigens that are targetable [157]. Current research on vaccines for dMMR CRC primarily focuses on utilizing these neoantigens. For instance, Nous-209, a vaccine targeting 209 frameshift peptides derived from dMMR/MSI-H tumors, has demonstrated safety and the ability to elicit immune responses when combined with ICIs [144]. Although loss-of-function mutations are limited in directly exposing antigens on cancer cells, vaccines targeting associated overexpressed proteins hold potential for effectively suppressing tumors.

5. Conclusions

In CRC, signature gene mutations play a pivotal role in tumor growth, progression, and the formation of the tumor microenvironment, highlighting their potential as targets for cancer vaccines. Current clinical trials and research efforts in cancer vaccine development have primarily focused on KRAS and BRAF mutations due to their ability to generate strong neoantigens. However, tumor heterogeneity and complex immune evasion mechanisms complicate therapeutic strategies, and the need for personalized approaches tailored to individual tumor characteristics presents a major limitation. Nonetheless, successful applications of cancer vaccines in other solid tumors offer promise for colorectal cancer treatment. For instance, neoantigen-based approaches and combination strategies with ICIs—proven effective in melanoma and lung cancer—may also be applied to colorectal cancer, a solid tumor, to develop effective therapeutic strategies [158,159]. Personalized approaches using individualized cancer vaccines, improvements in neoantigen prediction technologies, and advances in diverse vaccine platforms collectively support the potential for developing effective cancer vaccines for colorectal cancer. Notably, emerging technologies such as mRNA-based vaccines may facilitate the development of personalized vaccines targeting colorectal cancer-specific antigens [52,160]. In parallel, the development of advanced bioinformatics tools has played a crucial role in supporting personalized vaccine strategies. Platforms such as NetMHCpan and pVACtools assist in predicting immunogenic neoantigens and optimizing HLA typing, thereby streamlining the design of individualized mRNA-based vaccines [161,162]. As these computational approaches continue to advance, they are expected to narrow the gap between molecular tumor profiling and clinical application, ultimately making the translation of personalized immunotherapies into clinical practice. Future research should prioritize integrated strategies targeting multiple signature genes simultaneously. Additionally, elucidating the synergistic effects of combining cancer vaccines with conventional therapies is essential. In conclusion, cancer vaccines hold the potential to redefine colorectal cancer treatment and, with continued research, may offer more effective and personalized therapeutic options for patients.

Author Contributions

J.Y., H.M. and Y.J. mainly designed and J.Y., H.M., Y.J. and S.C. wrote the manuscript; H.Y. supervised, organized, and wrote the manuscript; all authors revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Catholic University of Korea, Research Fund, 2022 (# M2022B00080009) and the Ministry of Food and Drug Safety in Korea (22213MFDS421).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AKTProtein kinase B
APCAdenomatous polyposis coli
APCsAntigens-presenting cells
Ascl2Achaete-scute family bHLH transcription factor 2
BRAFB-Raf proto-oncogene, serine/threonine kinase
CAFsCancer-associated fibroblasts
ChAdChimpanzee adenovirus
CK IαCasein kinase I alpha
CRCColorectal cancer
CTLsCytotoxic T lymphocytes
dMMRMismatch repair deficiency
DCDendritic cell
DTHDelayed-type hypersensitivity
ECMExtracellular matrix
EPCAMEpithelial cell adhesion molecule
ERKExtracellular signal-regulated kinase
FSPFrameshift peptide
GaDGorilla adenovirus
GM-CSFGranulocyte-macrophage colony-stimulating factor
GSK-3βGlycogen synthase kinase-3β
HIF-1αHypoxia-inducible factor-1 alpha
HLAHuman leukocyte antigen
ICIsImmune checkpoint inhibitors
iCAFsInflammatory CAFs
ISCIntestinal stem cell
KRASKirsten rat sarcoma virus
LNPLipid nanoparticle
MEKMitogen-activated protein kinase
MHCMajor histocompatibility complex
MKI67Marker of proliferation Ki-67
MLH1mutL homolog1
MMRMismatch repair
MDSCsMyeloid-derived suppressor cells
MSIMicrosatellite instability
MSI-HMicrosatellite instability-high
MSSMicrosatellite stable
mTORC1Mammalian target of rapamycin complex1
myoCAFsMyofibroblastic CAFs
MSH2muts homolog2
MSH6muts homolog6
NONitric oxide
pDNAPlasmid DNA
PD-L1Programmed cell death-ligand 1
PCDProgrammed cell death
PI3KPhosphatidylinositol-3-kinase
PMS2Postmeiotic segregation increased2
PCDProgrammed cell death
ROSReactive oxygen species
samRNASelf-amplifying mRNA
SLPSynthetic long peptide
TALTumor-associated lymphocyte
TAMsTumor-associated macrophages
TAAsTumor-associated antigens
TILsTumor-infiltrating lymphocytes
TMBTumor mutation burden
TMETumor microenvironment
TregRegulatory T cell
TSAsTumor-specific antigens

References

  1. Xi, Y.; Xu, P. Global colorectal cancer burden in 2020 and projections to 2040. Transl. Oncol. 2021, 14, 101174. [Google Scholar] [CrossRef] [PubMed]
  2. Wang, D.; Zhang, H.; Xiang, T.; Wang, G. Clinical application of adaptive immune therapy in MSS colorectal cancer patients. Front. Immunol. 2021, 12, 762341. [Google Scholar] [CrossRef]
  3. Nunes, L.; Stenersen, J.M.; Kryeziu, K.; Sjöblom, T.; Glimelius, B.; Lothe, R.A.; Sveen, A. Co-occurring mutations identify prognostic subgroups of microsatellite stable colorectal cancer. Mol. Cancer 2024, 23, 264. [Google Scholar] [CrossRef] [PubMed]
  4. Lee, C.S.; Song, I.H.; Lee, A.; Kang, J.; Lee, Y.S.; Lee, I.K.; Song, Y.S.; Lee, S.H. Enhancing the landscape of colorectal cancer using targeted deep sequencing. Sci. Rep. 2021, 11, 8154. [Google Scholar] [CrossRef]
  5. Poturnajova, M.; Furielova, T.; Balintova, S.; Schmidtova, S.; Kucerova, L.; Matuskova, M. Molecular features and gene expression signature of metastatic colorectal cancer. Oncol. Rep. 2021, 45, 10. [Google Scholar] [CrossRef] [PubMed]
  6. Sahin, I.H.; Goyal, S.; Pumpalova, Y.; Sonbol, M.B.; Das, S.; Haraldsdottir, S.; Ahn, D.; Ciombor, K.K.; Chen, Z.; Draper, A. Mismatch Repair (MMR) gene alteration and BRAF V600E mutation are potential predictive biomarkers of immune checkpoint inhibitors in MMR-deficient colorectal cancer. Oncologist 2021, 26, 668–675. [Google Scholar] [CrossRef] [PubMed]
  7. Meng, M.; Zhong, K.; Jiang, T.; Liu, Z.; Kwan, H.Y.; Su, T. The current understanding on the impact of KRAS on colorectal cancer. Biomed. Pharmacother. 2021, 140, 111717. [Google Scholar] [CrossRef]
  8. Fanelli, G.N.; Dal Pozzo, C.A.; Depetris, I.; Schirripa, M.; Brignola, S.; Biason, P.; Balistreri, M.; Dal Santo, L.; Lonardi, S.; Munari, G. The heterogeneous clinical and pathological landscapes of metastatic Braf-mutated colorectal cancer. Cancer Cell Int. 2020, 20, 30. [Google Scholar] [CrossRef]
  9. Feng, F.; Sun, H.; Zhao, Z.; Sun, C.; Zhao, Y.; Lin, H.; Yang, J.; Xiao, Y.; Wang, W.; Wu, D. Identification of APC mutation as a potential predictor for immunotherapy in colorectal cancer. J. Oncol. 2022, 2022, 6567998. [Google Scholar] [CrossRef]
  10. Wang, C.; Ouyang, C.; Cho, M.; Ji, J.; Sandhu, J.; Goel, A.; Kahn, M.; Fakih, M. Wild-type APC Is Associated with Poor Survival in Metastatic Microsatellite Stable Colorectal Cancer. Oncologist 2021, 26, 208–214. [Google Scholar] [CrossRef]
  11. Liebl, M.C.; Hofmann, T.G. The role of p53 signaling in colorectal cancer. Cancers 2021, 13, 2125. [Google Scholar] [CrossRef] [PubMed]
  12. Liu, J.; Huang, X.; Liu, H.; Wei, C.; Ru, H.; Qin, H.; Lai, H.; Meng, Y.; Wu, G.; Xie, W. Immune landscape and prognostic immune-related genes in KRAS-mutant colorectal cancer patients. J. Transl. Med. 2021, 19, 27. [Google Scholar] [CrossRef] [PubMed]
  13. Underwood, P.W.; Ruff, S.M.; Pawlik, T.M. Update on targeted therapy and immunotherapy for metastatic colorectal cancer. Cells 2024, 13, 245. [Google Scholar] [CrossRef]
  14. Bejarano, L.; Jordāo, M.J.; Joyce, J.A. Therapeutic targeting of the tumor microenvironment. Cancer Discov. 2021, 11, 933–959. [Google Scholar] [CrossRef]
  15. Wang, L.; Geng, H.; Liu, Y.; Liu, L.; Chen, Y.; Wu, F.; Liu, Z.; Ling, S.; Wang, Y.; Zhou, L. Hot and cold tumors: Immunological features and the therapeutic strategies. MedComm 2023, 4, e343. [Google Scholar] [CrossRef]
  16. Ouyang, P.; Wang, L.; Wu, J.; Tian, Y.; Chen, C.; Li, D.; Yao, Z.; Chen, R.; Xiang, G.; Gong, J. Overcoming cold tumors: A combination strategy of immune checkpoint inhibitors. Front. Immunol. 2024, 15, 1344272. [Google Scholar] [CrossRef]
  17. Khosravi, G.R.; Mostafavi, S.; Bastan, S.; Ebrahimi, N.; Gharibvand, R.S.; Eskandari, N. Immunologic tumor microenvironment modulators for turning cold tumors hot. Cancer Commun. 2024, 44, 521–553. [Google Scholar] [CrossRef]
  18. Benoit, A.; Vogin, G.; Duhem, C.; Berchem, G.; Janji, B. Lighting up the fire in the microenvironment of cold tumors: A major challenge to improve cancer immunotherapy. Cells 2023, 12, 1787. [Google Scholar] [CrossRef]
  19. Weng, J.; Li, S.; Zhu, Z.; Liu, Q.; Zhang, R.; Yang, Y.; Li, X. Exploring immunotherapy in colorectal cancer. J. Hematol. Oncol. 2022, 15, 95. [Google Scholar] [CrossRef]
  20. Lin, M.J.; Svensson-Arvelund, J.; Lubitz, G.S.; Marabelle, A.; Melero, I.; Brown, B.D.; Brody, J.D. Cancer vaccines: The next immunotherapy frontier. Nat. Cancer 2022, 3, 911–926. [Google Scholar] [CrossRef]
  21. Miao, L.; Zhang, Y.; Huang, L. mRNA vaccine for cancer immunotherapy. Mol. Cancer 2021, 20, 41. [Google Scholar] [CrossRef] [PubMed]
  22. Morse, M.A.; Gwin, W.R., III; Mitchell, D.A. Vaccine therapies for cancer: Then and now. Target. Oncol. 2021, 16, 121–152. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, J.; Fu, M.; Wang, M.; Wan, D.; Wei, Y.; Wei, X. Cancer vaccines as promising immuno-therapeutics: Platforms and current progress. J. Hematol. Oncol. 2022, 15, 28. [Google Scholar] [CrossRef]
  24. Zhang, Y.; Ma, J.-A.; Zhang, H.-X.; Jiang, Y.-N.; Luo, W.-H. Cancer vaccines: Targeting KRAS-driven cancers. Expert Rev. Vaccines 2020, 19, 163–173. [Google Scholar] [CrossRef]
  25. Liu, C.; Zheng, S.; Jin, R.; Wang, X.; Wang, F.; Zang, R.; Xu, H.; Lu, Z.; Huang, J.; Lei, Y. The superior efficacy of anti-PD-1/PD-L1 immunotherapy in KRAS-mutant non-small cell lung cancer that correlates with an inflammatory phenotype and increased immunogenicity. Cancer Lett. 2020, 470, 95–105. [Google Scholar] [CrossRef]
  26. Kumar, A.; Gautam, V.; Sandhu, A.; Rawat, K.; Sharma, A.; Saha, L. Current and emerging therapeutic approaches for colorectal cancer: A comprehensive review. World J. Gastrointest. Surg. 2023, 15, 495. [Google Scholar] [CrossRef]
  27. Kaczmarek, M.; Poznańska, J.; Fechner, F.; Michalska, N.; Paszkowska, S.; Napierała, A.; Mackiewicz, A. Cancer vaccine therapeutics: Limitations and effectiveness—A literature review. Cells 2023, 12, 2159. [Google Scholar] [CrossRef]
  28. Wan, Y.; Zhang, Y.; Wang, G.; Mwangi, P.M.; Cai, H.; Li, R. Recombinant KRAS G12D protein vaccines elicit significant anti-tumor effects in mouse CT26 tumor models. Front. Oncol. 2020, 10, 1326. [Google Scholar] [CrossRef]
  29. Ahmad, R.; Singh, J.K.; Wunnava, A.; Al-Obeed, O.; Abdulla, M.; Srivastava, S.K. Emerging trends in colorectal cancer: Dysregulated signaling pathways. Int. J. Mol. Med. 2021, 47, 14. [Google Scholar] [CrossRef]
  30. Wan, M.-l.; Wang, Y.; Zeng, Z.; Deng, B.; Zhu, B.-s.; Cao, T.; Li, Y.-k.; Xiao, J.; Han, Q.; Wu, Q. Colorectal cancer (CRC) as a multifactorial disease and its causal correlations with multiple signaling pathways. Biosci. Rep. 2020, 40, BSR20200265. [Google Scholar] [CrossRef]
  31. Zheng, Y.; Fu, Y.; Wang, P.-P.; Ding, Z.-Y. Neoantigen: A promising target for the immunotherapy of colorectal cancer. Dis. Markers 2022, 2022, 8270305. [Google Scholar] [CrossRef] [PubMed]
  32. Alzahrani, S.M.; Al Doghaither, H.A.; Al-Ghafari, A.B. General insight into cancer: An overview of colorectal cancer. Mol. Clin. Oncol. 2021, 15, 271. [Google Scholar] [CrossRef] [PubMed]
  33. Ijsselsteijn, R.; Jansen, J.G.; de Wind, N. DNA mismatch repair-dependent DNA damage responses and cancer. DNA Repair 2020, 93, 102923. [Google Scholar] [CrossRef] [PubMed]
  34. Fan, W.-X.; Su, F.; Zhang, Y.; Zhang, X.-L.; Du, Y.-Y.; Gao, Y.-J.; Li, W.-L.; Hu, W.-Q.; Zhao, J. Oncological characteristics, treatments and prognostic outcomes in MMR-deficient colorectal cancer. Biomark. Res. 2024, 12, 89. [Google Scholar] [CrossRef]
  35. Dal Buono, A.; Gaiani, F.; Poliani, L.; Correale, C.; Laghi, L. Defects in MMR genes as a seminal example of personalized medicine: From diagnosis to therapy. J. Pers. Med. 2021, 11, 1333. [Google Scholar] [CrossRef]
  36. Venetis, K.; Frascarelli, C.; Bielo, L.B.; Cursano, G.; Adorisio, R.; Ivanova, M.; Mane, E.; Peruzzo, V.; Concardi, A.; Negrelli, M. Mismatch repair (MMR) and microsatellite instability (MSI) phenotypes across solid tumors: A comprehensive cBioPortal study on prevalence and prognostic impact. Eur. J. Cancer 2025, 217, 115233. [Google Scholar] [CrossRef]
  37. Taieb, J.; Svrcek, M.; Cohen, R.; Basile, D.; Tougeron, D.; Phelip, J.-M. Deficient mismatch repair/microsatellite unstable colorectal cancer: Diagnosis, prognosis and treatment. Eur. J. Cancer 2022, 175, 136–157. [Google Scholar] [CrossRef]
  38. Roudko, V.; Cimen Bozkus, C.; Greenbaum, B.; Lucas, A.; Samstein, R.; Bhardwaj, N. Lynch syndrome and MSI-H cancers: From mechanisms to “off-the-shelf” cancer vaccines. Front. Immunol. 2021, 12, 757804. [Google Scholar] [CrossRef]
  39. Patelli, G.; Tosi, F.; Amatu, A.; Mauri, G.; Curaba, A.; Patanè, D.; Pani, A.; Scaglione, F.; Siena, S.; Sartore-Bianchi, A. Strategies to tackle RAS-mutated metastatic colorectal cancer. ESMO Open 2021, 6, 100156. [Google Scholar] [CrossRef]
  40. Liu, P.; Wang, Y.; Li, X. Targeting the untargetable KRAS in cancer therapy. Acta Pharm. Sin. B 2019, 9, 871–879. [Google Scholar] [CrossRef]
  41. Zhu, G.; Pei, L.; Xia, H.; Tang, Q.; Bi, F. Role of oncogenic KRAS in the prognosis, diagnosis and treatment of colorectal cancer. Mol. Cancer 2021, 20, 143. [Google Scholar] [CrossRef] [PubMed]
  42. Kilowski, K.A.; Dietrich, M.F.; Xiu, J.; Baca, Y.; Hinton, A.; Ahmad, S.; Herzog, T.J.; Thaker, P.; Holloway, R.W. KRAS mutations in endometrial cancers: Possible prognostic and treatment implications. Gynecol. Oncol. 2024, 191, 299–306. [Google Scholar] [CrossRef] [PubMed]
  43. Ros, J.; Vaghi, C.; Baraibar, I.; Saoudi González, N.; Rodríguez-Castells, M.; García, A.; Alcaraz, A.; Salva, F.; Tabernero, J.; Elez, E. Targeting KRAS G12C Mutation in Colorectal Cancer, A Review: New Arrows in the Quiver. Int. J. Mol. Sci. 2024, 25, 3304. [Google Scholar] [CrossRef] [PubMed]
  44. Qunaj, L.; May, M.S.; Neugut, A.I.; Herzberg, B.O. Prognostic and therapeutic impact of the KRAS G12C mutation in colorectal cancer. Front. Oncol. 2023, 13, 1252516. [Google Scholar] [CrossRef]
  45. Poole, A.; Karuppiah, V.; Hartt, A.; Haidar, J.N.; Moureau, S.; Dobrzycki, T.; Hayes, C.; Rowley, C.; Dias, J.; Harper, S. Therapeutic high affinity T cell receptor targeting a KRASG12D cancer neoantigen. Nat. Commun. 2022, 13, 5333. [Google Scholar] [CrossRef]
  46. Cefalì, M.; Epistolio, S.; Palmarocchi, M.C.; Frattini, M.; De Dosso, S. Research progress on KRAS mutations in colorectal cancer. J. Cancer Metastasis Treat. 2021, 7, 26. [Google Scholar] [CrossRef]
  47. Negri, F.; Bottarelli, L.; de’Angelis, G.L.; Gnetti, L. KRAS: A druggable target in colon cancer patients. Int. J. Mol. Sci. 2022, 23, 4120. [Google Scholar] [CrossRef]
  48. Canon, J.; Rex, K.; Saiki, A.Y.; Mohr, C.; Cooke, K.; Bagal, D.; Gaida, K.; Holt, T.; Knutson, C.G.; Koppada, N. The clinical KRAS (G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 2019, 575, 217–223. [Google Scholar] [CrossRef]
  49. Wang, Q.J.; Yu, Z.; Griffith, K.; Hanada, K.-i.; Restifo, N.P.; Yang, J.C. Identification of T-cell receptors targeting KRAS-mutated human tumors. Cancer Immunol. Res. 2016, 4, 204–214. [Google Scholar] [CrossRef]
  50. Linette, G.P.; Bear, A.S.; Carreno, B.M. Facts and hopes in immunotherapy strategies targeting antigens derived from KRAS mutations. Clin. Cancer Res. 2024, 30, 2017–2024. [Google Scholar] [CrossRef]
  51. Xie, N.; Shen, G.; Gao, W.; Huang, Z.; Huang, C.; Fu, L. Neoantigens: Promising targets for cancer therapy. Signal Transduct. Target. Ther. 2023, 8, 9. [Google Scholar] [CrossRef] [PubMed]
  52. Zhu, Y.J.; Li, X.; Chen, T.T.; Wang, J.X.; Zhou, Y.X.; Mu, X.L.; Du, Y.; Wang, J.L.; Tang, J.; Liu, J.Y. Personalised neoantigen-based therapy in colorectal cancer. Clin. Transl. Med. 2023, 13, e1461. [Google Scholar] [CrossRef] [PubMed]
  53. Grothey, A.; Fakih, M.; Tabernero, J. Management of BRAF-mutant metastatic colorectal cancer: A review of treatment options and evidence-based guidelines. Ann. Oncol. 2021, 32, 959–967. [Google Scholar] [CrossRef] [PubMed]
  54. Guerrero, R.M.; Labajos, V.A.; Ballena, S.L.; Macha, C.A.; Lezama, M.S.; Roman, C.P.; Beltran, P.M.; Torrejon, A.F. Targeting BRAF V600E in metastatic colorectal cancer: Where are we today? Ecancermedicalscience 2022, 16, 1489. [Google Scholar]
  55. Grassi, E.; Corbelli, J.; Papiani, G.; Barbera, M.A.; Gazzaneo, F.; Tamberi, S. Current therapeutic strategies in BRAF-mutant metastatic colorectal cancer. Front. Oncol. 2021, 11, 601722. [Google Scholar] [CrossRef]
  56. Liu, M.; Liu, Q.; Hu, K.; Dong, Y.; Sun, X.; Zou, Z.; Ji, D.; Liu, T.; Yu, Y. Colorectal cancer with BRAF V600E mutation: Trends in immune checkpoint inhibitor treatment. Crit. Rev. Oncol./Hematol. 2024, 204, 104497. [Google Scholar] [CrossRef]
  57. Shan, K.S.; Rehman, T.U.; Ivanov, S.; Domingo, G.; Raez, L.E. Molecular targeting of the BRAF proto-oncogene/mitogen-activated protein kinase (MAPK) pathway across cancers. Int. J. Mol. Sci. 2024, 25, 624. [Google Scholar] [CrossRef]
  58. Rodriquenz, M.G.; Ciardiello, D.; Latiano, T.P.; Maiorano, B.A.; Martinelli, E.; Silvestris, N.; Ciardiello, F.; Maiello, E. Exploring biological heterogeneity and implications on novel treatment paradigm in BRAF-mutant metastatic colorectal cancer. Crit. Rev. Oncol./Hematol. 2022, 173, 103657. [Google Scholar] [CrossRef]
  59. Tabernero, J.; Ros, J.; Élez, E. The Evolving Treatment Landscape in BRAF-V600E-Mutated Metastatic Colorectal Cancer. In American Society of Clinical Oncology Educational Book; American Society of Clinical Oncology: Alexandria, VA, USA, 2022; pp. 1–10. [Google Scholar]
  60. Abushukair, H.M.; Zaitoun, S.M.a.; Saeed, A. Insight on BRAFV600E mutated colorectal cancer immune microenvironment. World J. Gastrointest. Oncol. 2022, 14, 1213. [Google Scholar] [CrossRef]
  61. Li, H.; Zhang, Y.; Xu, Y.; Huang, Z.; Cheng, G.; Xie, M.; Zhou, Z.; Yu, Y.; Xi, W.; Fan, Y. Tumor immune microenvironment and immunotherapy efficacy in BRAF mutation non-small-cell lung cancer. Cell Death Dis. 2022, 13, 1064. [Google Scholar] [CrossRef]
  62. Cho, S.M.; Esmail, A.; Abdelrahim, M. Triple-regimen of vemurafenib, irinotecan, and cetuximab for the treatment of BRAFV600E-mutant CRC: A case report and review. Front. Pharmacol. 2021, 12, 795381. [Google Scholar] [CrossRef] [PubMed]
  63. Ros, J.; Saoudi, N.; Baraibar, I.; Salva, F.; Tabernero, J.; Elez, E. Encorafenib plus cetuximab for the treatment of BRAF-V600E-mutated metastatic colorectal cancer. Ther. Adv. Gastroenterol. 2022, 15, 17562848221110644. [Google Scholar] [CrossRef] [PubMed]
  64. Ott, P.A.; Wu, C.J. Cancer vaccines: Steering T cells down the right path to eradicate tumors. Cancer Discov. 2019, 9, 476–481. [Google Scholar] [CrossRef]
  65. Sotirov, S.; Dimitrov, I. Tumor-derived antigenic peptides as potential cancer vaccines. Int. J. Mol. Sci. 2024, 25, 4934. [Google Scholar] [CrossRef]
  66. Malki, A.; ElRuz, R.A.; Gupta, I.; Allouch, A.; Vranic, S.; Al Moustafa, A.E. Molecular Mechanisms of Colon Cancer Progression and Metastasis: Recent Insights and Advancements. Int. J. Mol. Sci. 2020, 22, 130. [Google Scholar] [CrossRef]
  67. Tejeda-Muñoz, N.; Binder, G.; Mei, K.-C. Emerging therapeutic strategies for Wnt-dependent colon cancer targeting macropinocytosis. Cells Dev. 2024, 180, 203974. [Google Scholar] [CrossRef]
  68. Hankey, W.; Frankel, W.L.; Groden, J. Functions of the APC tumor suppressor protein dependent and independent of canonical WNT signaling: Implications for therapeutic targeting. Cancer Metastasis Rev. 2018, 37, 159–172. [Google Scholar] [CrossRef]
  69. Wang, T.; Fu, J.; Huang, Y.; Fu, C. Mechanism of APC truncation involved in colorectal cancer tumorigenesis. Oncol. Lett. 2024, 29, 2. [Google Scholar] [CrossRef]
  70. Song, P.; Gao, Z.; Bao, Y.; Chen, L.; Huang, Y.; Liu, Y.; Dong, Q.; Wei, X. Wnt/β-catenin signaling pathway in carcinogenesis and cancer therapy. J. Hematol. Oncol. 2024, 17, 46. [Google Scholar] [CrossRef]
  71. Xi, Y.; Cui, W.; Tan, Y.; Zhang, Q.; Duan, Q.; Chen, D.-s. Impact of APC mutations on prognosis and tumor microenvironment in colorectal signet ring cell carcinoma. J. Clin. Oncol. 2024, 42, e15500. [Google Scholar] [CrossRef]
  72. Aghabozorgi, A.S.; Bahreyni, A.; Soleimani, A.; Bahrami, A.; Khazaei, M.; Ferns, G.A.; Avan, A.; Hassanian, S.M. Role of adenomatous polyposis coli (APC) gene mutations in the pathogenesis of colorectal cancer; current status and perspectives. Biochimie 2019, 157, 64–71. [Google Scholar] [CrossRef] [PubMed]
  73. Kelson, C.O.; Zaytseva, Y.Y. Altered lipid metabolism in APC-driven colorectal cancer: The potential for therapeutic intervention. Front. Oncol. 2024, 14, 1343061. [Google Scholar] [CrossRef] [PubMed]
  74. Su, Y.; Sai, Y.; Zhou, L.; Liu, Z.; Du, P.; Wu, J.; Zhang, J. Current insights into the regulation of programmed cell death by TP53 mutation in cancer. Front. Oncol. 2022, 12, 1023427. [Google Scholar] [CrossRef]
  75. Wang, H.; Guo, M.; Wei, H.; Chen, Y. Targeting p53 pathways: Mechanisms, structures and advances in therapy. Signal Transduct. Target. Ther. 2023, 8, 92. [Google Scholar] [CrossRef]
  76. Hernández Borrero, L.J.; El-Deiry, W.S. Tumor suppressor p53: Biology, signaling pathways, and therapeutic targeting. Biochim. Biophys. Acta Rev. Cancer 2021, 1876, 188556. [Google Scholar] [CrossRef]
  77. Lei, L.; Lu, Q.; Ma, G.; Li, T.; Deng, J.; Li, W. P53 protein and the diseases in central nervous system. Front. Genet. 2023, 13, 1051395. [Google Scholar] [CrossRef]
  78. Kim, J.Y.; Jung, J.; Kim, K.M.; Lee, J.; Im, Y.H. TP53 mutations predict poor response to immunotherapy in patients with metastatic solid tumors. Cancer Med. 2023, 12, 12438–12451. [Google Scholar] [CrossRef]
  79. Hassin, O.; Nataraj, N.B.; Shreberk-Shaked, M.; Aylon, Y.; Yaeger, R.; Fontemaggi, G.; Mukherjee, S.; Maddalena, M.; Avioz, A.; Iancu, O.; et al. Different hotspot p53 mutants exert distinct phenotypes and predict outcome of colorectal cancer patients. Nat. Commun. 2022, 13, 2800. [Google Scholar] [CrossRef]
  80. Ho, V.; Chung, L.; Lim, S.H.; Ma, Y.; Wang, B.; Lea, V.; Abubakar, A.; Ng, W.; Lee, M.; Roberts, T.L. Prognostic impact of TP53 mutations and tumor mutational load in colorectal cancer. Gastrointest. Disord. 2022, 4, 165–179. [Google Scholar] [CrossRef]
  81. Zhao, Y.; Li, Y.; Sheng, J.; Wu, F.; Li, K.; Huang, R.; Wang, X.; Jiao, T.; Guan, X.; Lu, Y.; et al. P53-R273H mutation enhances colorectal cancer stemness through regulating specific lncRNAs. J. Exp. Clin. Cancer Res. 2019, 38, 379. [Google Scholar] [CrossRef]
  82. Liu, N.; Jiang, X.; Guo, L.; Zhang, C.; Jiang, M.; Sun, Z.; Zhang, Y.; Mi, W.; Li, J.; Fu, Y. Mutant p53 achieved Gain-of-Function by promoting tumor growth and immune escape through PHLPP2/AKT/PD-L1 pathway. Int. J. Biol. Sci. 2022, 18, 2419. [Google Scholar] [CrossRef] [PubMed]
  83. Li, T.; Liu, T.; Zhu, W.; Xie, S.; Zhao, Z.; Feng, B.; Guo, H.; Yang, R. Targeting MDSC for immune-checkpoint blockade in cancer immunotherapy: Current progress and new prospects. Clin. Med. Insights Oncol. 2021, 15, 11795549211035540. [Google Scholar] [CrossRef] [PubMed]
  84. Makaremi, S.; Asadzadeh, Z.; Hemmat, N.; Baghbanzadeh, A.; Sgambato, A.; Ghorbaninezhad, F.; Safarpour, H.; Argentiero, A.; Brunetti, O.; Bernardini, R. Immune checkpoint inhibitors in colorectal cancer: Challenges and future prospects. Biomedicines 2021, 9, 1075. [Google Scholar] [CrossRef] [PubMed]
  85. Zafari, N.; Khosravi, F.; Rezaee, Z.; Esfandyari, S.; Bahiraei, M.; Bahramy, A.; Ferns, G.A.; Avan, A. The role of the tumor microenvironment in colorectal cancer and the potential therapeutic approaches. J. Clin. Lab. Anal. 2022, 36, e24585. [Google Scholar] [CrossRef]
  86. Li, N.; Zhu, Q.; Tian, Y.; Ahn, K.J.; Wang, X.; Cramer, Z.; Jou, J.; Folkert, I.W.; Yu, P.; Adams-Tzivelekidis, S. Mapping and modeling human colorectal carcinoma interactions with the tumor microenvironment. Nat. Commun. 2023, 14, 7915. [Google Scholar] [CrossRef]
  87. Giraldo, N.A.; Sanchez-Salas, R.; Peske, J.D.; Vano, Y.; Becht, E.; Petitprez, F.; Validire, P.; Ingels, A.; Cathelineau, X.; Fridman, W.H. The clinical role of the TME in solid cancer. Br. J. Cancer 2019, 120, 45–53. [Google Scholar] [CrossRef]
  88. Zhuang, Y.; Wang, H.; Jiang, D.; Li, Y.; Feng, L.; Tian, C.; Pu, M.; Wang, X.; Zhang, J.; Hu, Y. Multi gene mutation signatures in colorectal cancer patients: Predict for the diagnosis, pathological classification, staging and prognosis. BMC Cancer 2021, 21, 380. [Google Scholar] [CrossRef]
  89. Park, K.; Veena, M.S.; Shin, D.S. Key players of the immunosuppressive tumor microenvironment and emerging therapeutic strategies. Front. Cell Dev. Biol. 2022, 10, 830208. [Google Scholar] [CrossRef]
  90. Wang, L.; Zhang, L.; Zhang, Z.; Wu, P.; Zhang, Y.; Chen, X. Advances in targeting tumor microenvironment for immunotherapy. Front. Immunol. 2024, 15, 1472772. [Google Scholar] [CrossRef]
  91. Liu, Y.-T.; Sun, Z.-J. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics 2021, 11, 5365. [Google Scholar] [CrossRef]
  92. Wang, J.X.; Choi, S.Y.; Niu, X.; Kang, N.; Xue, H.; Killam, J.; Wang, Y. Lactic acid and an acidic tumor microenvironment suppress anticancer immunity. Int. J. Mol. Sci. 2020, 21, 8363. [Google Scholar] [CrossRef] [PubMed]
  93. Sieminska, I.; Baran, J. Myeloid-derived suppressor cells in colorectal cancer. Front. Immunol. 2020, 11, 1526. [Google Scholar] [CrossRef]
  94. Sorolla, M.A.; Hidalgo, I.; Sorolla, A.; Montal, R.; Pallisé, O.; Salud, A.; Parisi, E. Microenvironmental reactive oxygen species in colorectal cancer: Involved processes and therapeutic opportunities. Cancers 2021, 13, 5037. [Google Scholar] [CrossRef]
  95. Wang, H.; Tian, T.; Zhang, J. Tumor-associated macrophages (TAMs) in colorectal cancer (CRC): From mechanism to therapy and prognosis. Int. J. Mol. Sci. 2021, 22, 8470. [Google Scholar] [CrossRef]
  96. Janssen, J.B.; Medema, J.P.; Gootjes, E.C.; Tauriello, D.V.; Verheul, H.M. Mutant RAS and the tumor microenvironment as dual therapeutic targets for advanced colorectal cancer. Cancer Treat. Rev. 2022, 109, 102433. [Google Scholar] [CrossRef]
  97. Li, C.; Jiang, P.; Wei, S.; Xu, X.; Wang, J. Regulatory T cells in tumor microenvironment: New mechanisms, potential therapeutic strategies and future prospects. Mol. Cancer 2020, 19, 116. [Google Scholar] [CrossRef]
  98. Kumagai, S.; Momoi, Y.; Nishikawa, H. Immunogenomic cancer evolution: A framework to understand cancer immunosuppression. Sci. Immunol. 2025, 10, eabo5570. [Google Scholar] [CrossRef]
  99. Dzobo, K.; Dandara, C. Architecture of cancer-associated fibroblasts in tumor microenvironment: Mapping their origins, heterogeneity, and role in cancer therapy resistance. Omics A J. Integr. Biol. 2020, 24, 314–339. [Google Scholar] [CrossRef]
  100. Shasha, T.; Gruijs, M.; van Egmond, M. Mechanisms of colorectal liver metastasis development. Cell. Mol. Life Sci. 2022, 79, 607. [Google Scholar] [CrossRef]
  101. Zeng, W.; Liu, H.; Mao, Y.; Jiang, S.; Yi, H.; Zhang, Z.; Wang, M.; Zong, Z. Myeloid-derived suppressor cells: Key immunosuppressive regulators and therapeutic targets in colorectal cancer. Int. J. Oncol. 2024, 65, 154711. [Google Scholar] [CrossRef]
  102. Le, I.; Dhandayuthapani, S.; Chacon, J.; Eiring, A.M.; Gadad, S.S. Harnessing the immune system with cancer vaccines: From prevention to therapeutics. Vaccines 2022, 10, 816. [Google Scholar] [CrossRef] [PubMed]
  103. Ros, J.; Balconi, F.; Baraibar, I.; Saoudi Gonzalez, N.; Salva, F.; Tabernero, J.; Elez, E. Advances in immune checkpoint inhibitor combination strategies for microsatellite stable colorectal cancer. Front. Oncol. 2023, 13, 1112276. [Google Scholar] [CrossRef] [PubMed]
  104. Najafi, S.; Mortezaee, K. Advances in dendritic cell vaccination therapy of cancer. Biomed. Pharmacother. 2023, 164, 114954. [Google Scholar] [CrossRef] [PubMed]
  105. Sobhani, N.; Scaggiante, B.; Morris, R.; Chai, D.; Catalano, M.; Tardiel-Cyril, D.R.; Neeli, P.; Roviello, G.; Mondani, G.; Li, Y. Therapeutic cancer vaccines: From biological mechanisms and engineering to ongoing clinical trials. Cancer Treat. Rev. 2022, 109, 102429. [Google Scholar] [CrossRef]
  106. Yu, J.; Sun, H.; Cao, W.; Song, Y.; Jiang, Z. Research progress on dendritic cell vaccines in cancer immunotherapy. Exp. Hematol. Oncol. 2022, 11, 3. [Google Scholar] [CrossRef]
  107. Filin, I.Y.; Kitaeva, K.V.; Rutland, C.S.; Rizvanov, A.A.; Solovyeva, V.V. Recent advances in experimental dendritic cell vaccines for cancer. Front. Oncol. 2021, 11, 730824. [Google Scholar] [CrossRef]
  108. Lee, K.-W.; Yam, J.W.P.; Mao, X. Dendritic cell vaccines: A shift from conventional approach to new generations. Cells 2023, 12, 2147. [Google Scholar] [CrossRef]
  109. Kim, V.M.; Pan, X.; Soares, K.C.; Azad, N.S.; Ahuja, N.; Gamper, C.J.; Blair, A.B.; Muth, S.; Ding, D.; Ladle, B.H. Neoantigen-based EpiGVAX vaccine initiates antitumor immunity in colorectal cancer. JCI Insight 2020, 5, e136368. [Google Scholar] [CrossRef]
  110. Abd-Aziz, N.; Poh, C.L. Development of peptide-based vaccines for cancer. J. Oncol. 2022, 2022, 9749363. [Google Scholar] [CrossRef]
  111. Buonaguro, L.; Tagliamonte, M. Peptide-based vaccine for cancer therapies. Front. Immunol. 2023, 14, 1210044. [Google Scholar] [CrossRef]
  112. Hamley, I.W. Peptides for vaccine development. ACS Appl. Bio Mater. 2022, 5, 905–944. [Google Scholar] [CrossRef] [PubMed]
  113. Stephens, A.J.; Burgess-Brown, N.A.; Jiang, S. Beyond just peptide antigens: The complex world of peptide-based cancer vaccines. Front. Immunol. 2021, 12, 696791. [Google Scholar] [CrossRef] [PubMed]
  114. O’Reilly, E.M.; Wainberg, Z.A.; Weekes, C.D.; Furqan, M.; Kasi, P.M.; Devoe, C.E.; Leal, A.D.; Chung, V.; Perry, J.; Seenappa, L. AMPLIFY-201, a first-in-human safety and efficacy trial of adjuvant ELI-002 2P immunotherapy for patients with high-relapse risk with KRAS G12D-or G12R-mutated pancreatic and colorectal cancer. J. Clin. Oncol. 2023, 41, 2528. [Google Scholar] [CrossRef]
  115. Pant, S.; Wainberg, Z.A.; Weekes, C.D.; Furqan, M.; Kasi, P.M.; Devoe, C.E.; Leal, A.D.; Chung, V.; Basturk, O.; VanWyk, H. Lymph-node-targeted, mKRAS-specific amphiphile vaccine in pancreatic and colorectal cancer: The phase 1 AMPLIFY-201 trial. Nat. Med. 2024, 30, 531–542. [Google Scholar] [CrossRef]
  116. Haldar, S.D.; Heumann, T.R.; Berg, M.; Ferguson, A.; Lim, S.J.; Wang, H.; Nauroth, J.; Laheru, D.; Jaffee, E.M.; Azad, N.S. A phase I study of a mutant KRAS-targeted long peptide vaccine combined with ipilimumab/nivolumab in resected pancreatic cancer and MMR-proficient metastatic colorectal cancer. J. Clin. Oncol. 2023, 41, TPS814. [Google Scholar] [CrossRef]
  117. Nusrat, M.; Yaeger, R. KRAS inhibition in metastatic colorectal cancer: An update. Curr. Opin. Pharmacol. 2023, 68, 102343. [Google Scholar] [CrossRef]
  118. Hager, S.; Fittler, F.J.; Wagner, E.; Bros, M. Nucleic acid-based approaches for tumor therapy. Cells 2020, 9, 2061. [Google Scholar] [CrossRef]
  119. Liao, H.-C.; Liu, S.-J. Advances in nucleic acid-based cancer vaccines. J. Biomed. Sci. 2025, 32, 10. [Google Scholar] [CrossRef]
  120. Chi, W.-Y.; Hu, Y.; Huang, H.-C.; Kuo, H.-H.; Lin, S.-H.; Kuo, C.-T.J.; Tao, J.; Fan, D.; Huang, Y.-M.; Wu, A.A. Molecular targets and strategies in the development of nucleic acid cancer vaccines: From shared to personalized antigens. J. Biomed. Sci. 2024, 31, 94. [Google Scholar] [CrossRef]
  121. Mollé, L.M.; Smyth, C.H.; Yuen, D.; Johnston, A.P. Nanoparticles for vaccine and gene therapy: Overcoming the barriers to nucleic acid delivery. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2022, 14, e1809. [Google Scholar] [CrossRef]
  122. Zhang, C.; Ma, Y.; Zhang, J.; Kuo, J.C.-T.; Zhang, Z.; Xie, H.; Zhu, J.; Liu, T. Modification of lipid-based nanoparticles: An efficient delivery system for nucleic acid-based immunotherapy. Molecules 2022, 27, 1943. [Google Scholar] [CrossRef] [PubMed]
  123. Fernandez, M.E.; Maurel, J.; Morris, V.; Kopetz, S.; Galligan, B.; Ali, S.; Derhovanessian, E.; Unsal-Kacmaz, K.; Manning, L.; Henn, H. 29P Characterization of T cell responses induced by the individualized mRNA neoantigen vaccine autogene cevumeran in adjuvant stage II (high risk)/stage III colorectal cancer (CRC) patients (pts) from the biomarker cohort of the phase II BNT122-01 trial. Ann. Oncol. 2024, 35, S14–S15. [Google Scholar] [CrossRef]
  124. Shanmugaraj, B.; Priya, L.B.; Mahalakshmi, B.; Subbiah, S.; Hu, R.-M.; Velmurugan, B.K.; Baskaran, R. Bacterial and viral vectors as vaccine delivery vehicles for breast cancer therapy. Life Sci. 2020, 250, 117550. [Google Scholar] [CrossRef] [PubMed]
  125. Travieso, T.; Li, J.; Mahesh, S.; Mello, J.D.F.R.E.; Blasi, M. The use of viral vectors in vaccine development. NPJ Vaccines 2022, 7, 75. [Google Scholar] [CrossRef]
  126. bin Umair, M.; Akusa, F.N.; Kashif, H.; Butt, F.; Azhar, M.; Munir, I.; Ahmed, M.; Khalil, W.; Sharyar, H.; Rafique, S. Viruses as tools in gene therapy, vaccine development, and cancer treatment. Arch. Virol. 2022, 167, 1387–1404. [Google Scholar] [CrossRef]
  127. Bezeljak, U. Cancer gene therapy goes viral: Viral vector platforms come of age. Radiol. Oncol. 2022, 56, 1. [Google Scholar] [CrossRef]
  128. Apolonio, J.S.; de Souza Gonçalves, V.L.; Santos, M.L.C.; Luz, M.S.; Souza, J.V.S.; Pinheiro, S.L.R.; de Souza, W.R.; Loureiro, M.S.; de Melo, F.F. Oncolytic virus therapy in cancer: A current review. World J. Virol. 2021, 10, 229. [Google Scholar] [CrossRef]
  129. Mondal, M.; Guo, J.; He, P.; Zhou, D. Recent advances of oncolytic virus in cancer therapy. Hum. Vaccines Immunother. 2020, 16, 2389–2402. [Google Scholar] [CrossRef]
  130. Hemminki, O.; Dos Santos, J.M.; Hemminki, A. Oncolytic viruses for cancer immunotherapy. J. Hematol. Oncol. 2020, 13, 84. [Google Scholar] [CrossRef]
  131. Monge Bonilla, M.C.; Xie, C.; Steinberg, S.M.; Fioraventi, S.; Walker, M.; Mabry-Hrones, D.; Wood, B.J.; Kleiner, D.E.; Greten, T.F. A phase I/II study of Pexa-Vec oncolytic virus in combination with immune checkpoint inhibition in refractory colorectal cancer. J. Clin. Oncol. 2020, 38, 117. [Google Scholar] [CrossRef]
  132. Monge, C.; Xie, C.; Myojin, Y.; Coffman, K.; Hrones, D.M.; Wang, S.; Hernandez, J.M.; Wood, B.J.; Levy, E.B.; Juburi, I. Phase I/II study of PexaVec in combination with immune checkpoint inhibition in refractory metastatic colorectal cancer. J. Immunother. Cancer 2023, 11, e005640. [Google Scholar] [CrossRef] [PubMed]
  133. Rappaport, A.R.; Kyi, C.; Lane, M.; Hart, M.G.; Johnson, M.L.; Henick, B.S.; Liao, C.-Y.; Mahipal, A.; Shergill, A.; Spira, A.I. A shared neoantigen vaccine combined with immune checkpoint blockade for advanced metastatic solid tumors: Phase 1 trial interim results. Nat. Med. 2024, 30, 1013–1022. [Google Scholar] [CrossRef] [PubMed]
  134. Yarchoan, M.; Huang, C.Y.; Zhu, Q.; Ferguson, A.K.; Durham, J.N.; Anders, R.A.; Thompson, E.D.; Rozich, N.S.; Thomas, D.L.; Nauroth, J.M. A phase 2 study of GVAX colon vaccine with cyclophosphamide and pembrolizumab in patients with mismatch repair proficient advanced colorectal cancer. Cancer Med. 2020, 9, 1485–1494. [Google Scholar] [CrossRef] [PubMed]
  135. Monsen, H.; Norseth, H.M.; Remen, N.; Abrahamsen, I.; Lysén, A.; Schjesvold, F. PB2118: TG01-Study: A phase 1/2 study with TG01 immunotherapy vaccination in patients with confirmed NRAS/KRAS mutation on codon 12/13 and multiple myeloma or high-risk smoldering myeloma. HemaSphere 2023, 7, e51467aa. [Google Scholar] [CrossRef]
  136. Seenappa, L.M.; Jakubowski, A.; Steinbuck, M.P.; Palmer, E.; Haqq, C.M.; Carter, C.; Fontenot, J.; Villinger, F.; McNeil, L.K.; DeMuth, P.C. Amphiphile-CpG vaccination induces potent lymph node activation and COVID-19 immunity in mice and non-human primates. NPJ Vaccines 2022, 7, 128. [Google Scholar] [CrossRef]
  137. Brown, T.A.; Byrd, K.; Vreeland, T.J.; Clifton, G.T.; Jackson, D.O.; Hale, D.F.; Herbert, G.S.; Myers, J.W.; Greene, J.M.; Berry, J.S. Final analysis of a phase I/IIa trial of the folate-binding protein-derived E39 peptide vaccine to prevent recurrence in ovarian and endometrial cancer patients. Cancer Med. 2019, 8, 4678–4687. [Google Scholar] [CrossRef]
  138. Belnoue, E.; Leystra, A.A.; Carboni, S.; Cooper, H.S.; Macedo, R.T.; Harvey, K.N.; Colby, K.B.; Campbell, K.S.; Vanderveer, L.A.; Clapper, M.L. Novel protein-based vaccine against self-antigen reduces the formation of sporadic colon adenomas in mice. Cancers 2021, 13, 845. [Google Scholar] [CrossRef]
  139. Kopetz, S.; Morris, V.K.; Alonso-Orduña, V.; Garcia-Alfonso, P.; Reboredo, M.; Fernandez Montes, A.; Maurel, J.; Paez, D.; Reinacher-Schick, A.C.; Höhler, T. A phase 2 multicenter, open-label, randomized, controlled trial in patients with stage II/III colorectal cancer who are ctDNA positive following resection to compare efficacy of autogene cevumeran versus watchful waiting. J. Clin. Oncol. 2022, 40, TPS3641. [Google Scholar] [CrossRef]
  140. Wei, J.; Hui, A.-M. The paradigm shift in treatment from COVID-19 to oncology with mRNA vaccines. Cancer Treat. Rev. 2022, 107, 102405. [Google Scholar] [CrossRef]
  141. D’Alise, M.; Willis, J.; Leoni, G.; Cruz-Correa, M.; Hall, M.J.; Idos, G.E.; Garzia, I.; Antonucci, L.; Cotugno, G.; Siani, L. 1526 Nous-209 genetic vaccine encoding shared cancer neoantigens is safe and elicits robust immune response in healthy Lynch syndrome carriers: Interim results from Phase 1 cancer interception trial. J. ImmunoTher. Cancer 2023, 11. [Google Scholar] [CrossRef]
  142. Pant, S.; Furqan, M.; Abdul-Karim, R.M.; Chung, V.; Devoe, C.E.; Johnson, M.L.; Leal, A.D.; Park, H.; Wainberg, Z.A.; Welkowsky, E. First-in-human phase 1 trial of ELI-002 immunotherapy as treatment for subjects with Kirsten rat sarcoma (KRAS)-mutated pancreatic ductal adenocarcinoma and other solid tumors. J. Clin. Oncol. 2022, 40, TPS2701. [Google Scholar] [CrossRef]
  143. Jou, J.; Harrington, K.J.; Zocca, M.-B.; Ehrnrooth, E.; Cohen, E.E. The changing landscape of therapeutic cancer vaccines—Novel platforms and neoantigen identification. Clin. Cancer Res. 2021, 27, 689–703. [Google Scholar] [CrossRef] [PubMed]
  144. Overman, M.J.; Maurel, J.; Oberstein, P.E.; Roselló-Keränen, S.; Le, D.T.; Pedersen, K.S.; Mukherjee, S.; D’Alise, A.M.; Leoni, G.; Siani, L. Results of phase I-II bridging study for Nous-209, a neoantigen cancer immunotherapy, in combination with pembrolizumab as first line treatment in patients with advanced dMMR/MSI-h colorectal cancer. J. Clin. Oncol. 2023, 41, e14665. [Google Scholar] [CrossRef]
  145. Formica, V.; Sera, F.; Cremolini, C.; Riondino, S.; Morelli, C.; Arkenau, H.-T.; Roselli, M. KRAS and BRAF mutations in stage II and III colon cancer: A systematic review and meta-analysis. JNCI J. Natl. Cancer Inst. 2022, 114, 517–527. [Google Scholar] [CrossRef]
  146. Testa, U.; Pelosi, E.; Castelli, G. Colorectal cancer: Genetic abnormalities, tumor progression, tumor heterogeneity, clonal evolution and tumor-initiating cells. Med. Sci. 2018, 6, 31. [Google Scholar] [CrossRef]
  147. Quandt, J.; Schlude, C.; Bartoschek, M.; Will, R.; Cid-Arregui, A.; Schölch, S.; Reissfelder, C.; Weitz, J.; Schneider, M.; Wiemann, S. Long-peptide vaccination with driver gene mutations in p53 and Kras induces cancer mutation-specific effector as well as regulatory T cell responses. Oncoimmunology 2018, 7, e1500671. [Google Scholar] [CrossRef]
  148. Zhou, S.; Fan, C.; Zeng, Z.; Young, K.H.; Li, Y. Clinical and immunological effects of p53-targeting vaccines. Front. Cell Dev. Biol. 2021, 9, 762796. [Google Scholar] [CrossRef]
  149. Chiang, Y.-T.; Chien, Y.-C.; Lin, Y.-H.; Wu, H.-H.; Lee, D.-F.; Yu, Y.-L. The function of the mutant p53-R175H in cancer. Cancers 2021, 13, 4088. [Google Scholar] [CrossRef]
  150. Chen, C.; Liu, S.; Qu, R.; Li, B. Recurrent neoantigens in colorectal cancer as potential immunotherapy targets. BioMed Res. Int. 2020, 2020, 2861240. [Google Scholar] [CrossRef]
  151. Wang, Z.; Burigotto, M.; Ghetti, S.; Vaillant, F.; Tan, T.; Capaldo, B.D.; Palmieri, M.; Hirokawa, Y.; Tai, L.; Simpson, D.S. Loss-of-function but not gain-of-function properties of mutant TP53 are critical for the proliferation, survival, and metastasis of a broad range of cancer cells. Cancer Discov. 2024, 14, 362–379. [Google Scholar] [CrossRef]
  152. Madden-Hennessey, K.; Gupta, D.; Radecki, A.A.; Guild, C.; Rath, A.; Heinen, C.D. Loss of mismatch repair promotes a direct selective advantage in human stem cells. Stem Cell Rep. 2022, 17, 2661–2673. [Google Scholar] [CrossRef] [PubMed]
  153. Peng, H.; Ying, J.; Zang, J.; Lu, H.; Zhao, X.; Yang, P.; Wang, X.; Li, J.; Gong, Z.; Zhang, D. Specific mutations in APC, with prognostic implications in metastatic colorectal cancer. Cancer Res. Treat. Off. J. Korean Cancer Assoc. 2023, 55, 1270–1280. [Google Scholar] [CrossRef] [PubMed]
  154. Capietto, A.-H.; Hoshyar, R.; Delamarre, L. Sources of cancer neoantigens beyond single-nucleotide variants. Int. J. Mol. Sci. 2022, 23, 10131. [Google Scholar] [CrossRef]
  155. Corulli, L.R.; Cecil, D.L.; Gad, E.; Koehnlein, M.; Coveler, A.L.; Childs, J.S.; Lubet, R.A.; Disis, M.L. Multi-Epitope-Based vaccines for colon cancer treatment and prevention. Front. Immunol. 2021, 12, 729809. [Google Scholar] [CrossRef]
  156. Zhang, D.; Ni, Q.-Q.; Liang, Q.-Y.; He, L.-L.; Qiu, B.-W.; Zhang, L.-J.; Mou, T.-Y.; Le, C.-C.; Huang, Y.; Li, T.-T. ASCL2 induces an immune excluded microenvironment by activating cancer-associated fibroblasts in microsatellite stable colorectal cancer. Oncogene 2023, 42, 2841–2853. [Google Scholar] [CrossRef]
  157. Mestrallet, G.; Brown, M.; Bozkus, C.C.; Bhardwaj, N. Immune escape and resistance to immunotherapy in mismatch repair deficient tumors. Front. Immunol. 2023, 14, 1210164. [Google Scholar] [CrossRef]
  158. Li, X.; You, J.; Hong, L.; Liu, W.; Guo, P.; Hao, X. Neoantigen cancer vaccines: A new star on the horizon. Cancer Biol. Med. 2024, 21, 274–311. [Google Scholar] [CrossRef]
  159. Fan, T.; Zhang, M.; Yang, J.; Zhu, Z.; Cao, W.; Dong, C. Therapeutic cancer vaccines: Advancements, challenges and prospects. Signal Transduct. Target. Ther. 2023, 8, 450. [Google Scholar] [CrossRef]
  160. Yu, Y.J.; Shan, N.; Li, L.Y.; Zhu, Y.S.; Lin, L.M.; Mao, C.C.; Hu, T.T.; Xue, X.Y.; Su, X.P.; Shen, X.; et al. Preliminary clinical study of personalized neoantigen vaccine therapy for microsatellite stability (MSS)-advanced colorectal cancer. Cancer Immunol. Immunother. 2023, 72, 2045–2056. [Google Scholar] [CrossRef]
  161. 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. [Google Scholar] [CrossRef]
  162. Hundal, J.; Kiwala, S.; McMichael, J.; Miller, C.A.; Xia, H.; Wollam, A.T.; Liu, C.J.; Zhao, S.; Feng, Y.-Y.; Graubert, A.P. pVACtools: A computational toolkit to identify and visualize cancer neoantigens. Cancer Immunol. Res. 2020, 8, 409–420. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Signaling pathways of signature gene mutations in CRC. It illustrates the molecular interactions involving signature gene mutations, including MMR genes, KRAS, BRAF, APC, and TP53. Mutant KRAS continuously activate downstream signaling pathways such as PI3K, AKT, and mTORC1, thereby promoting CRC cell survival and proliferation. BRAF activates MEK and ERK driving tumor cell proliferation and immune modulation. Abnormal APC fails to properly degrade β-catenin, leading to its accumulation. This activates the transcription of Wnt target genes involved in cell proliferation, survival, and metastasis, thereby promoting an immunosuppressive TME. Mutant p53 also facilitates cell proliferation and deregulated cell cycle progression. Moreover, they downregulate MHC class I expression, increase immunosuppressive cytokine production, and enhance immune evasion by upregulating PD-L1 and stimulating CAFs. CRC, colorectal cancer; CAFs, cancer-associated fibroblasts; MHC, major histocompatibility complex; PD-L1, programmed cell death-ligand1; dMMR, mismatch repair deficiency; GSK-3β, glycogen synthase kinase-3β; CK I α, casein kinase I alpha; APC, adenomatous polyposis coli; MSI, microsatellite instability; KRAS, Kirsten rat sarcoma virus; PI3K, phosphatidylinositol 3-kinase; AKT, protein kinase B; mTORC1, mammalian target of rapamycin complex1; MEK, mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase.
Figure 1. Signaling pathways of signature gene mutations in CRC. It illustrates the molecular interactions involving signature gene mutations, including MMR genes, KRAS, BRAF, APC, and TP53. Mutant KRAS continuously activate downstream signaling pathways such as PI3K, AKT, and mTORC1, thereby promoting CRC cell survival and proliferation. BRAF activates MEK and ERK driving tumor cell proliferation and immune modulation. Abnormal APC fails to properly degrade β-catenin, leading to its accumulation. This activates the transcription of Wnt target genes involved in cell proliferation, survival, and metastasis, thereby promoting an immunosuppressive TME. Mutant p53 also facilitates cell proliferation and deregulated cell cycle progression. Moreover, they downregulate MHC class I expression, increase immunosuppressive cytokine production, and enhance immune evasion by upregulating PD-L1 and stimulating CAFs. CRC, colorectal cancer; CAFs, cancer-associated fibroblasts; MHC, major histocompatibility complex; PD-L1, programmed cell death-ligand1; dMMR, mismatch repair deficiency; GSK-3β, glycogen synthase kinase-3β; CK I α, casein kinase I alpha; APC, adenomatous polyposis coli; MSI, microsatellite instability; KRAS, Kirsten rat sarcoma virus; PI3K, phosphatidylinositol 3-kinase; AKT, protein kinase B; mTORC1, mammalian target of rapamycin complex1; MEK, mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase.
Ijms 26 04559 g001
Figure 2. Differences in immune microenvironment between “cold and hot” tumors in CRC. The TME is composed of a complex structure with tightly interconnected cellular components—such as CAFs, Tregs, APCs, NK cells, and MDSCs—and non-cellular components. Hot tumors are characterized by high levels of infiltration by CD8+ T cells, NK cells, and APCs, resulting in an inflammatory phenotype. They secrete pro-inflammatory cytokines, such as IL-2, IFN-γ, and cancer cells present relatively high MHC-I. In contrast, cold tumors include immunosuppressive mediators, such as MDSCs, Tregs, TAMs, and CAFs, leading to a non-inflammatory phenotype. They suppress presenting MHC-I, and secrete anti-inflammatory cytokines, such as TGF-β, IL-10, and IL-35. TCR, T cell receptor; MHC, major histocompatibility complex; TGF, transforming growth factor; IL, interleukin; IFN, interferon; CAF, cancer-associated fibroblast; Treg, regulatory T cell; APC, antigen-presenting cell; NK cell, natural killer cell; MDSC, myeloid-derived suppressor cell; TAMs, tumor-associated macrophage.
Figure 2. Differences in immune microenvironment between “cold and hot” tumors in CRC. The TME is composed of a complex structure with tightly interconnected cellular components—such as CAFs, Tregs, APCs, NK cells, and MDSCs—and non-cellular components. Hot tumors are characterized by high levels of infiltration by CD8+ T cells, NK cells, and APCs, resulting in an inflammatory phenotype. They secrete pro-inflammatory cytokines, such as IL-2, IFN-γ, and cancer cells present relatively high MHC-I. In contrast, cold tumors include immunosuppressive mediators, such as MDSCs, Tregs, TAMs, and CAFs, leading to a non-inflammatory phenotype. They suppress presenting MHC-I, and secrete anti-inflammatory cytokines, such as TGF-β, IL-10, and IL-35. TCR, T cell receptor; MHC, major histocompatibility complex; TGF, transforming growth factor; IL, interleukin; IFN, interferon; CAF, cancer-associated fibroblast; Treg, regulatory T cell; APC, antigen-presenting cell; NK cell, natural killer cell; MDSC, myeloid-derived suppressor cell; TAMs, tumor-associated macrophage.
Ijms 26 04559 g002
Table 1. Cancer vaccines targeting signature genes in CRC.
Table 1. Cancer vaccines targeting signature genes in CRC.
Drug/FormulationRelated
Signature Genes
MechanismClinical Trial
Phase
Combination/
Adjuvant
Reference
GVAX/Cell-basedKRASSecretes GM-CSF to activate DCIIPembrolizumab[134]
ELI-002/Lipopeptide-basedKRASAMP-CpG activates immune system, AMP-mKRAS peptide targets KRASIb-[114,142]
-/Long peptide-basedKRASIncreases diversity of T-cell responses by long peptideINivolumab,
Ipilimumab
[116]
TG01/Peptide-basedKRASEnhances DTH response and T cell proliferationI/IIQS-21[135]
E-39/Peptide-basedNRAS/KRASTALs from E39-stimulated induce cytolysis in tumorI/II-[137]
KISIMA-Ascl2 vaccine/Peptide-basedAPCUtilizing Ascl2 upregulated in ISCs with KISIMA vaccine platform-Anti-PD-1/AS15[138]
BNT122/mRNA-basedKRAS/BRAFUpregulates tumor-specific cytotoxic immune responseIIAtezolizumab[139]
mRNA5681(V941)/
mRNA-based
KRASAdministered mRNA induces antigen expressionIIPembrolizumab[140]
GRT-R904/saRNA-based, Vector-basedKRASPrime-boost strategy with ChAdV and saRNA-LNP combinedI/IIKeytruda,
Pembrolizumab
[133,143]
Pexa-Vec(JX-594)/
Vector-based
KRAS/TP53Oncolytic virus-based that directly destroys tumors and promotes immune responseI/II-[131,132]
Nous-209/Vector-basedMMRPrime-boost strategy with GAd20-209-FSP eliciting robust T cell responseIb/IIPembrolizumab[141,144]
DC, dendritic cell; GM-CSF, granulocyte-macrophage colony-stimulating factor; DTH, delayed-type hypersensitivity; TAL, tumor-associated lymphocyte; ISC, intestinal stem cell; saRNA, self-amplifying RNA; ChAdV, chimpanzee adenovirus; GAd, gorilla adenovirus; FSP, frameshift peptide.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yoon, J.; Moon, H.; Jeon, Y.; Choe, S.; Yoon, H. Signature Gene Mutations in Colorectal Cancer: Potential Neoantigens for Cancer Vaccines. Int. J. Mol. Sci. 2025, 26, 4559. https://doi.org/10.3390/ijms26104559

AMA Style

Yoon J, Moon H, Jeon Y, Choe S, Yoon H. Signature Gene Mutations in Colorectal Cancer: Potential Neoantigens for Cancer Vaccines. International Journal of Molecular Sciences. 2025; 26(10):4559. https://doi.org/10.3390/ijms26104559

Chicago/Turabian Style

Yoon, Jaegoo, Haeun Moon, Yuna Jeon, Soohyun Choe, and Hyunho Yoon. 2025. "Signature Gene Mutations in Colorectal Cancer: Potential Neoantigens for Cancer Vaccines" International Journal of Molecular Sciences 26, no. 10: 4559. https://doi.org/10.3390/ijms26104559

APA Style

Yoon, J., Moon, H., Jeon, Y., Choe, S., & Yoon, H. (2025). Signature Gene Mutations in Colorectal Cancer: Potential Neoantigens for Cancer Vaccines. International Journal of Molecular Sciences, 26(10), 4559. https://doi.org/10.3390/ijms26104559

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop