Next Article in Journal
The Molecular Mechanisms Underlying Zucchini-Induced Changes in the Host Adaptation of Cotton- and Cucumber-Type Aphis gossypii
Previous Article in Journal
CLSI Validation of Exchangeable Copper Determination in Serum by ICP-MS: A Focus on Alzheimer’s Disease and Wilson Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Alternative Splicing in Tumorigenesis and Cancer Therapy

1
Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha 410013, China
2
Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
3
NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China
4
Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha 410013, China
*
Authors to whom correspondence should be addressed.
Biomolecules 2025, 15(6), 789; https://doi.org/10.3390/biom15060789
Submission received: 26 March 2025 / Revised: 21 April 2025 / Accepted: 1 May 2025 / Published: 29 May 2025
(This article belongs to the Section Molecular Genetics)

Abstract

:
Alternative splicing (AS) is a pivotal post-transcriptional mechanism that expands the functional diversity of the proteome by enabling a single gene to generate multiple mRNA and protein isoforms. This process, which involves the differential inclusion or exclusion of exons and introns, is tightly regulated by splicing factors (SFs), such as serine/arginine-rich proteins (SRs), heterogeneous nuclear ribonucleoproteins (hnRNPs), and RNA-binding motif (RBM) proteins. These factors recognize specific sequences, including 5′ and 3′ splice sites and branch points, to ensure precise splicing. While AS is essential for normal cellular function, its dysregulation is increasingly implicated in cancer pathogenesis. Aberrant splicing can lead to the production of oncogenic isoforms that promote tumorigenesis, metastasis, and resistance to therapy. Furthermore, such abnormalities can cause the loss of tumor-suppressing activity, thereby contributing to cancer development. Importantly, abnormal AS events can generate neoantigens, which are presented on tumor cell surfaces via major histocompatibility complex (MHC) molecules, suggesting novel targets for cancer immunotherapy. Additionally, splice-switching oligonucleotides (SSOs) have shown promise as therapeutic agents because they modulate splicing patterns to restore normal gene function or induce tumor-suppressive isoforms. This review explores the mechanisms of AS dysregulation in cancer, its role in tumor progression, and its potential as a therapeutic target. We also discuss innovative technologies, such as high-throughput sequencing and computational approaches, that are revolutionizing the study of AS in cancer. Finally, we address the challenges and future prospects of targeting AS for personalized cancer therapies, emphasizing its potential in precision medicine.

1. Introduction

AS is a cellular process that enables a single gene to generate multiple mRNA and protein isoforms by joining exons in different combinations. AS is a crucial mechanism for gene product diversity, leading to diverse protein isoforms with unique functions [1,2]. This approach provides significant evolutionary advantages by expanding the functional repertoire of genes. There are five primary modes of AS: exon skipping, alternative 5′ splice sites, alternative 3′ splice sites, intron retention and mutually exclusive exons [3,4,5,6,7,8]. AS events are regulated by a group of SFs, including SRs, hnRNPs, and RBM proteins [9,10,11,12]. These SFs recognize and bind to specific sequences, such as the 5′ splice site, 3′ splice site, and branch point, as well as exonic and intronic splicing enhancers (ESEs/ISEs) and silencers (ESSs/ISSs), to ensure precise and efficient splicing [13,14].
Abnormal AS can serve as a biomarker and therapeutic target for cancer. Dysregulation of AS can lead to the production of oncogenic splice variants that promote tumor growth, metastasis, and therapy resistance. Additionally, such abnormalities can cause the loss of tumor-suppressing activity, thereby contributing to cancer development. The identification of neoantigens resulting from abnormal AS events in cancer cells provides new therapeutic targets [1,15,16]. These neoantigens can be presented by MHC molecules on the surface of tumor cells, triggering immune responses and offering new avenues for immunotherapy. Additionally, splice-switching oligonucleotides (SSOs) have emerged as powerful tools to correct aberrant splicing or induce the expression of therapeutic splice variants [17]. By targeting specific splicing variants, SSOs can restore normal gene function or increase the production of tumor-suppressive isoforms. In this review, we explore the role of AS in cancer biology and therapy, focusing on the mechanisms underlying aberrant splicing, its impact on tumor progression, and its potential as a therapeutic target. We also discuss innovative technologies, such as high-throughput sequencing and computational tools, that are advancing our understanding of AS in cancer. Finally, we highlight the challenges and future directions in targeting AS for cancer treatment, emphasizing the potential of personalized therapies on the basis of splicing events.

2. Normal and Abnormal Alternative Splicing

AS is a normal phenomenon in eukaryotes. Approximately 95% of multiexonic genes are alternatively spliced to produce alternative transcript products from the same gene. However, the functional significance of these isoforms remains a subject of debate. While some scientists argue that most splice variants are due to splicing errors and lack functional relevance, others believe that some isoforms play critical roles in cellular processes. Studies indicate that fewer than 10% of genes produce functionally distinct splice isoforms (FDSIs) [18], with more than 90% of detected splice variants absent at the protein level [19]. Unproductive AS generally introduces premature termination codons and undergoes rapid nonsense-mediated decay (NMD) [20]. FDSIs are defined as those in which at least two variants are necessary for the gene’s normal function. Phenotypic analyses, including RNAi knockdown and isoform-specific rescue experiments, have identified only a limited number of genes with functionally distinct isoforms. In cases where the depletion of one splice isoform of a gene causes a phenotype, the depletion of the remaining splice isoforms of the same gene does not generate the same phenotype [18]. Notably, some studies have shown that different splice isoforms can rescue the same phenotype, suggesting functional redundancy. Fewer than 10% of human protein isoforms in UniProt have experimentally validated functional annotations [18]. Most alternatively spliced isoforms are expressed at low levels and lack cross-species conservation, suggesting that the majority of isoforms are nonfunctional transcripts resulting from mis-splicing [20]. There are several examples of normal AS, where specific isoforms play essential roles in regulating cellular functions and maintaining physiological homeostasis [21,22,23,24,25,26,27,28,29,30]. A notable example is the TP53, a crucial tumor suppressor gene, which produces multiple isoforms through alternative splicing, including full-length p53α, as well as Δ40p53, p53β, and p53γ. These isoforms have distinct roles in regulating genomic stability, apoptosis, tissue repair, and development. These isoforms are vital for both normal physiological processes and pathological states, such as cancer, by maintaining cellular and tissue balance.

3. Abnormal Alternative Splicing and Cancer

While normal AS plays a role in physiological processes, aberrant splicing is a hallmark of many diseases, including cancer and genetic disorders [31,32,33]. In cancers, splicing abnormalities are particularly prevalent, with tumors exhibiting up to 30% more AS events than normal samples [34]. These aberrant splicing events can generate cancer-specific isoforms that confer growth benefits and promote tumorigenesis [35]. Chemoresistant cancer cells often show a reduction in AS events, which may increase their survival under treatment [36]. Mutations that disrupt splicing regulation or cause reading frameshifts can lead to the production of dysfunctional proteins. Aberrant AS in cancer mainly arises from mutations in cis-acting regulatory elements, trans-acting SFs, and small nuclear RNAs (snRNAs), as well as the dysregulated expression of SFs [37].

3.1. Cis-Acting Splicing Mutations

A splice-site mutation refers to a genetic alteration in the DNA sequence that occurs at the junction of an exon and an intron (splice site). This change can disrupt normal RNA splicing, resulting in aberrant splicing patterns and an altered protein-coding sequence. Cis splicing mutations, which account for 15–60% of human disease-causing mutations, often disrupt splice site signals or splicing enhancer/silencer elements within pre-mRNAs. This disruption can lead to the production of aberrant mRNA and protein products [38]. Recurrent somatic point mutations near splicing sites can facilitate or inhibit specific splicing changes, resulting in erroneous splicing of cancer-associated genes and the generation of new splice isoforms [39,40,41]. Many driver mutations are thought to promote cancer through aberrant splicing. A notable example is mutations in the splice junctions of the MET gene, which lead to the skipping of exon 14. This AS results in a truncated MET protein that exhibits oncogenic activity and has clinical significance in lung adenocarcinoma (LUAD) [42]. This could serve as a druggable target, and detailed content will be discussed in the section “Targeting Novel Splice Variants”. A statistical framework for the full landscape of splice-altering variants (SAVs) was applied to whole exome and transcriptome sequencing data from 8976 cancer samples, systematically identifying 14,438 SAVs, approximately 50% of which disrupt or create splice sites. This study revealed the genomic features of SAVs, potential mutational processes, and their impact on cancer driver genes, including TP53, PIK3R1, GATA3, and CDKN2A. For example, TP53 splice site mutations often result in exon skipping or intron retention, resulting in the production of a nonfunctional p53 protein that fails to suppress tumor growth. Similarly, PIK3R1 mutations can dysregulate the PI3K signaling pathway, promoting cell survival and proliferation, while CDKN2A mutations can lead to loss of p16 function, disrupting cell cycle control [43].

3.2. Chromatin State

Splicing often occurs cotranscriptionally, suggesting that the chromatin state affects AS [44]. Nucleosomes localize to exons, with histone modifications such as H3K36me3 and H3K9me3, along with DNA methylation, forming a chromatin landscape for exon recognition and splicing. RNA-guided mechanisms, via small noncoding RNAs, influence splicing by recruiting histone-modifying enzymes. The kinetic coupling model shows that the transcription elongation rate impacts splicing. Slower rates facilitate weak splice site recognition and promote alternative exon inclusion. Histone methylation and acetylation modulate RNA polymerase II movement, affecting elongation and splicing [45]. For example, H3K9ac is related to fast elongation, whereas H3K36me3 is linked to slower elongation and more exon inclusion [46]. In splicing regulation, H3K36me3, which is abundant in exons, recruits SFs such as PTB and MRG15 to promote exon inclusion [47], whereas H3K9me3, a heterochromatin marker, is associated with exon skipping. DNA methylation near splice sites, regulated by factors like MeCP2, also influences splicing [48]. In colorectal cancer (CRC), the presence of H3K9me3 suppresses the transcription elongation rate of RNA polymerase II, which induces increased skipping of the CD44 exon, contributing to a partial epithelial-to-mesenchymal transition (EMT). This alteration in splicing promotes the invasion and metastasis of cancer cells in CRC, highlighting the role of chromatin modification in cancer progression [49].

3.3. RNA Structure

The structures of pre-mRNAs can influence AS by altering the function of splicing regulatory elements and proteins [50]. Secondary structures, such as hairpins, can either mask or expose splice sites, affecting the binding of SFs and small nuclear ribonucleoproteins (snRNPs). Stable structures near splice sites can prevent spliceosome recognition, causing exon skipping or intron retention. Conversely, the unfolding of these structures can enhance splice site recognition and promote exon inclusion, often facilitated by RNA-binding proteins (RBPs). Epigenetic RNA modifications, particularly N6-methyladenosine (m6A), play an important role in modulating AS. m6A, a prevalent RNA modifications, is found abundantly in both exons and introns. m6A-modified exons have a higher likelihood of being retained in mature mRNAs [51]. For instance, in oesophageal carcinoma [52], a negative correlation has been reported between m6A patterns and AS features in individual patients. The “m6A writer” complex, with METTL3 as the core enzyme, adds m6A to target RNAs, influencing AS in many genes, including MDM4, MDM2, FAS, BAX, and VEGFA [51,53]. In turn, AS can affect m6A deposition or recognition on mRNAs [51]. This dynamic relationship between AS and m6A deposition suggests that alterations in RNA structure and modifications can have profound implications for cancer biology.

3.4. Trans-Acting Splicing Mutations

Trans-acting splicing regulators such as SFs and RBPs are crucial in cancer splicing [38,54]. SF mutations are common and can cause mistakes in tumor suppressor gene mRNA splicing. For example, in pancreatic ductal adenocarcinoma (PDAC) [55] and lymphoid leukemia [56], SF3B1 (a U2 snRNP core) missense mutations lead to 3′ splice site recognition errors and abnormal splicing of tumor suppressor genes [57,58]. In sonic hedgehog medulloblastoma, U1 snRNP mutations disrupt mRNA splicing, inactivating genes such as PTCH1 and activating GLI2 [59,60,61,62,63]. An analysis of 32 cancer types revealed many splicing events linked to SF mutations, such as those in SF3B1 and U2AF1 [34]. The RBM family, which is important for alternative splicing regulation, is also key in cancers. RBM dysregulation from mutations or altered expression can produce abnormal splicing isoforms that drive tumor growth. In non-small cell lung cancer (NSCLC) [7], RBM4 exon 3 skipping changes RBM4-FL to RBM4-S. RBM4-FL inhibits the SRSF1-mTORC1 pathway, but RBM4-S does not, making the pathway hyperactive and promoting NSCLC cell growth [2]. In colorectal cancer (CRC), the regulatory role of RBM39 in splicing changes CDK5RAP2 from its long isoform to its short isoform, promoting CRC development [8,15].

3.5. Abnormal AS and NMD Regulation

NMD is a key cellular surveillance mechanism that degrades aberrant mRNAs containing premature termination codons (PTCs), which often arise from unproductive AS events [64]. Abnormal AS products can evade NMD through various mechanisms, such as altering splice sites, modulating mRNA stability, changing RNA secondary structures, or influencing interactions with RBPs [65]. These evasion strategies can lead to the accumulation of dysfunctional or truncated proteins, contributing to disease pathogenesis, including cancer pathogenesis.
Various cellular stresses, including amino acid deprivation, hypoxia, nutrient deprivation, infection, reactive oxygen species (ROS), and double-stranded RNA, can inhibit NMD [66]. The stress-induced inhibition of NMD is primarily mediated by the phosphorylation of eukaryotic initiation factor 2α (eIF2α), a key regulatory step in the integrated stress response (ISR) [67]. Phosphorylated eIF2α reduces global translation while selectively enhancing the expression of stress-responsive genes, such as ATF4, which plays a crucial role in cellular adaptation to stress. By dynamically regulating NMD, tumors can adapt to the tumor microenvironment (TME), such as hypoxia and nutrient deprivation, thereby promoting survival, proliferation, and metastasis. The inhibition of NMD in the TME may dynamically regulate the expression of key genes involved in tumorigenesis and cellular stress responses.
Under certain circumstances, activated NMD may paradoxically promote cancer progression. In myelodysplastic syndromes (MDS) [68], mutations in the SRSF2 induce aberrant recognition of specific exons, generating splice isoforms containing PTCs. These defective transcripts are targeted by NMD, leading to significant downregulation of critical genes EZH2 and INTS3. This NMD-driven depletion synergizes with RAS pathway activation, collectively contributing to the malignant transformation of MDS into acute leukemia.

3.6. Abberant Splicing in Breast Cancer

Aberrant splicing of genomic loci, including those encoding estrogen receptors (ERs), HER2/neu, Cyclin D1, BRCA1, BARD1, Tenscin-C, and CD44, has been implicated in breast carcinogenesis [69]. The ER gene produces multiple splice variants, including ERα66, ERα36 and ERα46. Among these, ERα36, a splice variant from the ESR1 locus, plays an important role in governing nongenomic membrane signaling pathways triggered by estrogen and confers 4-hydroxytamoxifen resistance in breast cancer therapy [69]. High levels of ERα36 have been associated with reduced benefits from endocrine therapy, indicating its role in mediating tamoxifen resistance. The balance between ERα66 and ERα36 impacts the antitumorigenic effects of vitamin D3 and its metabolites.
HER2, a known driver of breast cancer, undergoes AS resulting in variants like HER2 lacking exon 20 (Δ16HER2) with increased transforming ability compared to the wild-type HER2. This alteration can mimic phenotypes observed in endocrine therapy-resistant breast cancer cases.
The CD44 gene produces multiple splice variants, including the standard isoform (CD44s) and the variable isoform (CD44v), each involved in essential roles in breast cancer development. The CD44v subpopulation in 4T1 breast cancer cells exhibits increased metastatic potential through the expansion of stem-like cancer cells [70]. However, some studies indicate an association of CD44v with the luminal A subtype of breast cancer, which generally has a better prognosis [69], while the CD44 variant v3–10 has shown superior effectiveness compared to the standard isoform in slowing tumor growth and metastasis [71]. A chimeric monoclonal antibody that recognizes the CD44v6 isoform has demonstrated potential for radioimmunotherapy [72]. In clinical trials, this antibody labeled with 186Re could detect up to 66% of breast cancer lesions [73], underscoring its diagnostic and therapeutic potential.

3.7. Abberant Splicing in Colorectal Cancer

The KRAS gene is one of the most commonly mutated genes in human cancers, particularly in CRC, LUAD, and PDAC [74]. Despite its significant role in tumorigenesis, therapeutic strategies specifically targeting mutated KRAS are currently lacking. The functions and cell type-specific expression of the two known proteins produced by the KRAS locus have been described in normal tissues and during tumorigenesis. The human KRAS locus produces two distinct protein isoforms, KRAS4A and KRAS4B, through AS [75]. The less common KRAS4A isoform is prominently present in cancer stem-like cells and is responsive to hypoxic conditions, while the prevalent KRAS4B isoform is upregulated in response to endoplasmic reticulum (ER) stress [76]. Notably, the deletion of KRAS4A has been shown to suppress cancer stem cells. The abundance of the minor KRAS4A isoform in human tumors may serve as a biomarker for sensitivity to specific cancer treatment [76]. The primary developmental functions of KRAS are mediated through the KRAS4B isoform, whereas KRAS4A plays a critical role in cancer progression, possibly through effects on a minor population of stem cells [76]. Targeting RBM39/DCAF15, a key regulator of KRAS mRNA splicing, has demonstrated promising potential in inhibiting cancer stem cells [76].

3.8. Abberant Splicing in Lung Cancer

Aberrant splicing of key genes, including EGFR, KRAS, TP53, BCL2 and MET, plays a significant role in the pathogenesis of lung cancer, impacting tumor behavior and treatment responses.
The T790M mutation in EGFR, known as the “gatekeeper” mutation, confers resistance to first- and second-generation EGFR tyrosine kinase inhibitors (TKIs) by impeding drug binding to the ATP cleft [77]. Additionally, AS variants of EGFR have been identified in tumor cells and are being explored as potential targets for T-cell receptor (TCR)-based therapies [78]. HER2 amplification has been identified as a driver of first- and second-generation EGFR TKI resistance in tumors lacking T790M EGFR mutations [79]. The exon 16-skipping splice variant of HER2, known as HER2D16, has been recognized as a mediator of osimertinib resistance in patients with metastatic EGFR-mutant NSCLC.
TP53, a frequently mutated tumor suppressor gene in lung cancer, undergoes AS resulting in various isoforms. These isoforms include dominant-negative variants that exhibit contrasting functions to p53WT, as well as isoforms with distinct functions and regulatory roles in cellular processes. The presence of specific TP53 isoforms can influence tumor behavior, the response to therapy, and patient prognosis [80]. The activation of full-length p53 typically results in cellular apoptosis, whereas the induction of the AS isoform, the beta isoform of p53 (p53β), results in cellular senescence [81]. NSCLC cells expressing the Δ40p53 and Δ133p53 isoforms and treated with cisplatin exhibits enhanced apoptosis [82], and the Δ133p53 isoform can be a novel transcriptional enhancer of T-cell effector function to improve T-cell-based cancer immunotherapy [83].
Bcl-X, a critical apoptotic gene of BCL2 function, produces two antagonistic isoform splice variants, Bcl-Xl and Bcl-Xs, through AS. The expression of these variants can contribute to resistance against chemotherapeutic agents, highlighting the importance of splicing in therapeutic responses. Increased expression of Bcl-xL after irradiation promotes the malignant actions of lung cancer cells [84].
MET exon 14 skipping mutations occur in approximately 3–5% of NSCLCs, resulting in truncated MET receptor lacking the juxtamembrane regulatory domain, leading to abnormal MET signaling and oncogenesis. MET exon 14 skipping mutations represent a targetable alteration [85].

3.9. Other Malignancies

The BCL-2 gene, a critical regulator of apoptosis, initially identified for its role in B-cell lymphoma, produces two major isoforms through AS: BCL-2α and BCL-2β. BCL-2α is well known for its antiapoptotic properties, while the less characterized BCL-2β lacks exon 3, resulting in the absence of a transmembrane-anchoring domain [86]. Despite the difference, BCL-2β retains the same BH domains and general structure found in BCL-2α. BCL-2β features a unique 9-amino acid stretch at its C-terminal domain, distinguishing it from its α counterpart.
In acute myeloid leukemia (AML), mutations in the FLT3 gene often lead to aberrant splicing, generating constitutively active forms of the FLT3 receptor tyrosine kinase. These splice variants drive the uncontrolled proliferation and survival of leukemic cells. Particularly, internal tandem duplications (FLT3-ITDs) of FLT3 mutations are associated with poor prognosis and resistance to conventional therapies. Targeting FLT3 splice variants or their downstream signaling pathways emerges as a promising therapeutic strategy in AML [87].
In glioblastoma (GBM), the EGFRvIII mutation is a well-known driver of tumorigenesis. This mutation results in the deletion of exons 2–7, producing a constitutively active EGFR variant that promotes tumor growth and survival [88]. Recent studies have characterized the genome-wide AS induced by EGFRvIII in GBM, revealing its impact on tumor cell metabolism and transformation. EGFRvIII upregulates hnRNP A1 in SFs, which plays a key role in AS. hnRNP A1 promotes splicing of the Max transcript, generating a truncated isoform called Delta Max. Delta Max, unlike the full-length Max protein, enhances Myc-dependent transformation and metabolic reprogramming in GBM cells. Specifically, Delta Max rescues Myc-dependent glycolytic gene expression even after the loss of EGFRvIII, highlighting its role in sustaining tumor cell metabolism [89]. Table 1 summarizes the key abnormal AS of genes across the aforementioned cancer types.

3.10. AS and Cancer Immunotherapy

3.10.1. AS and Immune Activation

Immune therapy, including immune checkpoint blockade (ICB), adoptive cell therapy (ACT), and oncolytic virus therapy, has revolutionized the treatment landscape of various malignant tumors. T cell-based immunotherapies show promise in treating cancer by targeting cancer-specific antigens, but are limited in tumors with low mutations and high heterogeneity. Identification of novel tumor-wide neoantigens from RNA splicing erros, such as GNAS and RPL22, enhances T cell therapy by recognizing and eradicating cancer cells across various tumor types [106]. Neoantigens, recognized as nonself antigens newly formed by tumor cells due to various tumor-specific alterations such as genomic mutations, dysregulated RNA splicing, disordered post-translational modifications, and integrated viral ORFs, hold significant therapeutic potential [37]. Tumor cells undergo numerous AS events, resulting in the production of unique splicing isoforms absent in normal cells [107,108]. These splice isoforms can be translated into protein variants containing new epitopes [109,110,111,112], which act as neoantigens recognized by MHC molecules, particularly MHC I, and presented to CD8+ T cells, triggering an immune response [113,114,115]. The tumor mutational burden (TMB), representing the number of somatic nonsynonymous mutations per megabase in tumor cells, is a significant indicator for predicting tumor immunogenicity [116,117,118]. Higher TMBs are significantly associated with better survival after anti-PD-1 therapy [118]. Typically, a high TMB indicates that tumor cells generate more neoantigens, which may correlate with a enhanced response to immune checkpoint inhibitor (ICI) therapy [119].
However, in tumors with a low TMB, AS-derived neoantigens provide a rich source of immunogenic targets, often more abundant than mutation-derived neoantigens [120,121,122,123,124,125,126]. A comprehensive study across 32 different cancer types identified approximately 1.7 neoepitope junctions (NJs) and approximately 0.6 single-nucleotide variant (SNV)-derived peptides per tumor sample predicted to bind to MHC I molecules, making them potential neoantigens [34]. The ability of AS-derived neoantigens to elicit immune responses highlights their potential as targets for immunotherapy, particularly in cancers with low TMB. Exploiting these diverse neoantigens generated through splicing dysregulation expands the scope of immune therapy. Recent reports have reported that RECTAS compounds can induce tumor cells to produce neoantigens with immune-activating properties [127,128]. These neoantigens include six specific splice isoform variants of the Kifc1, Nf1, Acbd4, Rfx7, Qpctl, and Nup153 proteins, which are produced due to aberrant AS, offering a promising method for innovative cancer immunotherapy strategies. In breast and ovarian cancers, more than 68% of cases express an AS-derived neoepitope, whereas only 30% of cases express a neoepitope derived from a SNV [34]. The high frequency of AS-derived neoantigens in low-TMB tumors, such as breast and ovarian cancers, underscores their potential as targets for immunotherapy, particularly when mutation-derived neoantigens are limited.
In addition to affecting neoantigen generation, AS also plays a role in modulating cytokine and cytokine receptor activity. For example, in asthma, the AS of IL-33 results in the generation of isoforms that drive inflammation. In human airway epithelial cells, AS of the IL-33 transcript with deletion of exons 3 and 4 (Δ exon 3,4) activates basophils and mast cells to drive type 2 inflammation in chronic stable asthma [129]. In most cases, however, isoforms of IL-2, IL-4, IL-6, and others might act as antagonists to their corresponding wild-type forms and block their activity [130]. Similarly, soluble isoforms of cytokine receptors, such as IL-6R and TNFR2, often function as inhibitors by blocking ligand signaling.

3.10.2. AS in Immune Regulatory Molecules

AS plays a significant role in tumor immunity by generating tumor-specific neoantigens that can activate the immune system and, concurrently, enable tumor cells to evade immune surveillance. While AS-derived neoantigens have the potential to stimulate immune responses, some may lack immunogenicity, allowing tumors to evade immune detection [131,132,133]. Additionally, these neoantigens can trigger specific signaling pathways that reduce the expression of MHC I molecules on tumor cells’ surface, ultimately impeding the presentation of tumor antigens to T cells [134]. During the development of tumors, tumor cells often employ various mechanisms to reduce the expression of MHC I and decrease T cell recognition, facilitating immune evasion [135,136]. AS plays a crucial role in regulating immune checkpoint molecules, such as PD-1 and PD-L1, which are frequently dysregulated in tumors. In peripheral blood mononuclear cells (PBMCs) from healthy individuals, five splice isoforms of PD-1 mRNA have been identified [137,138], including PD-1∆ex2, PD-1∆ex3, PD-1∆ex2,3, PD-1∆ex2,3,4, and fIPD-1 [138,139]. In CRC, PD-L1 has distinct subtypes (e.g., PD-L1a, PD-L1b, PD-L1c) and soluble variants (e.g., PD-L1v229 and PD-L1v242) that act as decoys, Facilitating immune evasion by promoting PD-1/PD-L1 interactions [138,140,141,142]. Similarly, the AS of CTLA-4 generates soluble (sCTLA-4) and membrane-bound (mCTLA-4) forms [138,143,144], both of which contribute to immune escape.
SFs play a significant role in immune evasion mechanisms. For example, PTBP3 in SFs induces exon skipping in IL-18 in gallbladder cancer (GBC), producing a ΔIL-18 variant that reduces FBXO38-mediated PD-1 degradation in CD8+ T cells, thereby enhancing immune evasion [145]. This regulatory process substantially influences the diversity and specificity of immune-related gene expression, impacting the activity of cell surface receptors, including CD3, CD28, and CTLA-4, as well as kinases and phosphatases such as MAP4K2, MAP3K7 and CD45. Additionally, SFs regulate various transcription factors like GATA3 and FOXP3, and RBPs such as CELF2 and TIA-1 [28,146,147], essential for the proper development and response capabilities of the immune system.
Furthermore, tumor-derived splice isoforms can disrupt TCR signaling, hampering T cell activation and proliferation. Immune cells in the tumor microenvironment (TME), including regulatory T cells (Tregs) and tumor-associated macrophages (TAMs), are also influenced by tumor-specific AS events [148,149,150,151,152,153,154]. Notably, FOXP3, an important immunosuppressive molecule, exhibits two most abundant isoforms including full-length FOXP3 (FOXP3fl) and FOXP3 lacking exon 2 (FOXP3Δ2). FOXP3 lacking exons 2 and 7 (FOXP3Δ2Δ7) has been reported to inhibit other FOXP3 isoforms in a dominant negative manner, implying that exon 7 of FOXP3 is required for proper Treg cell function. The two different point mutations located near the intron 7 splice donor site result in the excision of FOXP3 exon 7 [155]. The splicing regulator USP39 modulates Treg function by maintaining CTLA-4 expression through lactate-mediated RNA splicing [156].

3.11. AS and Cancer Therapy

Advances in oncology research have shifted the focus of cancer treatment from traditional cytotoxic agents to targeted therapies. AS, a common event in cancer, has become a target for cancer therapy. By generating splice isoforms, AS contributes to tumorigenesis, immune evasion, and therapy resistance while also offering opportunities for innovative cancer treatments.

3.11.1. Cancer Immunotherapy

ACT therapies, particularly CAR-T and TCR-T cell therapies, have shown significant potential in cancer treatment [157]. CAR-T cell therapy involves genetically engineered T cells that can recognize and attack neoantigens produced by AS, although challenges such as limited neoantigen recognition, T-cell persistence, and the immunosuppressive TME hinder their efficacy in solid tumors [158]. In contrast, TCR-T cells utilize TCRs to recognize TSAs presented by MHC molecules on the cell membrane or derived from within the cell [159]. This allows TCR-T cells to recognize a broader range of target antigens and induce more sustained immune synapse formation, making them particularly promising for solid tumors. For example, TCR-T cell therapy targeting the cancer-testis antigen NY-ESO-1 has shown encouraging results in various cancers, including NSCLC, CRC, hepatocellular carcinoma (HCC), and multiple myeloma [160,161,162]. Tumor-specific neoantigens generated by aberrant AS events presented by MHC I complexes provide a theoretical basis for TCR-T cell immunotherapy. Despite challenges such as neoantigen cross-reactivity, MHC restrictions, and mismatch risks, TCR-T cell therapy has demonstrated safety and efficacy, as evidenced by the FDA-approved kimtrak (tebentafusp-tebn) for the treatment of metastatic melanoma [163].
The lack of targetable antigens remains a significant challenge for immunotherapies such as CAR-T, TCR-T, and vaccines. While proteomics data often fail to detect AS-derived peptides due to NMD, advances in AI and computational biology now enable the screening of immunogenic shared neoantigens derived from dysregulated AS [164]. These neoantigens can serve as targets for CAR-T and TCR-T cell therapies, with engineered T cell administered to patients to increase immunotherapy efficacy. This approach holds great promise for overcoming current limitations and improving treatment outcomes. The integration of AS regulation with ICIs, CAR-T/TCR-T cell therapies, and personalized cancer vaccines represents a transformative strategy in cancer immunotherapy. By targeting immunogenic splice isoforms and neoantigens, these approaches offer new avenues for enhancing immune recognition and overcoming tumor immune evasion, paving the way for more effective and personalized cancer treatments.
The regulation of AS plays a pivotal role in shaping the diversity and specificity of immune-related gene expression, influencing the activity of cell surface receptors (e.g., CD3, Fas, CTLA-4), kinases and phosphatases (e.g., MAP4K2, MAP3K7, CD45), and transcription factors (e.g., GATA3 and FOXP3) as well as RBPs (e.g., CELF2 and TIA-1) (Table 2). Numerous studies have shown that during the process of cell apoptosis, Fas is regulated by several SFs (e.g., SRSF6, hnRNPC, and PTB). In normal AS, SFSR6 binds to the UGCCAA region in exon 6 of the Fas gene [165]. This binding promotes the inclusion of exon 6. A fully functional Fas protein is subsequently translated. This Fas protein promotes the apoptosis of normal cells through the Fas/FasL pathway. In contrast, in abnormal AS, PTB binds to the uridine-rich sequence (URE6) of the CUCUCU region located in exon 6 of the Fas gene [166]. This binding promotes the skipping of exon 6. As a result, the FASΔ6 protein is translated. Additionally, this protein competitively binds to FasL with the fully functional Fas protein, thereby facilitating the immune escape of tumors (Figure 1).
The combined application of neoantigens produced by aberrant AS and ICIs represents an emerging therapeutic strategy [184,185]. This strategy is based on the immunogenic neoantigens generated by tumor-specific AS, which can be recognized by the immune system as “nonself” components, thereby stimulating an antitumor immune response [186,187]. For example, studies have shown that RBM39 of the SF degrader Indisulam can induce tumor cells to produce neoantigens that are presented by MHC I to stimulate an antitumor immune response. The combination of Indisulam and PD-1 antibodies has been shown to synergistically inhibit tumor growth, highlighting the therapeutic potential of integrating AS regulation with ICIs.

3.11.2. SSOs and Cancer Therapy

SSOs are small, synthetic nucleic acids that interfere with the pre-mRNA splicing process by disrupting RNA-RNA or protein-RNA interactions essential for the splicing machinery [32]. SSOs differ from siRNAs in regulating gene expression through splicing control rather than RNA degradation. AS has emerged as a promising target for cancer therapy, with SSOs being able to modulate the expression of critical oncogenic isoforms. By redirecting splicing toward proapoptotic isoforms, SSOs have demonstrated significant antitumor effects in both cell culture and xenograft models. For example, targeting Bcl-x, HER4, MDM4, and STAT3 splicing variants with SSOs has shown significant antitumor effects in cell culture and xenograft models. STAT3 exists in two main isoforms, namely, full-length STAT3α and truncated STAT3β, which are generated by the AS of exon 23 [35]. STAT3β acts as a dominant-negative regulator of transcription and promotes apoptosis. SSOs that redirect splicing from STAT3α to STAT3β have been shown to increase cancer cell death and induce tumor regression in xenograft models. In contrast to the effect mediated by total STAT3 knockdown-induced FSD-NMD, SSOs displayed a unique STAT3β signature, with the downregulation of specific targets. The Bcl-x gene produces two primary protein isoforms with opposing functions: the antiapoptotic protein Bcl-xL and the proapoptotic protein Bcl-xS. Bcl-xL is elevated in various cancers and contributes to resistance to a wide spectrum of chemotherapeutic agents. Conversely, Bcl-xS counteracts the antiapoptotic effects of Bcl-xL. SSOs that shift Bcl-x splicing from Bcl-xL to Bcl-xS induce apoptosis and increase sensitivity to chemotherapy in cancer cells cultured in vitro and inhibit tumor growth in vivo [188]. The insulin receptor (IR) is differentially spliced at exon 11 to generate two distinct isoforms—namely, IR-B (which includes exon 11) and IR-A (which excludes exon 11). SSOs that target IR splicing, either by modulating regulatory elements such as CELF1 or directly restoring splicing from IR-A to IR-B, have been shown to inhibit osteosarcoma growth and resistance to anoikis [189,190].

3.11.3. Targeting Novel Splice Variants

Given the importance of mRNA AS and the fundamental role of the spliceosome posttranscriptionally, the spliceosome has attracted attention as an anticancer target. Small-molecule compounds such as pladienolides [16] modulate the SF3b subunit of the spliceosome, interfering with its function in a dose-dependent manner. Drugs such as Indisulam (E7070), a small-molecule inhibitor, degrade RBM39, leading to widespread aberrant splicing and translation of key downstream effector proteins [191]. Indisulam has shown therapeutic effects in various cancer types, such as T-cell acute lymphoblastic leukemia (T-ALL) [109], high-grade serous ovarian cancer (HGSC) [110], and head and neck squamous cell carcinoma (HNSCC) [111]. Tepotinib [192] is a highly selective MET tyrosine kinase inhibitor, targeting the specific oncogenic driving mechanism of MET gene exon 14 skipping mutations in NSCLC [42]. This mutation leads to the deletion of exon 14 through AS of RNA, causing the MET receptor tyrosine kinase to avoid degradation and remain continuously activated, thereby abnormally activating the downstream pro-proliferative signaling pathways. As the first targeted drug approved for this target, Tepotinib precisely inhibits the kinase domain of the MET protein, blocking its abnormal signal transduction and demonstrating significant antitumor activity. This breakthrough validates the druggability of abnormal AS as a target for cancer treatment, marking a new stage in the clinical application of targeted therapies developed based on the RNA splicing mechanism.
Aberrant splicing in tumor cells generates neoantigens that can be presented by MHC I molecules, enhancing T cell recognition and attack [193]. Modulating AS with drugs such as Indisulam increases the presentation of these neoantigens, stimulating antitumor immune responses. Preclinical studies have demonstrated that combining Indisulam with PD-1 antibodies synergistically inhibits tumor growth, highlighting the potential of integrating AS regulation with ICIs for improved cancer immunotherapy [194].

4. Innovative Technologies in the Study of AS

Tumor-associated aberrant splicing events contribute to approximately 15–30% of oncogenic mutations, with frameshift proteins derived from these events potentially activating the immune system through neoantigen presentation [195]. Traditional genomic methods, such as whole-exome sequencing (WES), detect only approximately 1.5% of tumor neoantigens, whereas splicing-driven neoantigens account for 34–83% of cases [196]. Recent advances in computational biology and multi-omics technologies have shifted research focus toward high-precision splicing analysis tools and cross-omics integration strategies to systematically decode tumor-specific splicing events and their immunotherapeutic potential.

4.1. Splicing Analysis Tools and Databases

The diversification of differential splicing detection tools has propelled cancer research. Replicate Multivariate Analysis of Transcript Splicing (rMATS) [197] employs likelihood ratio tests and ΔPSI quantification to identify five classical splicing events, with outputs (e.g., BRAF exon skipping in melanoma) serving as starting points for neoantigen screening.
Whippet [198], leveraging lightweight algorithms, enables efficient quantification of complex splicing events on standard laptops, revealing that high-entropy AS events affect up to 40% of human genes.
Psichomics [199] integrates multi-omics data (RNA-seq, clinical outcomes) from TCGA, supporting AS event detection (e.g., exon skipping/intron retention), isoform quantification (via PSI values), and survival analysis (Cox models, KM curves). Its tumor-specific splicing outputs (e.g., exon-skipped aberrant transcripts) encode neoantigens, offering candidates for immunotherapy.
AS Cancer Atlas (https://ngdc.cncb.ac.cn/ascancer, accessed on 25 March 2025) [200], a core repository, catalogs 17,842 experimentally validated cancer-associated AS events, including splice sites, isoform expression, and prognostic data. This helps researchers gain deeper insights into splicing regulation and discover potential therapeutic targets.

4.2. Neoantigen Prediction Innovations

Splicing-driven neoantigen discovery relies on algorithmic advancements and multi-dimensional validation. The Spliced Neo Antigen Finder (SNAF) [201] tool integrates DeepImmuno-CNN and BayesTS models. Notably, more than 90% of more than 500 melanoma patients presented with shared splice neoantigens.
Researchers from the Children’s Hospital of Philadelphia (CHOP) and the University of California, Los Angeles (UCLA), have collaboratively developed a computational platform called isoform peptides from RNA splicing for immunotherapy target screening (IRIS) [202,203], that the platform combines tumor/normal differential expression (fold change >5) and HLA-I affinity prediction, prioritizing 48 TCR targets in neuroendocrine prostate cancer (NEPC) from 2939 splicing events.
A research team from the University of Lausanne has employed a revolutionary machine learning (ML) [204] method to significantly enhance the ability to identify immunogenic neoantigens and mutations, which is crucial for the development of personalized cancer immunotherapies. This technology has reprocessed WES and RNA-seq data from 112 cancer patients from the National Cancer Institute (NCI), 8 patients from the Tumor Neoantigen Selection Alliance (TESLA), and an internal dataset of 11 patients. The results revealed over 46,000 somatic single-nucleotide mutations and approximately 1.78 million new peptides, of which 212 mutations and 178 peptides showed immunogenicity. Moreover, the classifier trained on the large NCI dataset was able to accurately predict the immunogenicity of neoantigens across multiple datasets, and this method outperformed previous approaches in terms of orthogonal features, increasing the accuracy of predictions and the number of high-ranking immunogenic peptides by up to 30%. For specific details, refer to Table 3.

4.3. Multi-Omics Integration Strategy

Initial screening is conducted using NGS-based WES and RNA-seq (Illumina NovaSeq X Plus) to identify high-frequency mutations and differentially expressed genes [205,206,207,208,209,210,211]. Candidate splicing events are pinpointed via rMATS [197] and Whippet [198]. PacBio SMRT (10–15 kb reads) resolves complex structural variants (e.g., full-length ALK fusion isoforms), while HiFi sequencing [212] (CCS mode, 10–25 kb reads, accuracy ≥99.9%) validates low-abundance splice variants. Cross-referencing with AS Cancer Atlas identifies tumor-specific frameshift transcripts. Orbitrap Astral mass spectrometry (MS) [213,214,215,216] (DIA mode, 0.1 attomolar sensitivity) detects frameshift-derived peptides, followed by Spectronaut 16.0 spectral matching and XGBoost 3.0.1 for HLA-I/II binding prediction. Final immunogenicity validation employs the IRIS [202,203] platform and in vitro T-cell activation assays, establishing a closed-loop system from splicing discovery to therapeutic targets. This strategy increases neoantigen detection rates compared to traditional methods.
From differential splicing tools (rMATS, Whippet) to neoantigen algorithms (SNAF, XGBoost), and from long-read sequencing (HiFi) to multi-omics integration strategy, technological advancements are driving clinical translation. Future breakthroughs in cost-effective long-read sequencing, single-cell proteomics, and causal inference models will further overcome bottlenecks, enabling scalable personalized immunotherapy based on splicing-derived neoantigens.

5. Challenges and Difficulties

Despite the promising use of neoantigens in cancer immunotherapy, their discovery and clinical application remain limited due to several challenges. Tumor heterogeneity and dynamic changes lead to inconsistent neoantigen expression, complicating their identification and use as therapeutic targets. Additionally, only a subset of neoantigens possesses sufficient immunogenicity to trigger effective immune responses, whereas tumor immune escape mechanisms further reduce immune recognition. Predicting neoantigens requires advanced computational biology methods, and experimental validation is both time-consuming and costly. Other hurdles include individual variations in immune responses; technical limitations in high-throughput data analysis; and the complexities of clinical translation, such as production, safety, efficacy, and regulatory approval. These factors collectively create a multistep, interdisciplinary process that demands continuous technological innovation and cross-disciplinary collaboration.

6. Conclusions and Prospects

Aberrant AS in tumor cells plays a dual role in cancer immunotherapy. On the one hand, it can facilitate immune evasion by enabling tumor cells to escape immune surveillance. On the other hand, it generates immunogenic neoantigens that are presented by MHC I molecules to T cells, triggering antitumor immune responses. To expand the repertoire of potential immunotherapeutic antigens, recent studies have explored aberrant splicing events across multiple cancers as an additional source of TSAs. These cancer-specific splicing events, otherwise known as NJs, are prevalent in cancer cells and capable of generating novel TSAs that potentiate CD8+ T cell-mediated expansion and responses in select cancer types. When combined with ICIs (e.g., PD-1 or PD-L1 inhibitors), these neoantigens have synergistic potential in inhibiting tumor growth, laying the groundwork for personalized immunotherapy. However, neoantigens may lack sufficient immunogenicity, or the quantity may not be enough to activate an effective antitumor immune response. In addition, tumor cells may also regulate splice isoforms to evade immune surveillance. Although TCR-T and CAR-T cell therapies have shown promise in immunotherapy targeting neoantigens, they also face challenges such as MHC restrictions, potential mismatch risks, and immune suppression of the TME.
Advancements in computational tools and platforms, such as SNAF, ML, and IRIS, are revolutionizing the identification of immunogenic neoantigens. These tools enhance the precision of immunotherapy and improve patient outcomes. Additionally, AI algorithms and high-throughput sequencing technologies are enabling the design of personalized neoantigen vaccines tailored to the unique splicing profiles of individual tumors. Clinical trials have demonstrated that these vaccines can stimulate potent immune responses, particularly when combined with ICIs.
Despite challenges such as limited neoantigen immunogenicity, immune suppression mediated by the TME, and the high costs and technical demands of personalized therapies, ongoing clinical trials and technological innovations are paving the way for more effective and precise treatments. The discovery and utilization of neoantigens, along with strategies to modulate AS, will be pivotal in advancing cancer immunotherapy. As our understanding of tumor-specific splicing events deepens, more therapies targeting immunogenic neoantigens are expected to emerge. These neoantigens not only provide targets for personalized vaccines but also increase the efficacy of ICIs and CAR-T and TCR-T cell therapies. The development of personalized neoantigen vaccines represents a significant step toward precision medicine.

Author Contributions

H.C., J.T. and J.X. conceived the manuscript. H.C. prepared all the figures, Tables and wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Key Research and Development Program of Hunan [2019SK2253], the National Natural Science Foundation of Changsha [kq2208299, kq2403084] and the National Natural Science Foundation of Hunan [2025JJ50173].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable, no new data are presented in this review.

Conflicts of Interest

Authors declare that they have no competing interests.

Abbreviations

ACTAdoptive Cell Therapy.
AMLAcute Myeloid Leukemia.
ASAlternative Splicing.
AS Cancer AtlasAlternative Splicing Cancer Atlas.
BCBreast Cancer.
BCRB-cell Receptor.
CAR-TChimeric Antigen Receptor T-cell.
CCSCircular Consensus Sequencing.
CRCColorectal Cancer.
CHOPChildren’s Hospital of Philadelphia.
EMTEpithelial-to-Mesenchymal Transition.
ESEsExonic Splicing Enhancers.
ESSsExonic Splicing Silencers.
ERsEstrogen Receptors.
EREndoplasmic Reticulum.
FDSIsFunctionally Distinct Splice Isoforms.
FLT3-ITDsFLT3 Internal Tandem Duplications.
GBMGlioblastoma.
GBCGallbladder Cancer.
HCCHepatocellular Carcinoma.
HGSCHigh-grade serous ovarian cancer.
HNSCCHead and neck squamous cell carcinoma.
hnRNPsHeterogeneous Nuclear Ribonucleoproteins.
ISEsIntronic Splicing Enhancers.
ISSsIntronic Splicing Silencers.
ICBImmune Checkpoint Blockade.
ICIImmune Checkpoint Inhibitor.
IgImmunoglobulin.
IRISIsoform Peptides from RNA Splicing for Immunotherapy Target Screening.
ISRIntegrated Stress Response.
IRInsulin Receptor.
LUADLung Adenocarcinoma.
MLMachine Learning.
MHCMajor Histocompatibility Complex.
m6AN6-methyladenosine.
MDSMyelodysplastic syndromes.
MSMass Spectrometry.
NJsneojunctions.
NMDNonsense—mediated Decay.
NSCLCNon-small cell lung cancer.
NGSNext-Generation Sequencing.
NEPCNeuroendocrine prostate cancer.
NCINational Cancer Institute.
ORFsOpen Reading Frames.
PBMCsPeripheral Blood Mononuclear Cells.
PDACPancreatic ductal adenocarcinoma.
PTCsPremature Termination Codons.
RBPsRNA-binding proteins.
RBMRNA-binding Motif RNA.
ROSReactive Oxygen Species.
rMATSreplicate Multivariate Analysis of Transcript Splicing.
snRNAssmall nuclear RNAs.
snRNPssmall nuclear ribonucleoproteins.
SAVsSplice-altering Variants.
SNAFSpliced Neo Antigen Finder.
SNVSingle-Nucleotide Variant.
SMRTSingle Molecule, Real-Time.
SRsSerine/Arginine-rich Proteins.
SSOsSplice-switching Oligonucleotides.
TAMsTumor Associated Macrophages.
TCRT-cell Receptor.
TCR-TT-cell Receptor Engineered T-cell.
TregsRegulatory T Cells.
TMETumor Microenvironment.
TMBTumor Mutational Burden.
TKIsTyrosine Kinase Inhibitors.
TSAsTumor-Specific Antigens.
T-ALLT-cell acute lymphoblastic leukemia.
TESLATumor Neoantigen Selection Alliance.
URE6Uridine-rich sequence located in exon 6.
UCLAUniversity of California, Los Angeles.
WESWhole-exome sequencing.

References

  1. Huang, P.; Wen, F.; Tuerhong, N.; Yang, Y.; Li, Q. Neoantigens in cancer immunotherapy: Focusing on alternative splicing. Front. Immunol. 2024, 15, 1437774. [Google Scholar] [CrossRef]
  2. Blackwell, D.L.; Fraser, S.D.; Caluseriu, O.; Vivori, C.; Tyndall, A.V.; Lamont, R.E.; Parboosingh, J.S.; Innes, A.M.; Bernier, F.P.; Childs, S.J. Hnrnpul1 controls transcription, splicing, and modulates skeletal and limb development in vivo. G3 2022, 12, jkac067. [Google Scholar] [CrossRef]
  3. Sciarrillo, R.; Wojtuszkiewicz, A.; Assaraf, Y.G.; Jansen, G.; Kaspers, G.J.L.; Giovannetti, E.; Cloos, J. The role of alternative splicing in cancer: From oncogenesis to drug resistance. Drug Resist. Updat. 2020, 53, 100728. [Google Scholar] [CrossRef] [PubMed]
  4. Lu, S.; Jia, Z.; Meng, X.; Chen, Y.; Wang, S.; Fu, C.; Yang, L.; Zhou, R.; Wang, B.; Cao, Y. Combined Metabolomic and Transcriptomic Analysis Reveals Allantoin Enhances Drought Tolerance in Rice. Int. J. Mol. Sci. 2022, 23, 14172. [Google Scholar] [CrossRef] [PubMed]
  5. Li, D.; Yu, W.; Lai, M. Towards understandings of serine/arginine-rich splicing factors. Acta Pharm. Sin. B 2023, 13, 3181–3207. [Google Scholar] [CrossRef] [PubMed]
  6. Mehta, Z.; Touma, M. Post-Transcriptional Modification by Alternative Splicing and Pathogenic Splicing Variants in Cardiovascular Development and Congenital Heart Defects. Int. J. Mol. Sci. 2023, 24, 1555. [Google Scholar] [CrossRef]
  7. Schindler, N.R.; Braun, D.A. Antigenic targets in clear cell renal cell carcinoma. Kidney Cancer 2023, 7, 81–91. [Google Scholar] [CrossRef]
  8. Ouedraogo, W.; Ouangraoua, A. SimSpliceEvol2: Alternative splicing-aware simulation of biological sequence evolution and transcript phylogenies. BMC Bioinform. 2024, 25, 235. [Google Scholar] [CrossRef]
  9. Shender, V.O.; Anufrieva, K.S.; Shnaider, P.V.; Arapidi, G.P.; Pavlyukov, M.S.; Ivanova, O.M.; Malyants, I.K.; Stepanov, G.A.; Zhuravlev, E.; Ziganshin, R.H.; et al. Therapy-induced secretion of spliceosomal components mediates pro-survival crosstalk between ovarian cancer cells. Nat. Commun. 2024, 15, 5237. [Google Scholar] [CrossRef]
  10. Donnellan, L.; Young, C.; Simpson, B.S.; Acland, M.; Dhillon, V.S.; Costabile, M.; Fenech, M.; Hoffmann, P.; Deo, P. Proteomic Analysis of Methylglyoxal Modifications Reveals Susceptibility of Glycolytic Enzymes to Dicarbonyl Stress. Int. J. Mol. Sci. 2022, 23, 3689. [Google Scholar] [CrossRef]
  11. Freytag, M.; Kluth, M.; Bady, E.; Hube-Magg, C.; Makrypidi-Fraune, G.; Heinzer, H.; Höflmayer, D.; Weidemann, S.; Uhlig, R.; Huland, H.; et al. Epithelial splicing regulatory protein 1 and 2 (ESRP1 and ESRP2) upregulation predicts poor prognosis in prostate cancer. BMC Cancer 2020, 20, 1220. [Google Scholar] [CrossRef]
  12. Li, Y.; Zhang, S.; Li, Y.; Liu, J.; Li, Q.; Zang, W.; Pan, Y. The Regulatory Network of hnRNPs Underlying Regulating PKM Alternative Splicing in Tumor Progression. Biomolecules 2024, 14, 566. [Google Scholar] [CrossRef] [PubMed]
  13. Bei, M.; Xu, J. SR proteins in cancer: Function, regulation, and small inhibitor. Cell Mol. Biol. Lett. 2024, 29, 78. [Google Scholar] [CrossRef]
  14. Pan, D.; Long, L.; Li, C.; Zhou, Y.; Liu, Q.; Zhao, Z.; Zhao, H.; Lin, W.; Zheng, Z.; Peng, L.; et al. Splicing factor hnRNPA1 regulates alternative splicing of LOXL2 to enhance the production of LOXL2Δ13. J. Biol. Chem. 2024, 300, 107414. [Google Scholar] [CrossRef]
  15. Emilius, L.; Bremm, F.; Binder, A.K.; Schaft, N.; Dörrie, J. Tumor Antigens beyond the Human Exome. Int. J. Mol. Sci. 2024, 25, 4673. [Google Scholar] [CrossRef] [PubMed]
  16. Zhang, A.; Miao, K.; Sun, H.; Deng, C.X. Tumor heterogeneity reshapes the tumor microenvironment to influence drug resistance. Int. J. Biol. Sci. 2022, 18, 3019–3033. [Google Scholar] [CrossRef] [PubMed]
  17. Manabile, M.A.; Hull, R.; Khanyile, R.; Molefi, T.; Damane, B.P.; Mongan, N.P.; Bates, D.O.; Dlamini, Z. Alternative Splicing Events and Their Clinical Significance in Colorectal Cancer: Targeted Therapeutic Opportunities. Cancers 2023, 15, 3999. [Google Scholar] [CrossRef]
  18. Bhuiyan, S.A.; Ly, S.; Phan, M.; Huntington, B.; Hogan, E.; Liu, C.C.; Liu, J.; Pavlidis, P. Systematic evaluation of isoform function in literature reports of alternative splicing. BMC Genomics 2018, 19, 637. [Google Scholar] [CrossRef]
  19. Gueroussov, S.; Gonatopoulos-Pournatzis, T.; Irimia, M.; Raj, B.; Lin, Z.Y.; Gingras, A.C.; Blencowe, B.J. An alternative splicing event amplifies evolutionary differences between vertebrates. Science 2015, 349, 868–873. [Google Scholar] [CrossRef]
  20. Fair, B.; Buen Abad Najar, C.F.; Zhao, J.; Lozano, S.; Reilly, A.; Mossian, G.; Staley, J.P.; Wang, J.; Li, Y.I. Global impact of unproductive splicing on human gene expression. Nat. Genet. 2024, 56, 1851–1861. [Google Scholar] [CrossRef]
  21. Li, S.; Guo, W.; Dewey, C.N.; Greaser, M.L. Rbm20 regulates titin alternative splicing as a splicing repressor. Nucleic Acids Res. 2013, 41, 2659–2672. [Google Scholar] [CrossRef] [PubMed]
  22. Lin, J.C.; Yan, Y.T.; Hsieh, W.K.; Peng, P.J.; Su, C.H.; Tarn, W.Y. RBM4 promotes pancreas cell differentiation and insulin expression. Mol. Cell Biol. 2013, 33, 319–327. [Google Scholar] [CrossRef]
  23. Baralle, F.E.; Giudice, J. Alternative splicing as a regulator of development and tissue identity. Nat. Rev. Mol. Cell Biol. 2017, 18, 437–451. [Google Scholar] [CrossRef] [PubMed]
  24. Yano, M.; Hayakawa-Yano, Y.; Mele, A.; Darnell, R.B. Nova2 regulates neuronal migration through an RNA switch in disabled-1 signaling. Neuron 2010, 66, 848–858. [Google Scholar] [CrossRef] [PubMed]
  25. Kim, K.K.; Nam, J.; Mukouyama, Y.S.; Kawamoto, S. Rbfox3-regulated alternative splicing of Numb promotes neuronal differentiation during development. J. Cell Biol. 2013, 200, 443–458. [Google Scholar] [CrossRef]
  26. Sen, S.; Jumaa, H.; Webster, N.J. Splicing factor SRSF3 is crucial for hepatocyte differentiation and metabolic function. Nat. Commun. 2013, 4, 1336. [Google Scholar] [CrossRef]
  27. Rothrock, C.R.; House, A.E.; Lynch, K.W. HnRNP L represses exon splicing via a regulated exonic splicing silencer. Embo J. 2005, 24, 2792–2802. [Google Scholar] [CrossRef] [PubMed]
  28. Banerjee, S.; Galarza-Muñoz, G.; Garcia-Blanco, M.A. Role of RNA Alternative Splicing in T Cell Function and Disease. Genes 2023, 14, 1896. [Google Scholar] [CrossRef]
  29. Hirano, M.; Galarza-Muñoz, G.; Nagasawa, C.; Schott, G.; Wang, L.; Antonia, A.L.; Jain, V.; Yu, X.; Widen, S.G.; Briggs, F.B.S.; et al. The RNA helicase DDX39B activates FOXP3 RNA splicing to control T regulatory cell fate. eLife 2023, 12, e76927. [Google Scholar] [CrossRef]
  30. Yamamoto, M.L.; Clark, T.A.; Gee, S.L.; Kang, J.A.; Schweitzer, A.C.; Wickrema, A.; Conboy, J.G. Alternative pre-mRNA splicing switches modulate gene expression in late erythropoiesis. Blood 2009, 113, 3363–3370. [Google Scholar] [CrossRef]
  31. Shi, W.; Tang, J.; Xiang, J. Therapeutic strategies for aberrant splicing in cancer and genetic disorders. Clin. Genet. 2024, 105, 345–354. [Google Scholar] [CrossRef] [PubMed]
  32. Havens, M.A.; Hastings, M.L. Splice-switching antisense oligonucleotides as therapeutic drugs. Nucleic Acids Res. 2016, 44, 6549–6563. [Google Scholar] [CrossRef]
  33. Deng, K.; Yao, J.; Huang, J.; Ding, Y.; Zuo, J. Abnormal alternative splicing promotes tumor resistance in targeted therapy and immunotherapy. Transl. Oncol. 2021, 14, 101077. [Google Scholar] [CrossRef] [PubMed]
  34. Kahles, A.; Lehmann, K.V.; Toussaint, N.C.; Hüser, M.; Stark, S.G.; Sachsenberg, T.; Stegle, O.; Kohlbacher, O.; Sander, C.; Rätsch, G. Comprehensive Analysis of Alternative Splicing Across Tumors from 8705 Patients. Cancer Cell 2018, 34, 211–224.e6. [Google Scholar] [CrossRef]
  35. Zammarchi, F.; de Stanchina, E.; Bournazou, E.; Supakorndej, T.; Martires, K.; Riedel, E.; Corben, A.D.; Bromberg, J.F.; Cartegni, L. Antitumorigenic potential of STAT3 alternative splicing modulation. Proc. Natl. Acad. Sci. USA 2011, 108, 17779–17784. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, L.; Yin, N.; Shi, W.; Xie, Y.; Yi, J.; Tang, Z.; Tang, J.; Xiang, J. Splicing inhibition mediated by reduced splicing factors and helicases is associated with the cellular response of lung cancer cells to cisplatin. Comput. Struct. Biotechnol. J. 2024, 23, 648–658. [Google Scholar] [CrossRef]
  37. 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]
  38. Park, E.; Pan, Z.; Zhang, Z.; Lin, L.; Xing, Y. The Expanding Landscape of Alternative Splicing Variation in Human Populations. Am. J. Hum. Genet. 2018, 102, 11–26. [Google Scholar] [CrossRef]
  39. Cortés-López, M.; Schulz, L.; Enculescu, M.; Paret, C.; Spiekermann, B.; Quesnel-Vallières, M.; Torres-Diz, M.; Unic, S.; Busch, A.; Orekhova, A.; et al. High-throughput mutagenesis identifies mutations and RNA-binding proteins controlling CD19 splicing and CART-19 therapy resistance. Nat. Commun. 2022, 13, 5570. [Google Scholar] [CrossRef]
  40. Shlien, A.; Raine, K.; Fuligni, F.; Arnold, R.; Nik-Zainal, S.; Dronov, S.; Mamanova, L.; Rosic, A.; Ju, Y.S.; Cooke, S.L.; et al. Direct Transcriptional Consequences of Somatic Mutation in Breast Cancer. Cell Rep. 2016, 16, 2032–2046. [Google Scholar] [CrossRef]
  41. Luo, J.; Chen, C.; Liu, Z.; Wang, X. The mutation in splicing factor genes correlates with unfavorable prognosis, genomic instability, anti-tumor immunosuppression and increased immunotherapy response in pan-cancer. Front. Cell Dev. Biol. 2022, 10, 1045130. [Google Scholar] [CrossRef] [PubMed]
  42. Van Der Steen, N.; Giovannetti, E.; Pauwels, P.; Peters, G.J.; Hong, D.S.; Cappuzzo, F.; Hirsch, F.R.; Rolfo, C. cMET Exon 14 Skipping: From the Structure to the Clinic. J. Thorac. Oncol. 2016, 11, 1423–1432. [Google Scholar] [CrossRef]
  43. Shiraishi, Y.; Kataoka, K.; Chiba, K.; Okada, A.; Kogure, Y.; Tanaka, H.; Ogawa, S.; Miyano, S. A comprehensive characterization of cis-acting splicing-associated variants in human cancer. Genome Res. 2018, 28, 1111–1125. [Google Scholar] [CrossRef] [PubMed]
  44. Ullah, F.; Jabeen, S.; Salton, M.; Reddy, A.S.N.; Ben-Hur, A. Evidence for the role of transcription factors in the co-transcriptional regulation of intron retention. Genome Biol. 2023, 24, 53. [Google Scholar] [CrossRef] [PubMed]
  45. Zhou, H.L.; Luo, G.; Wise, J.A.; Lou, H. Regulation of alternative splicing by local histone modifications: Potential roles for RNA-guided mechanisms. Nucleic Acids Res. 2014, 42, 701–713. [Google Scholar] [CrossRef]
  46. Bar-Ziv, R.; Voichek, Y.; Barkai, N. Chromatin dynamics during DNA replication. Genome Res. 2016, 26, 1245–1256. [Google Scholar] [CrossRef]
  47. Do, H.T.T.; Shanak, S.; Barghash, A.; Helms, V. Differential exon usage of developmental genes is associated with deregulated epigenetic marks. Sci. Rep. 2023, 13, 12256. [Google Scholar] [CrossRef]
  48. Yang, H.; Wang, Y.; Zhang, Y. Characterization of H3K9me3 and DNA methylation co-marked CpG-rich regions during mouse development. BMC Genomics 2023, 24, 663. [Google Scholar] [CrossRef]
  49. Batsché, E.; Yi, J.; Mauger, O.; Kornobis, E.; Hopkins, B.; Hanmer-Lloyd, C.; Muchardt, C. CD44 alternative splicing senses intragenic DNA methylation in tumors via direct and indirect mechanisms. Nucleic Acids Res. 2021, 49, 6213–6237. [Google Scholar] [CrossRef]
  50. Buratti, E.; Baralle, F.E. Influence of RNA secondary structure on the pre-mRNA splicing process. Mol. Cell Biol. 2004, 24, 10505–10514. [Google Scholar] [CrossRef]
  51. Zhu, Z.M.; Huo, F.C.; Zhang, J.; Shan, H.J.; Pei, D.S. Crosstalk between m6A modification and alternative splicing during cancer progression. Clin. Transl. Med. 2023, 13, e1460. [Google Scholar] [CrossRef]
  52. Zhao, B.; Xiang, Z.; Wu, B.; Zhang, X.; Feng, N.; Wei, Y.; Zhang, W. Use of Novel m6A Regulator-mediated Methylation Modification Patterns in Distinct Tumor Microenvironment Profiles to Identify and Predict Glioma Prognosis and Progression, T-cell Dysfunction, and Clinical Response to ICI Immunotherapy. Curr. Pharm. Des. 2023, 29, 60–78. [Google Scholar] [CrossRef] [PubMed]
  53. Dominissini, D.; Moshitch-Moshkovitz, S.; Schwartz, S.; Salmon-Divon, M.; Ungar, L.; Osenberg, S.; Cesarkas, K.; Jacob-Hirsch, J.; Amariglio, N.; Kupiec, M.; et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 2012, 485, 201–206. [Google Scholar] [CrossRef]
  54. Yan, Y.; Ren, Y.; Bao, Y.; Wang, Y. RNA splicing alterations in lung cancer pathogenesis and therapy. Cancer Pathog. Ther. 2023, 1, 272–283. [Google Scholar] [CrossRef]
  55. Alors-Perez, E.; Blázquez-Encinas, R.; Alcalá, S.; Viyuela-García, C.; Pedraza-Arevalo, S.; Herrero-Aguayo, V.; Jiménez-Vacas, J.M.; Mafficini, A.; Sánchez-Frías, M.E.; Cano, M.T.; et al. Dysregulated splicing factor SF3B1 unveils a dual therapeutic vulnerability to target pancreatic cancer cells and cancer stem cells with an anti-splicing drug. J. Exp. Clin. Cancer Res. 2021, 40, 382. [Google Scholar] [CrossRef]
  56. Jyotsana, N.; Heuser, M. Exploiting differential RNA splicing patterns: A potential new group of therapeutic targets in cancer. Expert Opin. Ther. Targets 2018, 22, 107–121. [Google Scholar] [CrossRef]
  57. Wen, W.X.; Mead, A.J.; Thongjuea, S. MARVEL: An integrated alternative splicing analysis platform for single-cell RNA sequencing data. Nucleic Acids Res. 2023, 51, e29. [Google Scholar] [CrossRef] [PubMed]
  58. Alsafadi, S.; Dayot, S.; Tarin, M.; Houy, A.; Bellanger, D.; Cornella, M.; Wassef, M.; Waterfall, J.J.; Lehnert, E.; Roman-Roman, S.; et al. Genetic alterations of SUGP1 mimic mutant-SF3B1 splice pattern in lung adenocarcinoma and other cancers. Oncogene 2021, 40, 85–96. [Google Scholar] [CrossRef] [PubMed]
  59. Funakoshi, Y.; Sugihara, Y.; Uneda, A.; Nakashima, T.; Suzuki, H. Recent advances in the molecular understanding of medulloblastoma. Cancer Sci. 2023, 114, 741–749. [Google Scholar] [CrossRef]
  60. Zhang, Z.; Huang, R.; Lai, Y. Expression signature of ten small nuclear RNAs serves as novel biomarker for prognosis prediction of acute myeloid leukemia. Sci. Rep. 2023, 13, 18489. [Google Scholar] [CrossRef]
  61. George, J.; Chen, Y.; Abdelfattah, N.; Yamamoto, K.; Gallup, T.D.; Adamson, S.I.; Rybinski, B.; Srivastava, A.; Kumar, P.; Lee, M.G.; et al. Cancer stem cells, not bulk tumor cells, determine mechanisms of resistance to SMO inhibitors. Cancer Res. Commun. 2022, 2, 402–416. [Google Scholar] [CrossRef] [PubMed]
  62. Eisemann, T.; Wechsler-Reya, R.J. Coming in from the cold: Overcoming the hostile immune microenvironment of medulloblastoma. Genes Dev. 2022, 36, 514–532. [Google Scholar] [CrossRef] [PubMed]
  63. Richard, S.A. The pivotal role of irradiation-induced apoptosis in the pathogenesis and therapy of medulloblastoma. Cancer Rep. 2024, 7, e2048. [Google Scholar] [CrossRef]
  64. Padariya, M.; Vojtesek, B.; Hupp, T.; Kalathiya, U. In Vitro Cross-Linking MS Reveals SMG1-UPF2-SMG7 Assembly as Molecular Partners within the NMD Surveillance. Int. J. Mol. Sci. 2024, 25, 3182. [Google Scholar] [CrossRef]
  65. Ge, Y.; Porse, B.T. The functional consequences of intron retention: Alternative splicing coupled to NMD as a regulator of gene expression. Bioessays 2014, 36, 236–243. [Google Scholar] [CrossRef]
  66. Seo, J.; Singh, N.N.; Ottesen, E.W.; Lee, B.M.; Singh, R.N. A novel human-specific splice isoform alters the critical C-terminus of Survival Motor Neuron protein. Sci. Rep. 2016, 6, 30778. [Google Scholar] [CrossRef] [PubMed]
  67. Usuki, F.; Fujimura, M.; Yamashita, A. Endoplasmic reticulum stress preconditioning modifies intracellular mercury content by upregulating membrane transporters. Sci. Rep. 2017, 7, 12390. [Google Scholar] [CrossRef]
  68. Kim, E.; Ilagan, J.O.; Liang, Y.; Daubner, G.M.; Lee, S.C.; Ramakrishnan, A.; Li, Y.; Chung, Y.R.; Micol, J.B.; Murphy, M.E.; et al. SRSF2 Mutations Contribute to Myelodysplasia by Mutant-Specific Effects on Exon Recognition. Cancer Cell 2015, 27, 617–630. [Google Scholar] [CrossRef]
  69. Inoue, K.; Fry, E.A. Aberrant Splicing of Estrogen Receptor, HER2, and CD44 Genes in Breast Cancer. Genet. Epigenet. 2015, 7, 19–32. [Google Scholar] [CrossRef]
  70. Yae, T.; Tsuchihashi, K.; Ishimoto, T.; Motohara, T.; Yoshikawa, M.; Yoshida, G.J.; Wada, T.; Masuko, T.; Mogushi, K.; Tanaka, H.; et al. Alternative splicing of CD44 mRNA by ESRP1 enhances lung colonization of metastatic cancer cell. Nat. Commun. 2012, 3, 883. [Google Scholar] [CrossRef]
  71. Wallach-Dayan, S.B.; Rubinstein, A.M.; Hand, C.; Breuer, R.; Naor, D. DNA vaccination with CD44 variant isoform reduces mammary tumor local growth and lung metastasis. Mol. Cancer Ther. 2008, 7, 1615–1623. [Google Scholar] [CrossRef] [PubMed]
  72. Sandström, K.; Haylock, A.K.; Spiegelberg, D.; Qvarnström, F.; Wester, K.; Nestor, M. A novel CD44v6 targeting antibody fragment with improved tumor-to-blood ratio. Int. J. Oncol. 2012, 40, 1525–1532. [Google Scholar] [CrossRef] [PubMed]
  73. Chopra, A. Humanized anti-CD44v6 monoclonal antibody labeled with IRDye800CW. In Molecular Imaging and Contrast Agent Database (MICAD); National Center for Biotechnology Information: Bethesda, MD, USA, 2004. [Google Scholar]
  74. Lv, X.; Lu, X.; Cao, J.; Luo, Q.; Ding, Y.; Peng, F.; Pataer, A.; Lu, D.; Han, D.; Malmberg, E.; et al. Modulation of the proteostasis network promotes tumor resistance to oncogenic KRAS inhibitors. Science 2023, 381, eabn4180. [Google Scholar] [CrossRef]
  75. Whitley, M.J.; Tran, T.H.; Rigby, M.; Yi, M.; Dharmaiah, S.; Waybright, T.J.; Ramakrishnan, N.; Perkins, S.; Taylor, T.; Messing, S.; et al. Comparative analysis of KRAS4a and KRAS4b splice variants reveals distinctive structural and functional properties. Sci. Adv. 2024, 10, eadj4137. [Google Scholar] [CrossRef]
  76. Chen, W.C.; To, M.D.; Westcott, P.M.K.; Delrosario, R.; Kim, I.J.; Philips, M.; Tran, Q.; Bollam, S.R.; Goodarzi, H.; Bayani, N.; et al. Targeting KRAS4A splicing through the RBM39/DCAF15 pathway inhibits cancer stem cells. Nat. Commun. 2021, 12, 4288. [Google Scholar] [CrossRef]
  77. Yochum, Z.A.; Villaruz, L.C. Alternative splicing of HER2: A novel mediator of EGFR TKI resistance. Transl. Lung Cancer Res. 2020, 9, 1606–1612. [Google Scholar] [CrossRef] [PubMed]
  78. Weinholdt, C.; Wichmann, H.; Kotrba, J.; Ardell, D.H.; Kappler, M.; Eckert, A.W.; Vordermark, D.; Grosse, I. Prediction of regulatory targets of alternative isoforms of the epidermal growth factor receptor in a glioblastoma cell line. BMC Bioinform. 2019, 20, 434. [Google Scholar] [CrossRef]
  79. Takezawa, K.; Pirazzoli, V.; Arcila, M.E.; Nebhan, C.A.; Song, X.; de Stanchina, E.; Ohashi, K.; Janjigian, Y.Y.; Spitzler, P.J.; Melnick, M.A.; et al. HER2 amplification: A potential mechanism of acquired resistance to EGFR inhibition in EGFR-mutant lung cancers that lack the second-site EGFRT790M mutation. Cancer Discov. 2012, 2, 922–933. [Google Scholar] [CrossRef]
  80. Ray Das, S.; Delahunt, B.; Lasham, A.; Li, K.; Wright, D.; Print, C.; Slatter, T.; Braithwaite, A.; Mehta, S. Combining TP53 mutation and isoform has the potential to improve clinical practice. Pathology 2024, 56, 473–483. [Google Scholar] [CrossRef]
  81. Chen, J.; Zhang, D.; Qin, X.; Owzar, K.; McCann, J.J.; Kastan, M.B. DNA-Damage-Induced Alternative Splicing of p53. Cancers 2021, 13, 251. [Google Scholar] [CrossRef]
  82. Antonio-Vejar, V.; Ortiz-Sanchez, E.; Rosendo-Chalma, P.; Patino-Morales, C.C.; Guido-Jimenez, M.C.; Alvarado-Ortiz, E.; Hernandez, G.; Garcia-Carranca, A. New insights into the interactions of HPV-16 E6*I and E6*II with p53 isoforms and induction of apoptosis in cancer-derived cell lines. Pathol. Res. Pract. 2022, 234, 153890. [Google Scholar] [CrossRef] [PubMed]
  83. Legscha, K.J.; Antunes, E.; Guezguez, B.; Theobald, M.; Echchannaoui, H. p53 Isoform D133p53a: A Novel Transcriptional Enhancer of T-Cell Effector Function to Improve T-Cell Based Cancer Immunotherapy. Blood Adv. 2018, 132 (Suppl. S1), 3489. [Google Scholar] [CrossRef]
  84. Ho, J.N.; Kang, G.Y.; Lee, S.S.; Kim, J.; Bae, I.H.; Hwang, S.G.; Um, H.D. Bcl-XL and STAT3 mediate malignant actions of gamma-irradiation in lung cancer cells. Cancer Sci. 2010, 101, 1417–1423. [Google Scholar] [CrossRef] [PubMed]
  85. Wong, S.K.; Alex, D.; Bosdet, I.; Hughesman, C.; Karsan, A.; Yip, S.; Ho, C. MET exon 14 skipping mutation positive non-small cell lung cancer: Response to systemic therapy. Lung Cancer 2021, 154, 142–145. [Google Scholar] [CrossRef]
  86. Warren, C.F.A.; Wong-Brown, M.W.; Bowden, N.A. BCL-2 family isoforms in apoptosis and cancer. Cell Death Dis. 2019, 10, 177. [Google Scholar] [CrossRef] [PubMed]
  87. Long, J.; Chen, X.; Shen, Y.; Lei, Y.; Mu, L.; Wang, Z.; Xiang, R.; Gao, W.; Wang, L.; Wang, L.; et al. A combinatorial therapeutic approach to enhance FLT3-ITD AML treatment. Cell Rep. Med. 2023, 4, 101286. [Google Scholar] [CrossRef]
  88. Zheng, L.; Luthra, R.; Alvarez, H.A.; San Lucas, F.A.; Duose, D.Y.; Wistuba, I.I.; Fuller, G.N.; Ballester, L.Y.; Roy-Chowdhuri, S.; Sweeney, K.J.; et al. Intragenic EGFR::EGFR.E1E8 Fusion (EGFRvIII) in 4331 Solid Tumors. Cancers 2023, 16, 6. [Google Scholar] [CrossRef]
  89. Babic, I.; Anderson, E.S.; Tanaka, K.; Guo, D.; Masui, K.; Li, B.; Zhu, S.; Gu, Y.; Villa, G.R.; Akhavan, D.; et al. EGFR mutation-induced alternative splicing of Max contributes to growth of glycolytic tumors in brain cancer. Cell Metab. 2013, 17, 1000–1008. [Google Scholar] [CrossRef]
  90. Chaudhri, R.A.; Schwartz, N.; Elbaradie, K.; Schwartz, Z.; Boyan, B.D. Role of ERα36 in membrane-associated signaling by estrogen. Steroids 2014, 81, 74–80. [Google Scholar] [CrossRef]
  91. Chantalat, E.; Boudou, F.; Laurell, H.; Palierne, G.; Houtman, R.; Melchers, D.; Rochaix, P.; Filleron, T.; Stella, A.; Burlet-Schiltz, O.; et al. The AF-1-deficient estrogen receptor ERα46 isoform is frequently expressed in human breast tumors. Breast Cancer Res. 2016, 18, 123. [Google Scholar] [CrossRef]
  92. Anamthathmakula, P.; Kyathanahalli, C.; Ingles, J.; Hassan, S.S.; Condon, J.C.; Jeyasuria, P. Estrogen receptor alpha isoform ERdelta7 in myometrium modulates uterine quiescence during pregnancy. eBioMedicine 2019, 39, 520–530. [Google Scholar] [CrossRef] [PubMed]
  93. Guo, L.; Ke, H.; Zhang, H.; Zou, L.; Yang, Q.; Lu, X.; Zhao, L.; Jiao, B. TDP43 promotes stemness of breast cancer stem cells through CD44 variant splicing isoforms. Cell Death Dis. 2022, 13, 428. [Google Scholar] [CrossRef] [PubMed]
  94. Eilertsen, I.A.; Sveen, A.; Strømme, J.M.; Skotheim, R.I.; Nesbakken, A.; Lothe, R.A. Alternative splicing expands the prognostic impact of KRAS in microsatellite stable primary colorectal cancer. Int. J. Cancer 2019, 144, 841–847. [Google Scholar] [CrossRef]
  95. Zhang, B.; Guo, D.D.; Zheng, J.Y.; Wu, Y.A. Expression of KLF6-SV2 in colorectal cancer and its impact on proliferation and apoptosis. Eur. J. Cancer Prev. 2018, 27, 20–26. [Google Scholar] [CrossRef]
  96. Giménez-Capitán, A.; Sánchez-Herrero, E.; Robado de Lope, L.; Aguilar-Hernández, A.; Sullivan, I.; Calvo, V.; Moya-Horno, I.; Viteri, S.; Cabrera, C.; Aguado, C.; et al. Detecting ALK, ROS1, and RET fusions and the METΔex14 splicing variant in liquid biopsies of non-small-cell lung cancer patients using RNA-based techniques. Mol. Oncol. 2023, 17, 1884–1897. [Google Scholar] [CrossRef]
  97. Hsu, C.C.; Liao, B.C.; Liao, W.Y.; Markovets, A.; Stetson, D.; Thress, K.; Yang, J.C. Exon 16-Skipping HER2 as a Novel Mechanism of Osimertinib Resistance in EGFR L858R/T790M-Positive Non-Small Cell Lung Cancer. J. Thorac. Oncol. 2020, 15, 50–61. [Google Scholar] [CrossRef] [PubMed]
  98. Gudikote, J.P.; Cascone, T.; Poteete, A.; Sitthideatphaiboon, P.; Wu, Q.; Morikawa, N.; Zhang, F.; Peng, S.; Tong, P.; Li, L.; et al. Inhibition of nonsense-mediated decay rescues p53β/γ isoform expression and activates the p53 pathway in MDM2-overexpressing and select p53-mutant cancers. J. Biol. Chem. 2021, 297, 101163. [Google Scholar] [CrossRef]
  99. Senturk, S.; Yao, Z.; Camiolo, M.; Stiles, B.; Rathod, T.; Walsh, A.M.; Nemajerova, A.; Lazzara, M.J.; Altorki, N.K.; Krainer, A.; et al. p53Ψ is a transcriptionally inactive p53 isoform able to reprogram cells toward a metastatic-like state. Proc. Natl. Acad. Sci. USA 2014, 111, E3287–E3296. [Google Scholar] [CrossRef]
  100. Joruiz, S.M.; Bourdon, J.C. p53 Isoforms: Key Regulators of the Cell Fate Decision. Cold Spring Harb. Perspect. Med. 2016, 6, a026039. [Google Scholar] [CrossRef]
  101. Le Sénéchal, R.; Keruzoré, M.; Quillévéré, A.; Loaëc, N.; Dinh, V.T.; Reznichenko, O.; Guixens-Gallardo, P.; Corcos, L.; Teulade-Fichou, M.P.; Granzhan, A.; et al. Alternative splicing of BCL-x is controlled by RBM25 binding to a G-quadruplex in BCL-x pre-mRNA. Nucleic Acids Res. 2023, 51, 11239–11257. [Google Scholar] [CrossRef]
  102. Wang, Z.; Wang, S.; Qin, J.; Zhang, X.; Lu, G.; Liu, H.; Guo, H.; Wu, L.; Shender, V.O.; Shao, C.; et al. Splicing factor BUD31 promotes ovarian cancer progression through sustaining the expression of anti-apoptotic BCL2L12. Nat. Commun. 2022, 13, 6246. [Google Scholar] [CrossRef]
  103. Liang, W.C.; Wang, Y.; Xiao, L.J.; Wang, Y.B.; Fu, W.M.; Wang, W.M.; Jiang, H.Q.; Qi, W.; Wan, D.C.; Zhang, J.F.; et al. Identification of miRNAs that specifically target tumor suppressive KLF6-FL rather than oncogenic KLF6-SV1 isoform. RNA Biol. 2014, 11, 845–854. [Google Scholar] [CrossRef]
  104. Liu, X.; Zheng, Y.; Xiao, M.; Chen, X.; Zhu, Y.; Xu, C.; Wang, F.; Liu, Z.; Cao, K. SRSF10 stabilizes CDC25A by triggering exon 6 skipping to promote hepatocarcinogenesis. J. Exp. Clin. Cancer Res. 2022, 41, 353. [Google Scholar] [CrossRef]
  105. Sun, Y.; Xu, M.; Lee Wan, H.; Ding, X.; Wong, A.M.; Pu, D.; Ng, K.K.; Wong, N. Spliced exon9 ADRM1 promotes liver oncogenicity via selective degradation of tumor suppressor FBXW7. J. Hepatol. 2025. [Google Scholar] [CrossRef]
  106. Kwok, D.W.; Stevers, N.O.; Etxeberria, I.; Nejo, T.; Colton Cove, M.; Chen, L.H.; Jung, J.; Okada, K.; Lakshmanachetty, S.; Gallus, M.; et al. Tumour-wide RNA splicing aberrations generate actionable public neoantigens. Nature 2025, 639, 463–473. [Google Scholar] [CrossRef] [PubMed]
  107. Ma, X.; Dighe, A.; Maziarz, J.; Neumann, E.; Erkenbrack, E.; Hei, Y.Y.; Liu, Y.; Suhail, Y.; Pak, I.; Levchenko, A.; et al. Evolution of higher mesenchymal CD44 expression in the human lineage: A gene linked to cancer malignancy. Evol. Med. Public Health 2022, 10, 447–462. [Google Scholar] [CrossRef] [PubMed]
  108. Elliott, K.; Nilsson, J.; Van den Eynden, J. Pharmacologic RNA splicing modulation: A novel mechanism to enhance neoantigen-directed anti-tumor immunity and immunotherapy response. Signal Transduct. Target. Ther. 2021, 6, 373. [Google Scholar] [CrossRef] [PubMed]
  109. Lu, S.X.; De Neef, E.; Thomas, J.D.; Sabio, E.; Rousseau, B.; Gigoux, M.; Knorr, D.A.; Greenbaum, B.; Elhanati, Y.; Hogg, S.J.; et al. Pharmacologic modulation of RNA splicing enhances anti-tumor immunity. Cell 2021, 184, 4032–4047.e31. [Google Scholar] [CrossRef]
  110. Lo, A.; McSharry, M.; Berger, A.H. Oncogenic KRAS alters splicing factor phosphorylation and alternative splicing in lung cancer. BMC Cancer 2022, 22, 1315. [Google Scholar] [CrossRef]
  111. Lang, F.; Schrörs, B.; Löwer, M.; Türeci, Ö.; Sahin, U. Identification of neoantigens for individualized therapeutic cancer vaccines. Nat. Rev. Drug Discov. 2022, 21, 261–282. [Google Scholar] [CrossRef]
  112. Fuentes-Rodriguez, A.; Mitchell, A.; Guérin, S.L.; Landreville, S. Recent Advances in Molecular and Genetic Research on Uveal Melanoma. Cells 2024, 13, 1023. [Google Scholar] [CrossRef]
  113. Cai, Y.; Chen, R.; Gao, S.; Li, W.; Liu, Y.; Su, G.; Song, M.; Jiang, M.; Jiang, C.; Zhang, X. Artificial intelligence applied in neoantigen identification facilitates personalized cancer immunotherapy. Front. Oncol. 2022, 12, 1054231. [Google Scholar] [CrossRef] [PubMed]
  114. Peltier, D.C.; Roberts, A.; Reddy, P. LNCing RNA to immunity. Trends Immunol. 2022, 43, 478–495. [Google Scholar] [CrossRef]
  115. Pilger, D.; Seymour, L.W.; Jackson, S.P. Interfaces between cellular responses to DNA damage and cancer immunotherapy. Genes Dev. 2021, 35, 602–618. [Google Scholar] [CrossRef] [PubMed]
  116. Pelizzaro, F.; Farinati, F.; Trevisani, F. Immune Checkpoint Inhibitors in Hepatocellular Carcinoma: Current Strategies and Biomarkers Predicting Response and/or Resistance. Biomedicines 2023, 11, 1020. [Google Scholar] [CrossRef] [PubMed]
  117. Huang, X.; Chi, H.; Gou, S.; Guo, X.; Li, L.; Peng, G.; Zhang, J.; Xu, J.; Nian, S.; Yuan, Q. An Aggrephagy-Related LncRNA Signature for the Prognosis of Pancreatic Adenocarcinoma. Genes 2023, 14, 124. [Google Scholar] [CrossRef]
  118. Hugo, W.; Zaretsky, J.M.; Sun, L.; Song, C.; Moreno, B.H.; Hu-Lieskovan, S.; Berent-Maoz, B.; Pang, J.; Chmielowski, B.; Cherry, G.; et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 2016, 165, 35–44. [Google Scholar] [CrossRef]
  119. Liu, L.; Sun, J.; Zhong, C.; Zhang, A.; Wang, G.; Chen, S.; Zhang, S.; Wang, M.; Li, L. Identification of a fatty acid metabolism-related gene signature to predict prognosis in stomach adenocarcinoma. Aging 2024, 16, 8552–8571. [Google Scholar] [CrossRef]
  120. Zhang, Y.; Qian, J.; Gu, C.; Yang, Y. Alternative splicing and cancer: A systematic review. Signal Transduct. Target. Ther. 2021, 6, 78. [Google Scholar] [CrossRef]
  121. Advani, R.; Luzzi, S.; Scott, E.; Dalgliesh, C.; Weischenfeldt, J.; Munkley, J.; Elliott, D.J. Epithelial specific splicing regulator proteins as emerging oncogenes in aggressive prostate cancer. Oncogene 2023, 42, 3161–3168. [Google Scholar] [CrossRef]
  122. Pan, J.; Zhou, T.; Na, K.; Xu, K.; Yan, C.; Song, H.; Han, Y. Identification of hub modules and therapeutic targets associated with CD8(+)T-cells in HF and their pan-cancer analysis. Sci. Rep. 2024, 14, 18823. [Google Scholar] [CrossRef]
  123. Jiang, H.; Awuti, G.; Guo, X. Construction of an Immunophenoscore-Related Signature for Evaluating Prognosis and Immunotherapy Sensitivity in Ovarian Cancer. ACS Omega 2023, 8, 33017–33031. [Google Scholar] [CrossRef] [PubMed]
  124. Shi, X.; Wang, P.; Li, Y.; Xu, J.; Yin, T.; Teng, F. Using MRI radiomics to predict the efficacy of immunotherapy for brain metastasis in patients with small cell lung cancer. Thorac. Cancer 2024, 15, 738–748. [Google Scholar] [CrossRef] [PubMed]
  125. Liang, Y.; Wang, L.; Ma, P.; Ju, D.; Zhao, M.; Shi, Y. Enhancing anti-tumor immune responses through combination therapies: Epigenetic drugs and immune checkpoint inhibitors. Front. Immunol. 2023, 14, 1308264. [Google Scholar] [CrossRef] [PubMed]
  126. Tripodi, L.; Sasso, E.; Feola, S.; Coluccino, L.; Vitale, M.; Leoni, G.; Szomolay, B.; Pastore, L.; Cerullo, V. Systems Biology Approaches for the Improvement of Oncolytic Virus-Based Immunotherapies. Cancers 2023, 15, 1297. [Google Scholar] [CrossRef]
  127. Boussaad, I.; Obermaier, C.D.; Hanss, Z.; Bobbili, D.R.; Bolognin, S.; Glaab, E.; Wołyńska, K.; Weisschuh, N.; De Conti, L.; May, C.; et al. A patient-based model of RNA mis-splicing uncovers treatment targets in Parkinson’s disease. Sci. Transl. Med. 2020, 12, eaau3960. [Google Scholar] [CrossRef]
  128. Matsushima, S.; Ajiro, M.; Iida, K.; Chamoto, K.; Honjo, T.; Hagiwara, M. Chemical induction of splice-neoantigens attenuates tumor growth in a preclinical model of colorectal cancer. Sci. Transl. Med. 2022, 14, eabn6056. [Google Scholar] [CrossRef]
  129. Gordon, E.D.; Simpson, L.J.; Rios, C.L.; Ringel, L.; Lachowicz-Scroggins, M.E.; Peters, M.C.; Wesolowska-Andersen, A.; Gonzalez, J.R.; MacLeod, H.J.; Christian, L.S.; et al. Alternative splicing of interleukin-33 and type 2 inflammation in asthma. Proc. Natl. Acad. Sci. USA 2016, 113, 8765–8770. [Google Scholar] [CrossRef]
  130. Ren, P.; Lu, L.; Cai, S.; Chen, J.; Lin, W.; Han, F. Alternative Splicing: A New Cause and Potential Therapeutic Target in Autoimmune Disease. Front. Immunol. 2021, 12, 713540. [Google Scholar] [CrossRef]
  131. Reiter, S.; Schroeder, C.; Broche, J.; Sinnberg, T.; Bonzheim, I.; Süsskind, D.; Flatz, L.; Forschner, A. Successful treatment of metastatic uveal melanoma with ipilimumab and nivolumab after severe progression under tebentafusp: A case report. Front. Oncol. 2023, 13, 1167791. [Google Scholar] [CrossRef]
  132. 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] [PubMed]
  133. Kim, H.K.; Joung, J.G.; Choi, Y.L.; Lee, S.H.; Park, B.J.; Choi, Y.S.; Ryu, D.; Nam, J.Y.; Lee, M.S.; Park, W.Y.; et al. Earlier-Phased Cancer Immunity Cycle Strongly Influences Cancer Immunity in Operable Never-Smoker Lung Adenocarcinoma. iScience 2020, 23, 101386. [Google Scholar] [CrossRef] [PubMed]
  134. Yamamoto, K.; Venida, A.; Yano, J.; Biancur, D.E.; Kakiuchi, M.; Gupta, S.; Sohn, A.S.W.; Mukhopadhyay, S.; Lin, E.Y.; Parker, S.J.; et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature 2020, 581, 100–105. [Google Scholar] [CrossRef]
  135. Müller, L.; Kschischo, M.; Vokuhl, C.; Stahl, D.; Gütgemann, I. Stemness Correlates Inversely with MHC Class I Expression in Pediatric Small Round Blue Cell Tumors. Cancers 2022, 14, 3584. [Google Scholar] [CrossRef]
  136. Fu, X.; Liu, S.; Cao, D.; Li, C.; Ji, H.; Wang, G. Med23 deficiency reprograms the tumor microenvironment to promote lung tumorigenesis. Br. J. Cancer 2024, 130, 716–727. [Google Scholar] [CrossRef]
  137. Niu, M.; Liu, Y.; Yi, M.; Jiao, D.; Wu, K. Biological Characteristics and Clinical Significance of Soluble PD-1/PD-L1 and Exosomal PD-L1 in Cancer. Front. Immunol. 2022, 13, 827921. [Google Scholar] [CrossRef] [PubMed]
  138. Maebele, L.T.; Mulaudzi, T.V.; Yasasve, M.; Dlamini, Z.; Damane, B.P. Immunomodulatory Gene-Splicing Dysregulation in Tumorigenesis: Unmasking the Complexity. Molecules 2023, 28, 5984. [Google Scholar] [CrossRef]
  139. Raza, A.; Mohsen, R.; Kanbour, A.; Zar Gul, A.R.; Philip, A.; Vijayakumar, S.; Hydrose, S.; Prabhu, K.S.; Al-Suwaidi, A.K.; Inchakalody, V.P.; et al. Serum immune mediators as novel predictors of response to anti-PD-1/PD-L1 therapy in non-small cell lung cancer patients with high tissue-PD-L1 expression. Front. Immunol. 2023, 14, 1157100. [Google Scholar] [CrossRef]
  140. Hassounah, N.B.; Malladi, V.S.; Huang, Y.; Freeman, S.S.; Beauchamp, E.M.; Koyama, S.; Souders, N.; Martin, S.; Dranoff, G.; Wong, K.K.; et al. Identification and characterization of an alternative cancer-derived PD-L1 splice variant. Cancer Immunol. Immunother. 2019, 68, 407–420. [Google Scholar] [CrossRef]
  141. Gong, B.; Kiyotani, K.; Sakata, S.; Nagano, S.; Kumehara, S.; Baba, S.; Besse, B.; Yanagitani, N.; Friboulet, L.; Nishio, M.; et al. Secreted PD-L1 variants mediate resistance to PD-L1 blockade therapy in non-small cell lung cancer. J. Exp. Med. 2019, 216, 982–1000. [Google Scholar] [CrossRef]
  142. Incorvaia, L.; Fanale, D.; Badalamenti, G.; Porta, C.; Olive, D.; De Luca, I.; Brando, C.; Rizzo, M.; Messina, C.; Rediti, M.; et al. Baseline plasma levels of soluble PD-1, PD-L1, and BTN3A1 predict response to nivolumab treatment in patients with metastatic renal cell carcinoma: A step toward a biomarker for therapeutic decisions. Oncoimmunology 2020, 9, 1832348. [Google Scholar] [CrossRef]
  143. Bernard, A.; Boidot, R.; Végran, F. Alternative Splicing in Cancer and Immune Cells. Cancers 2022, 14, 1726. [Google Scholar] [CrossRef]
  144. Dursun, H.G.; Yılmaz, H.O.; Dursun, R.; Kulaksızoğlu, S. Association of Cytotoxic T Lymphocyte Antigen-4 Gene Polymorphisms with Psoriasis Vulgaris: A Case-Control Study in Turkish Population. J. Immunol. Res. 2018, 2018, 1643906. [Google Scholar] [CrossRef]
  145. Zhao, C.; Zhao, J.W.; Zhang, Y.H.; Zhu, Y.D.; Yang, Z.Y.; Liu, S.L.; Tang, Q.Y.; Yang, Y.; Wang, H.K.; Shu, Y.J.; et al. PTBP3 Mediates IL-18 Exon Skipping to Promote Immune Escape in Gallbladder Cancer. Adv. Sci. 2024, 11, e2406633. [Google Scholar] [CrossRef]
  146. Angarola, B.L.; Anczuków, O. Splicing alterations in healthy aging and disease. Wiley Interdiscip. Rev. RNA 2021, 12, e1643. [Google Scholar] [CrossRef]
  147. Martinez, N.M.; Lynch, K.W. Control of alternative splicing in immune responses: Many regulators, many predictions, much still to learn. Immunol. Rev. 2013, 253, 216–236. [Google Scholar] [CrossRef]
  148. Leowattana, W.; Leowattana, P.; Leowattana, T. Systemic treatments for resectable carcinoma of the esophagus. World J. Gastroenterol. 2023, 29, 4628–4641. [Google Scholar] [CrossRef]
  149. Furmanski, A.L.; Barbarulo, A.; Solanki, A.; Lau, C.I.; Sahni, H.; Saldana, J.I.; D’Acquisto, F.; Crompton, T. The transcriptional activator Gli2 modulates T-cell receptor signalling through attenuation of AP-1 and NFκB activity. J. Cell Sci. 2015, 128, 2085–2095. [Google Scholar] [CrossRef]
  150. Conserva, M.R.; Redavid, I.; Anelli, L.; Zagaria, A.; Tarantini, F.; Cumbo, C.; Tota, G.; Parciante, E.; Coccaro, N.; Minervini, C.F.; et al. IKAROS in Acute Leukemia: A Positive Influencer or a Mean Hater? Int. J. Mol. Sci. 2023, 24, 3282. [Google Scholar] [CrossRef]
  151. McNamee, E.N.; Korns Johnson, D.; Homann, D.; Clambey, E.T. Hypoxia and hypoxia-inducible factors as regulators of T cell development, differentiation, and function. Immunol. Res. 2013, 55, 58–70. [Google Scholar] [CrossRef]
  152. Du, J.; Wang, Q.; Yang, S.; Chen, S.; Fu, Y.; Spath, S.; Domeier, P.; Hagin, D.; Anover-Sombke, S.; Haouili, M.; et al. FOXP3 exon 2 controls T(reg) stability and autoimmunity. Sci. Immunol. 2022, 7, eabo5407. [Google Scholar] [CrossRef]
  153. Legrand, J.M.D.; Chan, A.L.; La, H.M.; Rossello, F.J.; Änkö, M.L.; Fuller-Pace, F.V.; Hobbs, R.M. DDX5 plays essential transcriptional and post-transcriptional roles in the maintenance and function of spermatogonia. Nat. Commun. 2019, 10, 2278. [Google Scholar] [CrossRef] [PubMed]
  154. Roganowicz, M.; Bär, D.; Bersaglieri, C.; Aprigliano, R.; Santoro, R. BAZ2A-RNA mediated association with TOP2A and KDM1A represses genes implicated in prostate cancer. Life Sci. Alliance 2023, 6, e202301950. [Google Scholar] [CrossRef]
  155. Mailer, R.K.; Joly, A.L.; Liu, S.; Elias, S.; Tegner, J.; Andersson, J. IL-1beta promotes Th17 differentiation by inducing alternative splicing of FOXP3. Sci. Rep. 2015, 5, 14674. [Google Scholar] [CrossRef] [PubMed]
  156. Ding, R.; Yu, X.; Hu, Z.; Dong, Y.; Huang, H.; Zhang, Y.; Han, Q.; Ni, Z.Y.; Zhao, R.; Ye, Y.; et al. Lactate modulates RNA splicing to promote CTLA-4 expression in tumor-infiltrating regulatory T cells. Immunity 2024, 57, 528–540.e6. [Google Scholar] [CrossRef] [PubMed]
  157. Marima, R.; Basera, A.; Miya, T.; Damane, B.P.; Kandhavelu, J.; Mirza, S.; Penny, C.; Dlamini, Z. Exosomal long non-coding RNAs in cancer: Interplay, modulation, and therapeutic avenues. Noncoding RNA Res. 2024, 9, 887–900. [Google Scholar] [CrossRef]
  158. Lin, H.; Li, C.; Zhang, W.; Wu, B.; Wang, Y.; Wang, S.; Wang, D.; Li, X.; Huang, H. Synthetic Cells and Molecules in Cellular Immunotherapy. Int. J. Biol. Sci. 2024, 20, 2833–2859. [Google Scholar] [CrossRef]
  159. Hwang, A.; Mehra, V.; Chhetri, J.; Ali, S.; Tran, M.; Roddie, C. Current Treatment Options for Renal Cell Carcinoma: Focus on Cell-Based Immunotherapy. Cancers 2024, 16, 1209. [Google Scholar] [CrossRef]
  160. Yu, G.; Wang, W.; He, X.; Xu, J.; Xu, R.; Wan, T.; Wu, Y. Synergistic Therapeutic Effects of Low Dose Decitabine and NY-ESO-1 Specific TCR-T Cells for the Colorectal Cancer with Microsatellite Stability. Front. Oncol. 2022, 12, 895103. [Google Scholar] [CrossRef]
  161. Lu, F.; Ma, X.J.; Jin, W.L.; Luo, Y.; Li, X. Neoantigen Specific T Cells Derived from T Cell-Derived Induced Pluripotent Stem Cells for the Treatment of Hepatocellular Carcinoma: Potential and Challenges. Front. Immunol. 2021, 12, 690565. [Google Scholar] [CrossRef]
  162. Filin, I.Y.; Mayasin, Y.P.; Kharisova, C.B.; Gorodilova, A.V.; Kitaeva, K.V.; Chulpanova, D.S.; Solovyeva, V.V.; Rizvanov, A.A. Cell Immunotherapy against Melanoma: Clinical Trials Review. Int. J. Mol. Sci. 2023, 24, 2413. [Google Scholar] [CrossRef] [PubMed]
  163. Feng, Y.; He, C.; Liu, C.; Shao, B.; Wang, D.; Wu, P. Exploring the Complexity and Promise of Tumor Immunotherapy in Drug Development. Int. J. Mol. Sci. 2024, 25, 6444. [Google Scholar] [CrossRef]
  164. Nejo, T.; Wang, L.; Leung, K.K.; Wang, A.; Lakshmanachetty, S.; Gallus, M.; Kwok, D.W.; Hong, C.; Chen, L.H.; Carrera, D.A.; et al. Challenges in the discovery of tumor-specific alternative splicing-derived cell-surface antigens in glioma. Sci. Rep. 2024, 14, 6362. [Google Scholar] [CrossRef] [PubMed]
  165. Choi, N.; Jang, H.N.; Oh, J.; Ha, J.; Park, H.; Zheng, X.; Lee, S.; Shen, H. SRSF6 Regulates the Alternative Splicing of the Apoptotic Fas Gene by Targeting a Novel RNA Sequence. Cancers 2022, 14, 1990. [Google Scholar] [CrossRef]
  166. Izquierdo, J.M.; Majós, N.; Bonnal, S.; Martínez, C.; Castelo, R.; Guigó, R.; Bilbao, D.; Valcárcel, J. Regulation of Fas alternative splicing by antagonistic effects of TIA-1 and PTB on exon definition. Mol. Cell 2005, 19, 475–484. [Google Scholar] [CrossRef] [PubMed]
  167. Takeiwa, T.; Ikeda, K.; Horie, K.; Inoue, S. Role of RNA binding proteins of the Drosophila behavior and human splicing (DBHS) family in health and cancer. RNA Biol. 2024, 21, 1–17. [Google Scholar] [CrossRef]
  168. Ji, C.; Bao, L.; Yuan, S.; Qi, Z.; Wang, F.; You, M.; Yu, G.; Liu, J.; Cui, X.; Wang, Z.; et al. SRSF1 Deficiency Impairs the Late Thymocyte Maturation and the CD8 Single-Positive Lineage Fate Decision. Front. Immunol. 2022, 13, 838719. [Google Scholar] [CrossRef]
  169. Lee, K.; Hyung, D.; Cho, S.Y.; Yu, N.; Hong, S.; Kim, J.; Kim, S.; Han, J.Y.; Park, C. Splicing signature database development to delineate cancer pathways using literature mining and transcriptome machine learning. Comput. Struct. Biotechnol. J. 2023, 21, 1978–1988. [Google Scholar] [CrossRef]
  170. Mickleburgh, I.; Kafasla, P.; Cherny, D.; Llorian, M.; Curry, S.; Jackson, R.J.; Smith, C.W. The organization of RNA contacts by PTB for regulation of FAS splicing. Nucleic Acids Res. 2014, 42, 8605–8620. [Google Scholar] [CrossRef]
  171. Chen, T.; Dai, X.; Dai, J.; Ding, C.; Zhang, Z.; Lin, Z.; Hu, J.; Lu, M.; Wang, Z.; Qi, Y.; et al. AFP promotes HCC progression by suppressing the HuR-mediated Fas/FADD apoptotic pathway. Cell Death Dis. 2020, 11, 822. [Google Scholar] [CrossRef]
  172. Gu, Z.; Yang, Y.; Ma, Q.; Wang, H.; Zhao, S.; Qi, Y.; Li, Y. HNRNPC, a predictor of prognosis and immunotherapy response based on bioinformatics analysis, is related to proliferation and invasion of NSCLC cells. Respir. Res. 2022, 23, 362. [Google Scholar] [CrossRef] [PubMed]
  173. Soni, K.; Martínez-Lumbreras, S.; Sattler, M. Conformational Dynamics from Ambiguous Zinc Coordination in the RanBP2-Type Zinc Finger of RBM5. J. Mol. Biol. 2020, 432, 4127–4138. [Google Scholar] [CrossRef]
  174. Cheng, Y.; Li, L.; Wei, X.; Xu, F.; Huang, X.; Qi, F.; Zhang, Y.; Li, X. HNRNPC suppresses tumor immune microenvironment by activating Treg cells promoting the progression of prostate cancer. Cancer Sci. 2023, 114, 1830–1845. [Google Scholar] [CrossRef] [PubMed]
  175. Nagasawa, C.K.; Bailey, A.O.; Russell, W.K.; Garcia-Blanco, M.A. Inefficient recruitment of DDX39B impedes pre-spliceosome assembly on FOXP3 introns. RNA 2024, 30, 824–838. [Google Scholar] [CrossRef]
  176. Subramani, P.G.; Fraszczak, J.; Helness, A.; Estall, J.L.; Möröy, T.; Di Noia, J.M. Conserved role of hnRNPL in alternative splicing of epigenetic modifiers enables B cell activation. EMBO Rep. 2024, 25, 2662–2697. [Google Scholar] [CrossRef] [PubMed]
  177. Szelest, M.; Giannopoulos, K. Biological relevance of alternative splicing in hematologic malignancies. Mol. Med. 2024, 30, 62. [Google Scholar] [CrossRef]
  178. Liu, Z.; Yoshimi, A.; Wang, J.; Cho, H.; Chun-Wei Lee, S.; Ki, M.; Bitner, L.; Chu, T.; Shah, H.; Liu, B.; et al. Mutations in the RNA Splicing Factor SF3B1 Promote Tumorigenesis through MYC Stabilization. Cancer Discov. 2020, 10, 806–821. [Google Scholar] [CrossRef]
  179. Tangye, S.G.; Nguyen, T.; Deenick, E.K.; Bryant, V.L.; Ma, C.S. Inborn errors of human B cell development, differentiation, and function. J. Exp. Med. 2023, 220, e20221105. [Google Scholar] [CrossRef]
  180. Escherich, C.; Chen, W.; Miyamoto, S.; Namikawa, Y.; Yang, W.; Teachey, D.T.; Li, Z.; Raetz, E.A.; Larsen, E.; Devidas, M.; et al. Identification of TCF3 germline variants in pediatric B-cell acute lymphoblastic leukemia. Blood Adv. 2023, 7, 2177–2180. [Google Scholar] [CrossRef]
  181. Bruijnesteijn, J.; van der Wiel, M.K.H.; de Groot, N.; Otting, N.; de Vos-Rouweler, A.J.M.; Lardy, N.M.; de Groot, N.G.; Bontrop, R.E. Extensive Alternative Splicing of KIR Transcripts. Front. Immunol. 2018, 9, 2846. [Google Scholar] [CrossRef]
  182. Geng, G.; Xu, C.; Peng, N.; Li, Y.; Liu, J.; Wu, J.; Liang, J.; Zhu, Y.; Shi, L. PTBP1 is necessary for dendritic cells to regulate T-cell homeostasis and antitumour immunity. Immunology 2021, 163, 74–85. [Google Scholar] [CrossRef]
  183. Chen, W.; Geng, D.; Chen, J.; Han, X.; Xie, Q.; Guo, G.; Chen, X.; Zhang, W.; Tang, S.; Zhong, X. Roles and mechanisms of aberrant alternative splicing in melanoma—Implications for targeted therapy and immunotherapy resistance. Cancer Cell Int. 2024, 24, 101. [Google Scholar] [CrossRef] [PubMed]
  184. Ando, T.; Okamoto, K.; Ueda, Y.; Kataoka, N.; Shintani, T.; Yanamoto, S.; Miyauchi, M.; Kajiya, M. YAP/TAZ interacts with RBM39 to confer resistance against indisulam. Oncogenesis 2024, 13, 25. [Google Scholar] [CrossRef] [PubMed]
  185. Donisi, C.; Pretta, A.; Pusceddu, V.; Ziranu, P.; Lai, E.; Puzzoni, M.; Mariani, S.; Massa, E.; Madeddu, C.; Scartozzi, M. Immunotherapy and Cancer: The Multi-Omics Perspective. Int. J. Mol. Sci. 2024, 25, 3563. [Google Scholar] [CrossRef]
  186. Wang, B.; Han, Y.; Zhang, Y.; Zhao, Q.; Wang, H.; Wei, J.; Meng, L.; Xin, Y.; Jiang, X. Overcoming acquired resistance to cancer immune checkpoint therapy: Potential strategies based on molecular mechanisms. Cell Biosci. 2023, 13, 120. [Google Scholar] [CrossRef] [PubMed]
  187. Cao, J.; Yang, X.; Chen, S.; Wang, J.; Fan, X.; Fu, S.; Yang, L. The predictive efficacy of tumor mutation burden in immunotherapy across multiple cancer types: A meta-analysis and bioinformatics analysis. Transl. Oncol. 2022, 20, 101375. [Google Scholar] [CrossRef]
  188. Bauman, J.A.; Li, S.D.; Yang, A.; Huang, L.; Kole, R. Anti-tumor activity of splice-switching oligonucleotides. Nucleic Acids Res. 2010, 38, 8348–8356. [Google Scholar] [CrossRef]
  189. Sen, S.; Talukdar, I.; Webster, N.J. SRp20 and CUG-BP1 modulate insulin receptor exon 11 alternative splicing. Mol. Cell Biol. 2009, 29, 871–880. [Google Scholar] [CrossRef]
  190. Khurshid, S.; Venkataramany, A.S.; Montes, M.; Kipp, J.F.; Roberts, R.D.; Wein, N.; Rigo, F.; Wang, P.Y.; Cripe, T.P.; Chandler, D.S. Employing splice-switching oligonucleotides and AAVrh74.U7 snRNA to target insulin receptor splicing and cancer hallmarks in osteosarcoma. Mol. Ther. Oncol. 2024, 32, 200908. [Google Scholar] [CrossRef]
  191. Xu, Y.; Spear, S.; Ma, Y.; Lorentzen, M.P.; Gruet, M.; McKinney, F.; Xu, Y.; Wickremesinghe, C.; Shepherd, M.R.; McNeish, I.; et al. Pharmacological depletion of RNA splicing factor RBM39 by indisulam synergizes with PARP inhibitors in high-grade serous ovarian carcinoma. Cell Rep. 2023, 42, 113307. [Google Scholar] [CrossRef]
  192. Paik, P.K.; Felip, E.; Veillon, R.; Sakai, H.; Cortot, A.B.; Garassino, M.C.; Mazieres, J.; Viteri, S.; Senellart, H.; Van Meerbeeck, J.; et al. Tepotinib in Non-Small-Cell Lung Cancer with MET Exon 14 Skipping Mutations. N. Engl. J. Med. 2020, 383, 931–943. [Google Scholar] [CrossRef] [PubMed]
  193. Calderon-Aparicio, A.; Wang, B.D. Prostate cancer: Alternatively spliced mRNA transcripts in tumor progression and their uses as therapeutic targets. Int. J. Biochem. Cell Biol. 2021, 141, 106096. [Google Scholar] [CrossRef] [PubMed]
  194. Mossmann, D.; Müller, C.; Park, S.; Ryback, B.; Colombi, M.; Ritter, N.; Weißenberger, D.; Dazert, E.; Coto-Llerena, M.; Nuciforo, S.; et al. Arginine reprograms metabolism in liver cancer via RBM39. Cell 2023, 186, 5068–5083.e23. [Google Scholar] [CrossRef]
  195. Huang, L.; Zeng, X.; Ma, H.; Yang, Y.; Akimoto, Y.; Wei, G.; Ni, T. Pan-Cancer Profiling of Intron Retention and Its Clinical Significance in Diagnosis and Prognosis. Cancers 2023, 15, 5689. [Google Scholar] [CrossRef]
  196. Li, G.; Iyer, B.; Prasath, V.B.S.; Ni, Y.; Salomonis, N. DeepImmuno: Deep learning-empowered prediction and generation of immunogenic peptides for T-cell immunity. Brief. Bioinform. 2021, 22, bbab160. [Google Scholar] [CrossRef] [PubMed]
  197. Shen, S.; Park, J.W.; Lu, Z.X.; Lin, L.; Henry, M.D.; Wu, Y.N.; Zhou, Q.; Xing, Y. rMATS: Robust and flexible detection of differential alternative splicing from replicate RNA-Seq data. Proc. Natl. Acad. Sci. USA 2014, 111, E5593–E5601. [Google Scholar] [CrossRef]
  198. Sterne-Weiler, T.; Weatheritt, R.J.; Best, A.J.; Ha, K.C.H.; Blencowe, B.J. Efficient and Accurate Quantitative Profiling of Alternative Splicing Patterns of Any Complexity on a Laptop. Mol. Cell 2018, 72, 187–200.e6. [Google Scholar] [CrossRef]
  199. Saraiva-Agostinho, N.; Barbosa-Morais, N.L. psichomics: Graphical application for alternative splicing quantification and analysis. Nucleic Acids Res. 2019, 47, e7. [Google Scholar] [CrossRef]
  200. Database Resources of the National Genomics Data Center, China National Center for Bioinformation in 2023. Nucleic Acids Res. 2023, 51, D18–D28. [CrossRef]
  201. Li, G.; Mahajan, S.; Ma, S.; Jeffery, E.D.; Zhang, X.; Bhattacharjee, A.; Venkatasubramanian, M.; Weirauch, M.T.; Miraldi, E.R.; Grimes, H.L.; et al. Splicing neoantigen discovery with SNAF reveals shared targets for cancer immunotherapy. Sci. Transl. Med. 2024, 16, eade2886. [Google Scholar] [CrossRef]
  202. Pan, Y.; Phillips, J.W.; Zhang, B.D.; Noguchi, M.; Kutschera, E.; McLaughlin, J.; Nesterenko, P.A.; Mao, Z.; Bangayan, N.J.; Wang, R.; et al. IRIS: Discovery of cancer immunotherapy targets arising from pre-mRNA alternative splicing. Proc. Natl. Acad. Sci. USA 2023, 120, e2221116120. [Google Scholar] [CrossRef] [PubMed]
  203. Wang, L.; Shamardani, K.; Babikir, H.; Catalan, F.; Nejo, T.; Chang, S.; Phillips, J.J.; Okada, H.; Diaz, A.A. The evolution of alternative splicing in glioblastoma under therapy. Genome Biol. 2021, 22, 48. [Google Scholar] [CrossRef] [PubMed]
  204. Müller, M.; Huber, F.; Arnaud, M.; Kraemer, A.I.; Altimiras, E.R.; Michaux, J.; Taillandier-Coindard, M.; Chiffelle, J.; Murgues, B.; Gehret, T.; et al. Machine learning methods and harmonized datasets improve immunogenic neoantigen prediction. Immunity 2023, 56, 2650–2663.e6. [Google Scholar] [CrossRef]
  205. Yang, R.L.; Qian, G.L.; Wu, D.W.; Miao, J.K.; Yang, X.; Wu, B.Q.; Yan, Y.Q.; Li, H.B.; Mao, X.M.; He, J.; et al. A multicenter prospective study of next-generation sequencing-based newborn screening for monogenic genetic diseases in China. World J. Pediatr. 2023, 19, 663–673. [Google Scholar] [CrossRef] [PubMed]
  206. Lin, J.; Ngiam, K.Y. How data science and AI-based technologies impact genomics. Singap. Med. J. 2023, 64, 59–66. [Google Scholar] [CrossRef]
  207. Zhang, X.; Zhao, C.; Shao, M.W.; Chen, Y.L.; Liu, P.; Chen, G.Q. The roadmap of bioeconomy in China. Eng. Biol. 2022, 6, 71–81. [Google Scholar] [CrossRef]
  208. Hashemi, S.; Hashemi, S.E.; Lien, K.M.; Lamb, J.J. Molecular Microbial Community Analysis as an Analysis Tool for Optimal Biogas Production. Microorganisms 2021, 9, 1162. [Google Scholar] [CrossRef]
  209. Chokr, N.; Pine, A.B.; Bewersdorf, J.P.; Shallis, R.M.; Stahl, M.; Zeidan, A.M. Getting personal with myelodysplastic syndromes: Is now the right time? Expert Rev. Hematol. 2019, 12, 215–224. [Google Scholar] [CrossRef]
  210. Zhao, Y.C.; Ding, Y.Z.; Zhao, X.; Fu, G.W.; Huang, M.J.; Li, X.X.; Sun, Q.Q.; Kan, Y.B.; Li, J.; Wang, S.L.; et al. Role and Clinical Application of Metagenomic Next-Generation Sequencing in Immunocompromised Patients with Acute Respiratory Failure During Veno-Venous Extracorporeal Membrane Oxygenation. Front. Cell Infect. Microbiol. 2022, 12, 877205. [Google Scholar] [CrossRef]
  211. Peng, W.; Cui, S.; Song, C. One-time-pad cipher algorithm based on confusion mapping and DNA storage technology. PLoS ONE 2021, 16, e0245506. [Google Scholar] [CrossRef]
  212. Ma, J.; Xiang, Y.; Xiong, Y.; Lin, Z.; Xue, Y.; Mao, M.; Sun, L.; Zhou, Y.; Li, X.; Huang, Z. SMRT sequencing analysis reveals the full-length transcripts and alternative splicing patterns in Ananas comosus var. bracteatus. PeerJ 2019, 7, e7062. [Google Scholar] [CrossRef]
  213. Zhang, F.; Chen, J.Y. A method for identifying discriminative isoform-specific peptides for clinical proteomics application. BMC Genomics 2016, 17 (Suppl. 7), 522. [Google Scholar] [CrossRef]
  214. Liu, H.; Liu, H.; Wang, L.; Song, L.; Jiang, G.; Lu, Q.; Yang, T.; Peng, H.; Cai, R.; Zhao, X.; et al. Cochlear transcript diversity and its role in auditory functions implied by an otoferlin short isoform. Nat. Commun. 2023, 14, 3085. [Google Scholar] [CrossRef]
  215. Chang, J.; Zhang, Y.; Zhou, T.; Qiao, Q.; Shan, J.; Chen, Y.; Jiang, W.; Wang, Y.; Liu, S.; Wang, Y.; et al. RBM10 C761Y mutation induced oncogenic ASPM isoforms and regulated β-catenin signaling in cholangiocarcinoma. J. Exp. Clin. Cancer Res. 2024, 43, 104. [Google Scholar] [CrossRef] [PubMed]
  216. Su, T.; Hollas, M.A.R.; Fellers, R.T.; Kelleher, N.L. Identification of Splice Variants and Isoforms in Transcriptomics and Proteomics. Annu. Rev. Biomed. Data Sci. 2023, 6, 357–376. [Google Scholar] [CrossRef] [PubMed]
  217. Trincado, J.L.; Entizne, J.C.; Hysenaj, G.; Singh, B.; Skalic, M.; Elliott, D.J.; Eyras, E. SUPPA2: Fast, accurate, and uncertainty-aware differential splicing analysis across multiple conditions. Genome Biol. 2018, 19, 40. [Google Scholar] [CrossRef] [PubMed]
  218. Li, Y.I.; Knowles, D.A.; Humphrey, J.; Barbeira, A.N.; Dickinson, S.P.; Im, H.K.; Pritchard, J.K. Annotation-free quantification of RNA splicing using LeafCutter. Nat. Genet. 2018, 50, 151–158. [Google Scholar] [CrossRef]
  219. 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.; et al. pVACtools: A Computational Toolkit to Identify and Visualize Cancer Neoantigens. Cancer Immunol. Res. 2020, 8, 409–420. [Google Scholar] [CrossRef]
  220. O’Donnell, T.J.; Rubinsteyn, A.; Laserson, U. MHCflurry 2.0: Improved Pan-Allele Prediction of MHC Class I-Presented Peptides by Incorporating Antigen Processing. Cell Syst. 2020, 11, 42–48.e7. [Google Scholar] [CrossRef]
Figure 1. Normal and abnormal alternative splicing. Alternative splicing of the Fas gene was used as an example to compare normal and abnormal alternative splicing. Normal alternative splicing: The splicing factor SFSR6 binds to the UGCCAA region in exon 6 of the Fas gene, promoting the inclusion of exon 6. As a result, it is translated into a fully functional Fas protein, which promotes the apoptosis of tumor cells through the Fas-FasL pathway. Abnormal alternative splicing: The splicing factor PTB binds to the uridine-rich sequence located in exon 6 (URE6) of the CUCUCU region of the Fas gene, promoting the skipping of exon 6. Consequently, it is translated into the FASΔ6 protein. Moreover, this protein competes with the fully functional Fas protein for binding to FasL, thus facilitating the immune escape of tumors.
Figure 1. Normal and abnormal alternative splicing. Alternative splicing of the Fas gene was used as an example to compare normal and abnormal alternative splicing. Normal alternative splicing: The splicing factor SFSR6 binds to the UGCCAA region in exon 6 of the Fas gene, promoting the inclusion of exon 6. As a result, it is translated into a fully functional Fas protein, which promotes the apoptosis of tumor cells through the Fas-FasL pathway. Abnormal alternative splicing: The splicing factor PTB binds to the uridine-rich sequence located in exon 6 (URE6) of the CUCUCU region of the Fas gene, promoting the skipping of exon 6. Consequently, it is translated into the FASΔ6 protein. Moreover, this protein competes with the fully functional Fas protein for binding to FasL, thus facilitating the immune escape of tumors.
Biomolecules 15 00789 g001
Table 1. Some of abnormal alternative splicing in various malignant tumors.
Table 1. Some of abnormal alternative splicing in various malignant tumors.
CancerTarget GeneAS IsoformExon/IntronAS TypeFunctionReference
Breast CancerERERα36exon 7, 8Exon skipping, InclusionResistance to endocrine therapies.[90,91,92]
ERα46exon 1, exon 9
ERΔ7exon 7
HER2Δ16HER2exon 20Exon skippingIncreased transforming ability.[69]
CD44CD44v2-v10exon v2-v10Exon InclusionBC growth and metastasis.[72,93]
CD44v3-v10exon v3-v10
CD44v8-v10exon v8-v10
CD44v6exon v6
Colorectal CancerKRASKRAS4Aexon 4AExon InclusionCancer stemness.[94]
KRAS4Bexon 4AExon skippingResponse to endoplasmic reticulum stress.
KLF6KLF6-SV2exon 2Exon skippingCRC cell proliferation and apoptosis.[95]
Lung CancerRBM4RBM4-Sexon3Exon skippingActivation of the SRSF1-mTORC1 pathway promotes NSCLC cell growth.[2]
METMETΔex14exon 14Exon skippingOncogenic activity.[96]
HER2HER2D16exon 16Exon skippingA mediator of osimertinib resistance in patients with metastatic EGFR-mutant NSCLC.[97]
TP53P53βexon 9βExon InclusionCellular senescence.[98,99,100]
p53γexon 9γCell differentiation/Antioxidant response.
p53Ψintron 6Alternative 3′ splicingEpigenetic regulation.
Δ40p53intron 2Intron RetentionApoptosis.
Δ133p53intron 4A novel transcriptional enhancer of T-cell effector function.
Δ160p53intron 4Tumor cell migration and invasion.
Bcl—XBcl-Xsexon 2Alternative 5′ splicingResistance against chemotherapeutic agents.[101]
Hematological MalignanciesBCL—2BCL-2βexon 3Exon skippingAntiapoptotic.[86]
Ovarian CancerBCL—2BCL2L12-Lexon 3Exon InclusionApoptosis.[102]
BCL2L12-Sexon 3Exon skipping
Hepatocellular CarcinomaKLF6KLF6-SV1exon 2Alternative 5′ splicingCancer metastasis, progression.[103]
CDC25ACDC25A ΔE6exon 6Exon skippingHCC growth.[104]
ADRM1ADRM1-ΔEx9exon 9Exon skippingUbiquitin proteasome specificity.[105]
Table 2. Splicing factors that regulate genes related to immune cells.
Table 2. Splicing factors that regulate genes related to immune cells.
Splicing FactorsTypes of Immune CellsAffected GenesReferences
SRSF1, SFPQ, CELF2T cellIrf7, Il27ra, CD45[167,168,169]
PTB, HuR, hnRNPC, RBM5T cellFas, PD-L1, CTLA-4[170,171,172,173,174]
DDX39BT cellFOXP3[175]
hnRNPLB cellMYC, E2F[176]
SF3B1B cellBCL2, MYC[177,178]
TCF3B cellE12, E47[179,180]
KIRNK cell___[181]
PTBP1DCsMHC II[182,183]
Table 3. Some of the innovative technologies for detecting aberrant alternative splicing events, neoantigen immunogenicity, and neoantigen target screening.
Table 3. Some of the innovative technologies for detecting aberrant alternative splicing events, neoantigen immunogenicity, and neoantigen target screening.
Technology/Database Core Functions/
Technical Features
AdvantagesLimitationComplementary TechnologiesClinical Translational ValueReferences
rMATSQuantifies 5 classical splicing events using ΔPSIHigh sensitivity, standardized pipelineRequires replicate samples, cannot resolve complex isoformsWhippetInitial screening of splicing events linked to high-frequency mutations[197]
WhippetLightweight algorithm for high-entropy AS detectionRapid single-sample analysis, covers 40% human genesLow sensitivity for weakly expressed genes, lacks clinical data integrationPacBio SMRTLarge-scale screening of potential therapeutic targets[198]
PsichomicsTCGA integration with Cox survival modelingDirect patient prognosis association, target prioritizationPublic database dependency, low flexibilityAS Cancer AtlasPrognostic biomarker discovery[199]
AS Cancer AtlasPan-cancer AS event database integrating TCGA/GTEx (33 cancer types) with survival-mutation linksInteractive visualization of splicing-clinical correlationsInfrequent updates, sparse data for rare cancersPsichomicsIdentification of pan-cancer splicing targets and therapy-response biomarkers[200]
SNAFDeepImmuno-CNN + BayesTS framework for splicing-derived neoantigensHigh specificity (>85%), high shared antigen ratio (>90%)RNA-seq coverage dependency, lacks MS validationIRISDevelopment of universal TCR therapies[201]
IRISDifferential expression + HLA-I affinity filtering30% reduced false positives, high verifiabilityRequires matched normal samples, HLA typing constraintsMLPersonalized TCR therapy development[202,203]
MLXGBoost + immunogenicity classifier30% AUC improvement, strong generalizabilityData-hungry, computationally intensiveOrbitrap Astral MSEnhanced personalized vaccine design[204]
NGSShort-read detection of high-frequency mutations/Differential genesCost-effective, standardized workflowMisses splicing driversrMATS/WhippetFoundational mutation profiling[205,206,207,208,209,210,211]
PacBio SMRTLong-read (10–15 kb) resolution of complex SVsError-free assembly, full-length isoform detectionHigh cost, low throughputPacBio HiFiGuidance for fusion protein targeting[212]
PacBio HiFiHigh-fidelity long-reads (≥99.9%) for rare isoform validation0.1 attomolar sensitivityLarge storage requirementsOrbitrap Astral MSEnhanced neoantigen authenticity validation[212]
Orbitrap Astral MSDIA-MS detection of frameshift peptidesAntibody-free direct validationDatabase dependency, low-abundance peptide challengesSpectronautConfirmation of neoantigen presentation[213,214,215,216]
SUPPA2AS analysis via transcript abundance (PSI calculation)Single-sample compatibility, no replicates requiredLimited by transcript reconstruction accuracy, low sensitivity for rare isoformsSalmonRapid identification of prognosis-associated splicing events[217]
LeafCutterReference-free splicing analysis using intron excision sitesNovel isoform discovery without exon annotationHigh sequencing depth requirement, limited complex SV resolutionSTARDetection of non-canonical splicing drivers[218]
pVACtoolsNetMHCpan + expression filtering + immunogenicity scoringOpen-source multi-threading supportManual parameter tuning, lacks long-read integrationNeoPredPipePersonalized vaccine candidate prioritization[219]
MHCflurry2.0DL-based HLA-I/II affinity predictionCovers >10,000 HLA alleles, cross-validation supportReduced accuracy for rare HLA typesPrimeRankImproved T-cell response prediction[220]
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

Chen, H.; Tang, J.; Xiang, J. Alternative Splicing in Tumorigenesis and Cancer Therapy. Biomolecules 2025, 15, 789. https://doi.org/10.3390/biom15060789

AMA Style

Chen H, Tang J, Xiang J. Alternative Splicing in Tumorigenesis and Cancer Therapy. Biomolecules. 2025; 15(6):789. https://doi.org/10.3390/biom15060789

Chicago/Turabian Style

Chen, Huiping, Jingqun Tang, and Juanjuan Xiang. 2025. "Alternative Splicing in Tumorigenesis and Cancer Therapy" Biomolecules 15, no. 6: 789. https://doi.org/10.3390/biom15060789

APA Style

Chen, H., Tang, J., & Xiang, J. (2025). Alternative Splicing in Tumorigenesis and Cancer Therapy. Biomolecules, 15(6), 789. https://doi.org/10.3390/biom15060789

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