Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Aug 20.
Published in final edited form as: Ann Oncol. 2022 Mar 23;33(6):578–592. doi: 10.1016/j.annonc.2022.03.011

Alternative RNA Splicing Defects in Pediatric Cancers: New Insights in Tumorigenesis and Potential Therapeutic Vulnerabilities

A S Venkataramany 1,2, K M Schieffer 3, K Lee 3,4,5, C E Cottrell 3,4,5, PY Wang 4,6, ER Mardis 3,4, TP Cripe 4,6,7, D S Chandler 6,8,*
PMCID: PMC12361925  NIHMSID: NIHMS2100498  PMID: 35339647

Abstract

Background:

Compared to adult cancers, pediatric cancers are uniquely characterized by a genomically stable landscape and lower tumor mutational burden. However, alternative splicing, a global cellular process that produces different mRNA/protein isoforms from a single mRNA transcript, has been increasingly implicated in the development of pediatric cancers.

Design:

We review the current literature on the role of alternative splicing in adult cancer, cancer predisposition syndromes, and pediatric cancers. We also describe multiple splice variants identified in adult cancers and confirmed through comprehensive genomic profiling in our institutional cohort of rare, refractory and relapsed pediatric and adolescent young adult cancer patients. Finally, we summarize the contributions of alternative splicing events to neoantigens and chemoresistance and prospects for splicing-based therapies.

Results:

Published dysregulated splicing events can be categorized as exon inclusion, exon exclusion, splicing factor upregulation, or splice site alterations. We observe these phenomena in cancer predisposition syndromes (Lynch syndrome, Li-Fraumeni syndrome, CHEK2) and pediatric leukemia (B-ALL), sarcomas (Ewing sarcoma, rhabdomyosarcoma, osteosarcoma), retinoblastoma, Wilms tumor, and neuroblastoma. Within our institutional cohort, we demonstrate splice variants in key regulatory genes (CHEK2, TP53, PIK3R1, MDM2, KDM6A, NF1) that resulted in exon exclusion or splice site alterations, which were predicted to impact functional protein expression and promote tumorigenesis. Differentially spliced isoforms and splicing proteins also impact neoantigen creation and treatment resistance, such as imatinib or glucocorticoid regimens. Additionally, splice-altering strategies with the potential to change the therapeutic landscape of pediatric cancers include antisense oligonucleotides, adeno-associated virus gene transfers, and small molecule inhibitors.

Conclusions:

Alternative splicing plays a critical role in the formation and growth of pediatric cancers, and our institutional cohort confirms and highlights the broad spectrum of affected genes in a variety of cancers. Further studies that elucidate the mechanisms of disease-inducing splicing events will contribute toward the development of novel therapeutics.

Keywords: Pediatric cancer, alternative splicing, exon inclusion/exclusion, splicing factors, alterations, therapy

Introduction

By 2040, approximately 27.5 million new cancer cases, with nearly 200,000 in children, and 16.3 million cancer deaths are expected annually worldwide1. Cancer is a leading cause of death due to disease in children24, and there remains a need for improved understanding of childhood cancer biology and the development of more effective therapeutics.

Genomic alteration is an initiating event in tumorigenesis. In comparison to adult cancers, pediatric cancers are typically associated with epigenetic dysregulation rather than numerous genetic alterations59. Some pediatric cancers are characterized by copy number variants or alterations in master regulatory factors, such as transcription factors or epigenetic regulators, that affect expression of a broad range of downstream genes6,1013. Between 6 and 18% of pediatric cases arise in the context of a genetic predisposition syndrome14. Recent reports also suggest that alterations in pre-mRNA splicing contribute to tumorigenesis15,16.

Splicing has been repeatedly described as a critical driver in adult cancers. The Cancer Genome Atlas (TCGA) studies on soft tissue sarcomas and bladder urothelial cancer have identified well over 35,000 alternative splicing events in thousands of analyzed genes17,18. The extent to which splicing plays a role in pediatric cancers, however, is largely unknown.

Herein, we review what is currently known about alternative mRNA splicing in pediatric cancers. We categorize various splicing events in tumorigenesis and identify commonalities between cancer types. We also highlight recent splicing variations in a series of patients at our own institution, emphasizing how the study of alternative splicing mechanisms is essential to better understand pediatric oncogenesis. Furthermore, we discuss opportunities for the development of novel therapeutics targeting splicing-driven cancer.

Normal Alternative Splicing

Splicing is an essential cellular mechanism in the nucleus that results in distinct protein isoforms that can be structurally and functionally different from the parent protein. In constitutive splicing, introns are removed, and exons are retained sequentially. During alternative splicing, sequences are selectively excluded or included. Alternative splicing is categorized into distinct events—exon skipping, mutually exclusive exons, alternative 3’ and 5’ splice site selection, and intron retention19. The process of splicing is sequential, requiring the recruitment and assembly of various ribonucleoprotein complexes and splicing factors (Figure 1).

Figure 1: Normal splicing through spliceosome assembly.

Figure 1:

Pre-mRNA splicing is a sequential, two-step transesterification process that requires the recruitment of various ribonucleoprotein complexes (U1, U2, U2AF, U4, U5, U6) and RNA binding proteins (RBPs), such as hnRNPs or SRSFs. Splice sites on the 5’ (donor – GU) and 3’ (acceptor – AG) ends of the introns dictate the exact locations where the spliceosome, the macromolecular machine that mediates this process, cuts the pre-mRNA. Following the binding of the U1 snRNP at the 5’ intronic splice site and the U2 snRNP at the branch site (denoted by adenosine near the 3’ end of the intron), the U4-U6 snRNPs are recruited to form the mature spliceosome. Next, the removal of the 5’ intronic end and subsequent ligation of the 5’-GU site to the branch site creates a lariat. Finally, cleavage of the 3’ intronic splice site removes the intron completely, and the exons are ligated together to form a mature RNA transcript.

Splicing in Adult Cancers: A Broad Overview

Splicing defects are a feature of many adult cancers15,16,20. To provide context for the pediatric landscape, we briefly examine a selection of dysregulatory splicing events in the four most prevalent adult cancers: prostate, breast, lung, and colorectal21 (Table 1).

Table 1:

Key Aberrant Splicing Events in Prevalent Adult Cancers

Adult Cancer Splicing Events
Exon Inclusion
Description Outcome References
Inclusion of exon 4A or 4B of KRAS gene in colorectal cancer Pro-apoptotic (exon 4A) or anti-apoptotic effect (exon 4B) with overall effects on cell migration and tumorigenesis 22,23,25,26,28
Exon Exclusion
Partial or complete skipping of BRCA exon 11 in breast cancer Dysfunction of tumor suppressor and drug resistance to PARP inhibitors and cisplatin 30
Skipping of part of KLF6 exon 2 in breast cancer Localization to cytoplasm instead of nucleus, leading to cancer cell proliferation and metastasis 31,32
Skipping of exons 4–8, containing the ligand-binding domain, in AR in prostate cancer Produce AR-V7 alternative transcript → Constitutive activation of androgen receptor in absence of androgen and cell cycle dysregulation
Produce AR-V9 alternative transcript (different cryptic terminal exons than AR-V7) → Acquired drug resistance against abiraterone
3336
Skipping exon 11 of INSR gene in breast cancer, prostate cancer, non-small cell lung cancer, colorectal cancer Increased cancer cell proliferation
Drug resistance (colorectal cancer)
Increased binding of IGF2, which promotes cancer cell proliferation and drug resistance (breast cancer specifically)
4346
Splicing Factor Upregulation
SRSF1 overexpression in lung cancer (induced by MYC overexpression) More aggressive cancer phenotype, increased oncogenic potential, drug resistance 47,48
SRSF1, SRSF3, hnRNPA1/A2/I overexpression in breast cancer Promote oncogenesis, drug resistance
Differential expression of PKM2 isoform→ tumorigenic, Warburg effect
29,4951
Splice Site Alterations
5’ or 3’ splice site mutations in MET exon 14 Increased stability and oncogenic potential of METΔ14 isoform 52,53

AR: androgen receptor, INSR: insulin receptor

Exon Inclusion Events

KRAS has two isoforms (KRAS4A, KRAS4B) characterized by the inclusion of alternative fourth exons. Perhaps influenced by epigenetic modifications, the ratio of KRAS4A:4B expression is altered in colorectal cancer and affects apoptosis, cell migration, and overall tumorigenesis2228.

Exon Exclusion Events

Aberrant exon exclusion events are numerous in adult cancers, especially in breast cancer29. BRCA1 exon 11 can be partially or completely skipped, resulting in dysfunction of the tumor suppressor and drug resistance to common therapies30. In KLF6, another tumor suppressor, skipping of the nuclear localization signal creates multiple isoforms that aberrently localize to the cytoplasm, inducing proliferation and metastasis31,32. In prostate cancer, the androgen receptor (AR)’s ligand-binding domain is encoded in exons 4–8, and skipping of this region creates isoforms that constitutively activate the AR and dysregulate the cell cycle (AR-V7) or promote abiraterone resistance (AR-V9)3336.

Notably, the insulin receptor (INSR) is alternatively spliced in multiple cancers, including breast, prostate, non-small cell lung, and colorectal cancer3741. INSR has two isoforms—INSR-A (excludes exon 11) and INSR-B (includes exon 11)—that are differentially expressed in a cell- and development-specific manner42. In adults, INSR-B is preferentially expressed in highly metabolic tissues, whereas INSR-A is predominant in the embryo and in sarcomas because of its high-affinity binding of insulin-like growth factor II (IGF-II), which promotes increased cancer cell growth, migration, and viability43,44. INSR exon 11 exclusion facilitates cell proliferation and contributes to drug resistance4446.

Splicing Factor Upregulation

Overexpression of splicing factors can increase splicing events. In lung cancer, upregulated MYC leads to overexpression of serine-rich splicing factor 1 (SRSF1), which causes a more aggressive cancer phenotype47,48. In breast cancer, SRSFs and heterogenous nuclear ribonucleoprotein (hnRNP) proteins, known to promote oncogenesis and drug resistance, are commonly upregulated29,49,50. hnRNPA1/A2/I all contribute to the differential expression of pyruvate kinase (PKM), which has two mutually exclusive alternative exons51. PKM2 expression is favorable as a tumorigenic isoform and is necessary for the Warburg effect51.

Splice Site Alterations

Splice site alterations can introduce a premature termination codon (PTC) that truncates the mRNA transcript (functionally similar to a nonsense or other classical truncating mutation) or promote alternative sequence inclusion/exclusion. In lung cancer, where splice site alterations are common, the proto-oncogene MET can harbor mutations in both 5’ or 3’ splice sites surrounding exon 14 that result in exon exclusion. The alternative MET isoforms lack a key phosphorylation site to bind CBL, a ubiquitin ligase that degrades MET52,53. The METΔ14 splice variant is more stable and potentiates oncogenesis, making it a therapeutic target for tyrosine kinase inhibitors52.

Cancer Predisposition Syndromes

Pediatric cancer patients may have a cancer predisposition syndrome, which originates from germline pathogenic variants associated with splicing defects54,55. Patients with Lynch syndrome have a predisposition to colon and endometrial cancers, some of which can occur during childhood. Many cases are associated with germline alterations of the mismatch repair genes MSH2 and MLH1 that result in low or absent expression of these proteins56. An intronic variant in MSH2 (c.212–553_c.212–479) creates a novel splice donor site that introduces a PTC57. Additionally, splice site alterations in MSH2 (c.211+1G>C) and MLH1 (c.1731+5G>A) can induce nonsense-mediated decay (NMD) of the mRNA transcripts58. Other studies have shown that several MLH1 variants in the exon 10 coding region lead to aberrant splicing59. Additionally, the key genes associated with Lynch syndrome (MLH1, MSH2, MSH6 and PMS2) have been reported to be highly enriched for splicing alterations60.

Li-Fraumeni syndrome is characterized by alterations in the tumor suppressor TP53, increasing the risk for childhood/young adulthood sarcomas, breast cancer, leukemia, etc.6164. Familial studies have identified both intronic and exonic splice site alterations that generate PTCs and aberrant TP53 transcripts65,66. Loss of the initiation codon, synonymous variants, or missense variants in TP53 that localize in exonic splicing enhancers can contribute to the creation of novel splice sites and the expression of different TP53 isoforms67,68.

Additionally, alterations in the Checkpoint Kinase 2 (CHEK2) gene on chromosome 22, encoding the CHK2 protein, are associated with an increased risk of inherited breast cancers and therapy resistance69,70. In an analysis of 13 meningiomas, frameshift alterations resulting in alternative splicing and lack of the CHK2 kinase domain are hypothesized to contribute to the loss of DNA repair mechanisms and increased chromosomal instability71. Within our institutional cohort of rare, refractory, and relapsed pediatric and adolescent young adult (AYA) cancer patients on whom comprehensive genomic profiling was performed (see Supplementary Materials), we identified a novel CHEK2 splice variant in a seven-year-old diagnosed with a subependymal giant cell astrocytoma, World Health Organization (WHO) grade I (Figure 2). Paired somatic disease/germline comparator enhanced exome sequencing revealed a germline splice site variant in CHEK2 (NM_007194.3:c.444+1G>A), also referred to as IVS3+1G>A or IVS2+1G>A72. This variant has been classified as a pathogenic alteration by other clinical laboratories (ClinVar Variation ID:128075) and is associated with cancer predisposition, most frequently breast cancer69. This splice donor variant in intron 3 is a founder variant in the Polish population (general minor allele frequency (MAF): 0.0001, enriched in European population MAF: 0.0005)73. Functional studies have described the use of an alternative 5’ splice site, four base pairs (4bp) within intron 3, predicted to encode a PTC that truncates the transcript and result in reduced CHK2 protein levels72.

Figure 2: CHEK2 splice variant in a seven-year-old diagnosed with a subependymal giant cell astrocytoma.

Figure 2:

A) Schematic of CHEK2 alternative splicing (adapted from Dong et al.72 and Dufault et al.204). The G>A single nucleotide variant in intron 3 results in the use of an alternative splice site, which leads to the insertion of four extra nucleotides between exons 3 and 4 and an out-of-frame transcript. The frameshift creates a premature termination codon (PTC) in exon 4 and reduced CHK2 protein. B) Enhanced exome sequencing was performed on tissues collected from consenting patients enrolled on an institutional translational research protocol (see Supplementary Materials for more information). Aberrant RNA splicing due to germline/somatic variation is shown for each patient (red) compared to a reference cohort of tissue- and disease-matched controls (blue) in a Sashimi plot (methodology described in205). The corresponding genomic sequence is shown (top: reference sequence; bottom: alternative sequence) with the variant highlighted in orange. Coordinates are provided for GRCh38. The patient had a germline splice site variant in CHEK2 (NM_007194.3:c.444+1G>A) confirmed to alter RNA splicing through intron inclusion.

Aberrant Splicing in Pediatric Cancers

While the relationship between alternative splicing and cancer formation is well established in adult cancer literature, the discovery and mechanistic study of disease-causing splice variants in pediatric cancer is a growing field. Most splicing studies have been conducted in the context of leukemias and sarcomas (Table 2).

Table 2:

Summary of Aberrant Splicing Events in Pediatric Cancers

Pediatric Cancer Splicing Events
Exon Inclusion
Description Outcome References
Inclusion of exon 9i in TP53 transcript in pediatric B-ALL Increased expression of p53β isoform that is regulated by SRSF3 74,75
Inclusion of alternative exon 4A in NT5C2 transcript in pediatric B-ALL Dysfunction in metabolism of nucleoside analog drugs → drug resistance 74,7678
Exon Exclusion
Skipping exon 11 of hnRNPA1 gene in pediatric B-ALL NMD activated → changes in cell metabolism, DICER1 deregulation 74
Skipping of exons 4–11 of MDM2 in rhabdomyosarcoma MDM2-ALT1 expression → loss of p53 regulation 7982
Skipping of exons 4–9 of MDM2 in patient with metastatic aggressive alveolar rhabdomyosarcoma (institutional cohort) MDM2-A expression → diminished direct p53 interaction * 201,202
** 203
Skipping exon 11 of INSR gene in rhabdomyosarcoma and osteosarcoma Increased INSR-A: INSR-B ratio → increased cell growth, migration and viability 44,86,88
Skipping exon 6A of DHX9 gene Increased DHX9 and enhanced EWSR1-FLI1 activity 91,95
Skipping exon 8 of EWSR1 gene in certain Ewing sarcoma translocations Produce functional EWSR1-FLI1 protein 92,93
Skipping of exons 6 or 9 in MDM4 in retinoblastoma MDM4-S (exon 6) and MDM4-A (exon 9) expression → oncogenesis 101
Skipping of various exons of RB1 (some induced by mutations) in retinoblastoma Aberrant function of RB1 protein, inheritance of retinoblastoma 103109
Splicing Factor Upregulation
SRSF3 upregulation in osteosarcoma FoxM1, PLK1, CDC25B upregulation and ILF3 alternative splicing → increased proliferation, loss of cell cycle regulation 110112
hnRNPa1 and PTBP1 upregulation caused by N-myc upregulation in neuroblastoma Increased neuroblastoma cell proliferation, differential splicing of PKM gene (isoform 2, inclusion of exon 10) 114
Splice Sites and Splice Site Alterations
Use of alternative splice sites after exons 5 or 9 of WT1 in Wilms tumor Production of four WT1 isoforms (−17AA/−KTS, +17AA/−KTS, −17AA/+KTS and +17AA/+KTS) → undifferentiated tumor cells, upregulation of VEGF and other proliferation genes, altered DNA/RNA binding, RNA splicing/processing, functional abnormalities 119,121128
Altered splice donor site of intron 6 of TP53 in patient with osteosarcoma (institutional cohort) Inclusion of 5 bp of intron 6 and altered splicing * 129,130
Altered splice donor site of intron 11 of PI3KR1 in patient with osteosarcoma (institutional cohort) PI3KR1 exon 11 skipping → constitutive activation of PI3K signaling pathway * 132
** 131
Indel event spanning intron 10 splice acceptor site and adjacent exonic sequence of KDM6A in patient with WHO grade IV medulloblastoma 12 bp deletion (including 10 bp of intron 10 and 2 bp of exon 11) and 2 bp insertion → loss of splice acceptor site and reintroduction of new splice acceptor site → predicted shift of open reading frame, termination codon Present paper
Altered splice donor site of intron 33 of NF1 in patient with WHO grade I ganglioglioma Exon 33 skipping event 140142

ALL: acute lymphoid leukemia, INSR: insulin receptor, NMD: nonsense-mediated decay

*

reference in adult literature that describes the splice variant

**

reference in pediatric literature (cancer or other disease type) that describes the splice variant or a functionally similar variant

Exon Inclusion Events

Dysregulated splicing resulting in exon inclusion has been identified in pediatric B-cell acute lymphoblastic leukemia (B-ALL). B-ALL datasets reveal increased inclusion of TP53 exon 9i, resulting in a TP53 transcript that promotes the p53β isoform regulated by SRSF374,75. Another example involves the cytosolic 5’-nucleosidase II gene NT5C2, which encodes a protein that dephosphorylates purine monophosphates (IMP, GMP). Cancers with NT5C2 transcripts harboring alternative exon 4A transcripts produce an enzyme necessary for inactivating nucleoside-analog drugs used in ALL treatment, leading to drug resistance and cancer relapse74,7678.

Exon Exclusion Events

Most splicing events in pediatric cancers appear to be exon exclusion events in splicing factor or disease-driving genes. In pediatric B-ALL, exon 11 skipping in the splicing factor hnRNPA1 gene activates NMD of the hnRNPA1 transcript74. The decay and decreased expression of hnRNPA1 then affects cell metabolism/DICER1 regulation74.

In sarcomas, exon exclusion within critical genes promotes oncogene function and increased cell growth. Rhabdomyosarcoma is the most common soft tissue tumor in children. Skipping of exons 4–11 in the mouse double minute 2 homolog (MDM2) proto-oncogene, an E3 ubiquitin ligase that targets p53 toward degradation, produces the alternative transcript MDM2-ALT179. This isoform is constitutively expressed in alveolar and embryonal rhabdomyosarcoma tumors and is unable to fulfill the full-length transcript’s role of negatively regulating p5380. In vitro and in vivo studies have shown that MDM2-ALT1 expression leads to accelerated tumorigenesis and increased incidence of rhabdomyosarcoma80,81. These splicing responses are mediated in part through the splicing factor SRSF1, which binds an exonic splicing silencer element in MDM2 exon 11 and acts as a negative regulator to promote MDM2-ALT1 expression82. In our institutional cohort, we identified a one-year-old patient diagnosed with metastatic aggressive alveolar rhabdomyosarcoma harboring a PAX3-FOXO1 fusion and a hotspot missense variant in the TP53 gene (NM_000546.5:c.742C>T (p.Arg248Trp)). Additionally, in the same patient, we identified alternative splicing of MDM2 (NM_002392.5), specifically deletion of exons 4–9 (known as MDM2-A)83 (Figure 3A). MDM2 and TP53 interact though an autoregulatory negative feedback loop to inhibit cell growth84. The MDM2-A transcript lacks its TP53 binding domain, resulting in diminished direct p53 protein interaction85. Multiple dysregulation events within the MDM2-TP53 pathway are hypothesized to contribute to this tumor’s aggressive nature.

Figure 3: Sashimi plots of splicing events in pediatric cancers.

Figure 3:

A) Schematic of MDM2 alternative splicing seen in a one-year old rhabdomyosarcoma patient harboring a hotspot missense TP53 variant (NM_000546.5:c.742C>T). MDM2 exons 4–9 are skipped to generate the MDM2-A variant. B) A 16-year old osteosarcoma patient had a somatic splice site variant in TP53 (NM_000546.5:c.672+1G>T) that was confirmed to alter RNA splicing through intron inclusion in the primary tumor. This patient also had a metastatic lung nodule with an additional somatic splice site variant in PIK3R1 (NM_181523.3: c.1425+1G>C) confirmed to result in exon 11 skipping. (C) A 27-year old medulloblastoma patient had a somatic splice site variant in KDM6A (NM_001291415.2:c.876–10_877delinsAG), which generated a novel splice site 2bp within exon 11. D) A 16-year old ganglioglioma patient presented with a somatic variant in NF1 (NM_000267.3:c.4514+1G>T), which was confirmed to result in exon 33 skipping.

In rhabdomyosarcoma and osteosarcomas, multiple studies have noted the increased expression of INSR44,86,87. An analysis of 19 osteosarcoma patient samples showed that INSR-A transcripts, which are formed through exon 11 exclusion and demonstrate higher-affinity binding to IGF-II, were consistently expressed at higher levels than INSR-B transcripts in all samples86. Similarly, a recent study of 30 embryonal and alveolar rhabdomyosarcoma patient samples found that all tumors expressed significant levels of INSR-A compared to normal muscle control samples88.

Interestingly, exon exclusion events in Ewing sarcoma affect both the hallmark oncoprotein and genes enhancing its function. The EWSR1-FLI1 translocation (t(11;22)(q24;q12)) encodes a namesake aberrant transcription factor that has a widespread impact on DNA regulation, transcription, and post-transcriptional processes, including splicing89,90. The EWSR1-FLI1 chimeric interacts with the spliceosome by inducing alternative splicing of oncogenic regulatory genes or associating with splicing factors, a phenomenon observed in Ewing sarcoma cell lines and patient tumors before chemotherapy91. The most common EWSR-FLI1 translocation is composed of EWSR1 exons 1–7 and FLI1 exons 6–9. One fusion variant includes EWSR1 exon 8, which must be spliced out by hnRNPH1 to generate an in-frame EWSR1-FLI1 transcript and promote Ewing sarcoma pathogenesis9294. In another example, expression of DHX9, which participates in RNA metabolism and maintenance of genomic stability, enhances EWSR1-FLI1 activity and correlates with worse patient prognosis91,95. DHX9 exon 6A contains a PTC, and splicing factors hnRNPM and SRSF3, among others, have been shown to splice this exon out, ensuring high expression of the DHX9 transcript91.

Retinoblastoma, an ocular cancer, presents with all categories of aberrant splicing9699. Out of 1453 differentially spliced genes in retinoblastoma tumor samples, exon exclusion and mutually exclusive exons were the most predominant events100. Furthermore, these differential splicing events enriched the activity of E2F family transcription factors, the visual sense gene ABCA4, and the splicing factor DAZAP1100. One other well known event in retinoblastoma occurs in MDM4, where exclusion of exons 6 or 9 produces the oncogenic isoforms MDM4-S or MDM4-A respectively101. Additionally, the Dab1 and RB1 genes undergo both exon inclusion and exclusion events, and various exon skipping events, some resulting from alterations, in RB1 have also been reported through genetic analyses of families with retinoblastoma102109.

Splicing Factor Upregulation

In osteosarcoma, SRSF3 upregulation contributes to the subsequent upregulation of cell cycle regulators FoxM1, PLK1, and CDC25B, all of which increase cell proliferation and transformation110. Furthermore, SRSF3 promotes alternative splicing of interleukin enhancer-binding factor 3 (ILF3) isoforms 1 and 2 and loss of cell cycle regulation110112.

In neuroblastoma, the most common extracranial solid tumor in children, the splicing factors hnRNPA1 and PTBP1 are upregulated due to N-Myc overexpression113,114. Both splicing factors increase neuroblastoma cell proliferation and the differential splicing of PKM2 through the inclusion of exon 10114. Like INSR, the PKM2 splice variant is primarily expressed during fetal development and in several cancers due to its favorable effect on cell metabolism115.

Splice Sites and Splice Site Alterations

Wilms Tumor, a pediatric nephroblastoma characterized by the WT1 gene, has four major isoforms generated through the use of alternate splice sites after exons 5 or 9, though other variants have been observed116120. The resulting functional isoforms and their ratios are involved in undifferentiated tumor cells, upregulation of proliferation genes, DNA/RNA binding, and post-transcriptional regulation, and functional abnormalities (seen in Frasier syndrome)119,121128.

Within our institutional pediatric and AYA cancer cohort, we found evidence of the contribution of splice site alterations. In a 16-year-old female diagnosed with osteosarcoma, we identified a variant in TP53 affecting the splice donor site of intron 6 (NM_000546.5:c.672+1G>T) (Figure 3B). This variant is predicted to result in the inclusion of 5bp of intron 6 as previously described (MutSpliceDB, https://brb.nci.nih.gov/splicing/) and has been documented in multiple cancer types in the Catalogue of Somatic Mutations in Cancer (COSMIC) and the International Agency for Research on Cancer (IARC) TP53 databases129,130. The metastatic lung nodule harbored the same TP53 variant as the primary tumor, as well as a newly acquired PIK3R1 variant affecting the splice donor site of intron 11 (NM_181523.3:c.1425+1G>C) (Figure 3B). This PIK3R1 variant results in exon 11 skipping and is predicted to constitutively activate the PI3K signaling pathway131,132. Activating alterations within the PI3K/AKT/mTOR signaling pathway are a newly described mechanism of cell growth and proliferation in osteosarcoma133.

Variants within genes involved in epigenetic regulation are frequent within medulloblastoma134. KDM6A, a histone demethylase, undergoes recurrent frameshift, splice-site, and predicted truncating variants that are well documented in cancer, including medulloblastoma135138. In our cohort, a 27-year-old WHO grade IV medulloblastoma patient was found to harbor a novel KDM6A complex insertion-and-deletion (indel) event spanning the intron 10 splice acceptor site and adjacent exonic sequence (NM_001291415.2:c.876–10_877delinsAG) (Figure 3C). This complex alteration includes a 12bp deletion and a 2bp insertion. The deletion encompasses 10bp of intron 10, including the splice acceptor site (AG), and 2bp of exon 11 (CCCTTCTCAG|GT). A 2bp insertion (AG) then reintroduces a splice acceptor site. Using ExPASy Translate, we found this change is predicted to result in a shift of the reading frame, leading to introduction of a PTC (p.(Cys293LeufsTer19)) and truncated transcript139.

We also identified a splice variant in a 16-year-old patient diagnosed with a ganglioglioma, WHO grade I. The tumor was found to harbor an NF1 splice site variant affecting the donor site of intron 33 (NM_000267.3:c.4514+1G>T) (Figure 3D). Disruption of this splice site has been previously reported in clinical databases and literature as a germline finding in patients with neurofibromatosis type 1 (NF1) (ClinVar Variation ID 576239) and as a somatic finding in NF-1 associated tumors140143. Similar to predictions in the literature, this splice site variant results in an exon 33 skipping event140,142.

Alternative Splicing and Neoantigens

The multitude of transcripts generated from alternative splicing can create novel epitopes and antigens that diversify a cancer’s transcriptional profile144. Widespread analysis of the TCGA has demonstrated the creation of novel exon-exon junction sites (coined “neojunctions”) that are independent of tumor mutational burden145. These sites, and others generated through noncoding regions, intron retention, indels, or splicing errors, can generate polypeptides that bind the MHC complex and become cancer-specific neoantigens, which contribute to a cancer’s immunogenicity and response to immunotherapy145150. For example, non-small cell lung cancer patient samples had a higher expression of neoantigens derived from aberrantly spliced gene isoforms151.

Within pediatric cancers, the contribution of neoantigens arising from alternative splicing is an emerging field; however, studies in hematologic malignancies and medulloblastoma have demonstrated integral roles of neoantigens in cancer progression. An analysis of RNA-seq data from 400 pediatric B-ALL samples showed an increased prevalence of the CD22 exon 5–6 skipping event, which was further validated in 18 primary B-ALL samples152. Furthermore, this CD22 isoform, and not the full-length isoform, was targetable with a novel, highly specific monoclonal antibody in BALB/c mice152. Neoantigens in medulloblastoma, a cancer characterized by low mutational load, were created primarily through aberrant splice junctions that joined two non-exonic sequences153,154. Pharmacologic modulation of splicing also affects neoepitope production. The in vitro and in vivo use of splicing modulators like indisulam on various cancer types has resulted in splicing-derived neoantigens that favorably alter tumor immunogenicity155. Such an approach suggests that alternative splicing in pediatric cancers could be clinically targeted to enhance current immune therapies.

Alternative Splicing and Chemoresistance

One of the first clinically used small molecule inhibitors, imatinib effectively abrogates signaling of the BCR-ABL chimeric fusion protein in chronic myelogenous leukemia (CML)156,157. BCR-ABL is a constitutively activated tyrosine kinase resulting from a t(9;22) gene fusion between the ABL1 and BCR genes on chromosomes 9 and 22 respectively. By binding close to the ATP binding site of BCR-ABL, imatinib sterically competes with ATP and hinders the downstream signaling cascade that would normally promote oncogenesis. However, imatinib is less effective when BCR-ABL acquires alterations in the kinase domain or is alternatively spliced to include intronic nucleotides between exons 8 and 9158160. These changes to BCR-ABL structurally alter the kinase domain or induce conformational shifts in the BCR-ABL protein, effectively decreasing the response to imatinib158162. A second potential mechanism of imatinib resistance in CML and other tyrosine kinase-driven cancers is the expression of a BCL2-like 11 (BIM) alternatively spliced isoform, which leads to loss of the apoptosis-inducing BH3 domain on BCL2161,163,164. Notably, this splicing event can be circumvented through antisense oligonucleotide (ASO) therapy, and tyrosine kinase inhibitor response can be successfully restored164.

Another common example of chemoresistance resulting from alternative splicing can be found in BRCA1 isoforms161. Mice implanted with BRCA1 exon 11 mutant tumors expressed high levels of BRCA1-Δ11q, which correlated with treatment resistance to PARP1 inhibitors and cisplatin30. Interestingly, BRCA1-Δ11q levels can be decreased via the spliceosome inhibitor PB-1 or ASOs to resensitize cells to PARP1 inhibitors30,165.

Likewise, there are examples of alternative splicing leading to therapy resistance in pediatric cancers. Chimeric antigen receptor T-cell (CAR-T) immunotherapy involves the infusion of autologous T cells that targeted against CD19, commonly expressed by neoplastic and non-neoplastic B cells166. A subset of pediatric B-ALL patients who relapse following CAR-T therapy have downregulated CD-19 expression on their cancer cells, which sometimes results from CD19 pre-mRNA alternative splicing167,168. When CD19 exon 2 is skipped, the final protein is truncated at the N-terminus and loses its CAR recognition site, rendering the CAR-T therapy ineffective168. Another example of splicing-mediated chemoresistance is seen in glucocorticoid treatment resistance in pediatric ALL. An RNA sequencing study of 38 ALL samples identified several splicing pathways that were altered in glucocorticoid-resistant ALL cells, including variants in hnRNP and SRSF protein families that were specific to ALL subtypes169. However, further investigation in a lineage-specific manner would be necessary to elucidate the exact splicing changes and their roles in steroid-resistant ALL cells169.

Therapies for Splicing-Induced Diseases

Antisense Oligonucleotides and Alternative Vehicles

Therapeutic development for pediatric diseases in recent years has been pioneered by the success of ASO therapies that modulate mRNA splicing, via exon inclusion or exclusion, in spinal muscular atrophy (SMA) and Duchenne muscular dystrophy (DMD). For SMA, nusinersen is an 18-mer ASO that blocks hnRNPA1 intronic binding sites to promote SMN2 exon 7 inclusion and full-length SMN2 mRNA levels170174 (Figure 4AC). In DMD, the morpholino treatment eteplirsen induces exon 51 skipping in dystrophin pre-mRNA and restores the open reading frame to produce functional dystrophin175. New therapies have since been developed to restore dystrophin through exon 45 and exon 53 skipping176. Both treatments have improved motor function and clinical outcomes in patients, suggesting that RNA-based therapeutics can be viable approaches for splicing-induced diseases like cancer16,177,178.

Figure 4: Mechanisms of ASO therapy to modulate alternative splicing in SMA and cancer.

Figure 4:

Humans have two copies of the SMN gene, which are nearly identical except for a C vs. T nucleotide difference in exon 7171. SMA is caused by low levels of SMN protein due to A) loss of function of SMN1 and B) exon 7 skipping in SMN2. C) ASOs targeting the intronic splicing silencer (ISS) located in intron 7–8 can prevent hnRNPA1 binding, which promotes exon 7 inclusion and functional SMN2 transcripts/SMN protein production172174. D) Full-length MDM2 expression is facilitated through SRSF2 binding at exon 1182. E) In the setting of wild-type p53 in cancer, ASOs can block the SRSF2 binding site and promote skipping of MDM2 exons 4–11, resulting in MDM2-ALT1 and loss of MDM2-induced p53 degradation82.

In in vitro rhabdomyosarcoma models, ASOs can block splicing factor binding sites to switch the splicing of cancer-driving genes, such as INSR. The splicing factors SRSF3 and CUGBP-1 mediate INSR alternative splicing in rhabdomyosarcoma, and blocking these binding sites with splice switching oligonucleotides in rhabdomyosarcoma cell lines reduces the levels of INSR-A, resulting in decreased angiogenesis and proliferation179. In the case of MDM2, assessing p53 status of the tumor is essential to determining therapeutic ASO design. For example, in the setting of wild-type p53, ASOs can target the SRSF2 binding site on MDM2 exon 11 to induce skipping of exons 4–1182 (Figure 4DE). This exon skipping results in the production of the MDM2-ALT1 isoform, which can no longer degrade wild-type p53 in cancer cells.

By binding to the mRNA and affecting mRNA/protein levels, ASOs are an attractive alternative used to target large proteins. In diffuse large B-cell lymphoma, STAT3 is one such protein against which ASOs have shown therapeutic efficacy. In a phase 1B trial, AZD9150, a 16-mer ASO that downregulates STAT3, showed 13% clinical benefit180. Additionally, several pilot clinical trials have been launched to explore ASO use in solid tumors, leukemias, and lymphomas, though many of these are not in pediatric cancers181.

The barriers of ASO therapy in vivo lie in targeted delivery to tumors, lack of efficacy due to degradation, and toxicity182184. Systemic intravenous administration, for example, deposits the ASO compound in both target and non-target tissues, such as the liver and kidney. The ASO’s decreased bioavailability might not simply be overcome with increased dosing due to accumulation, immune, or aptameric binding toxicities. Some off-target effects, instability, and dosing issues are avoided through different chemical formulations, which may include various backbones, ribose modification, or polymer/peptide conjugations. Nanoparticles and other technologies are also a growing field to mitigate obstacles in ASO delivery185187. However, each chemical class of ASOs or delivery systems has its own associated toxicities that must be considered during therapeutic design.

Additionally, vehicles such as non-pathogenic adeno-associated viruses (AAVs) offer a solution for issues with drug delivery. AAVs are single-stranded DNA viruses that contain rep and cap genes between inverted terminal repeats (ITRs) on the 5’ and 3’ ends of the viral genome. Recombinant vectors can be engineered by replacing the rep and cap genes with a promoter followed by transgenes and a poly-A tail188. Multiple studies have explored the insertion of ASO sequences instead of a transgene in the recombinant AAV vector189,190. In DMD, sustained exon 51 skipping in the dystrophin gene was achieved by encoding antisense sequences following a U7 promoter in an AAV vector, and this approach was extremely effective in inducing long-term exon 51 skipping and therapeutic dystrophin levels in the mdx52 mouse model189,191. Therefore, similar principles might be applicable to pediatric cancers with splicing defects, where the antisense sequence necessary to therapeutically alter splicing is expressed via a U7 promoter in an AAV vector.

Small Molecule Inhibitors

The dynamic assembly of the spliceosome can be difficult to target, but small molecule inhibitors (SMI) can inhibit the individual proteins within the complex, making them effective choices for diseases driven by splicing factors15,192,193. Multiple SMIs, including spliceostatin A or pladienolides, directly target SF3B1, a commonly altered component of the spliceosome identified in uveal melanoma, AML, and myelodysplastic syndromes194198. Upon treatment with SF3B1 inhibitors, in vitro and in vivo assays show arrested cell growth and increased tumor cell death199. Though early attempts at clinical trials were unsuccessful due to toxicity, SMIs are still being explored as promising agents against myeloid tumors with splicing factor mutations and defects (www.clinicaltrials.gov, NCT02841540)193. To mitigate the unfavorable side effects often seen with SMIs, examining and repurposing currently approved FDA drugs with therapeutic effects on splicing could be an alternative approach200.

Conclusions

A growing body of work in the splicing field has shown that events such as exon inclusion/exclusion, splicing factor upregulation, and splice site alterations are prevalent in pediatric cancers and contribute to oncogenesis, neoantigens, and chemoresistance. Through a better understanding of how splicing and other post-transcriptional mechanisms act in tumorigenesis, we can develop more effective treatments outside of the standard regimens for pediatric patients.

Supplementary Material

Supplementary Materials

Highlights.

  • Alternative splicing has been increasingly implicated in pediatric cancer, neoantigens, and chemoresistance.

  • Exon inclusion/exclusion, splicing factor upregulation, or splice site alterations encompass most of these splicing events.

  • Splice variants previously observed in several cancers have also been observed in our institutional cohort of pediatric patients.

  • Current therapies against splicing events use ASO, AAV, and small molecule inhibitors.

  • Better understanding of splicing biology will contribute toward the development of novel therapeutics for pediatric cancer.

Funding and Acknowledgements

This work was supported by the Nationwide Children’s Hospital Technology Development Fund, The Ohio Development Services Agency and Third Frontier Technology Validation and Start-Up Fund, Alex’s Lemonade Stand Foundation, Cancer Free Kids Pediatric Cancer Research Alliance, the Ohio State University Comprehensive Cancer Center, and the National Institutes of Health under grant number P30 CA016058, R21CA227926, R03CA259865, and R01CA262873. We thank the patients and their families for participating in our translational research protocol. We thank the Nationwide Foundation Pediatric Innovation Fund for generously supporting sequencing, data production, and analysis.

Figures 1, 2A, and 4 were created with BioRender.com.

Footnotes

Disclosures

Elaine Mardis serves on the scientific advisory boards of Scorpion Therapeutics, LLC and of PACT Pharma LLC, for which she receives honoraria and restricted stock units. All remaining authors have declared no conflicts of interest.

References

  • 1.Society AC. Global Cancer Facts & Figures 4th Edition. 2020. Atlanta: American Cancer Society; 2018. [Google Scholar]
  • 2.Cunningham RM, Walton MA, Carter PM. The major causes of death in children and adolescents in the United States. New England Journal of Medicine 2018; 379 (25): 2468–2475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Pritchard-Jones K, Pieters R, Reaman GH et al. Sustaining innovation and improvement in the treatment of childhood cancer: lessons from high-income countries. The lancet oncology 2013; 14 (3): e95–e103. [DOI] [PubMed] [Google Scholar]
  • 4.Lam CG, Howard SC, Bouffet E et al. Science and health for all children with cancer. Science 2019; 363 (6432): 1182–1186. [DOI] [PubMed] [Google Scholar]
  • 5.Filbin M, Monje M. Developmental origins and emerging therapeutic opportunities for childhood cancer. Nat Med 2019; 25 (3): 367–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Huether R, Dong L, Chen X et al. The landscape of somatic mutations in epigenetic regulators across 1,000 paediatric cancer genomes. Nature communications 2014; 5 (1): 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kattner P, Strobel H, Khoshnevis N et al. Compare and contrast: pediatric cancer versus adult malignancies. Cancer and Metastasis Reviews 2019; 38 (4): 673–682. [DOI] [PubMed] [Google Scholar]
  • 8.Ma X, Liu Y, Liu Y et al. Pan-cancer genome and transcriptome analyses of 1,699 paediatric leukaemias and solid tumours. Nature 2018; 555 (7696): 371–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gröbner SN, Worst BC, Weischenfeldt J et al. The landscape of genomic alterations across childhood cancers. Nature 2018; 555 (7696): 321–327. [DOI] [PubMed] [Google Scholar]
  • 10.Bax DA, Mackay A, Little SE et al. A Distinct Spectrum of Copy Number Aberrations in Pediatric High-Grade Gliomas. Clinical Cancer Research 2010; 16 (13): 3368–3377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Diskin SJ, Hou C, Glessner JT et al. Copy number variation at 1q21. 1 associated with neuroblastoma. Nature 2009; 459 (7249): 987–991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gambale A, Russo R, Andolfo I et al. Germline mutations and new copy number variants among 40 pediatric cancer patients suspected for genetic predisposition. Clinical genetics 2019; 96 (4): 359–365. [DOI] [PubMed] [Google Scholar]
  • 13.Downing JR, Wilson RK, Zhang J et al. The pediatric cancer genome project. Nat Genet 2012; 44 (6): 619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Capasso M, Montella A, Tirelli M et al. Genetic Predisposition to Solid Pediatric Cancers. Frontiers in Oncology 2020; 10 (2083). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zhang Y, Qian J, Gu C et al. Alternative splicing and cancer: a systematic review. Signal transduction and targeted therapy 2021; 6 (1): 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Cherry S, Lynch KW. Alternative splicing and cancer: insights, opportunities, and challenges from an expanding view of the transcriptome. Genes & Development 2020; 34 (15–16): 1005–1016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yang X, Huang W-t, He R-q et al. Determining the prognostic significance of alternative splicing events in soft tissue sarcoma using data from The Cancer Genome Atlas. Journal of Translational Medicine 2019; 17 (1): 283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.He R-q, Zhou X-g, Yi Q-y et al. Prognostic signature of alternative splicing events in bladder urothelial carcinoma based on spliceseq data from 317 cases. Cellular Physiology and Biochemistry 2018; 48 (3): 1355–1368. [DOI] [PubMed] [Google Scholar]
  • 19.Montes M, Sanford BL, Comiskey DF et al. RNA Splicing and Disease: Animal Models to Therapies. Trends Genet 2019; 35 (1): 68–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Urbanski LM, Leclair N, Anczuków O. Alternative-splicing defects in cancer: Splicing regulators and their downstream targets, guiding the way to novel cancer therapeutics. Wiley Interdisciplinary Reviews: RNA 2018; 9 (4): e1476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Cancer Facts & Figures 2022. American Cancer Society; 2022. [Google Scholar]
  • 22.Abubaker J, Bavi P, Al-Haqawi W et al. Prognostic significance of alterations in KRAS isoforms KRAS-4A/4B and KRAS mutations in colorectal carcinoma. The Journal of Pathology: A Journal of the Pathological Society of Great Britain and Ireland 2009; 219 (4): 435–445. [DOI] [PubMed] [Google Scholar]
  • 23.Eilertsen IA, Sveen A, Strømme JM et al. Alternative splicing expands the prognostic impact of KRAS in microsatellite stable primary colorectal cancer. International journal of cancer 2019; 144 (4): 841–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Riffo-Campos ÁL, Gimeno-Valiente F, Rodríguez FM et al. Role of epigenetic factors in the selection of the alternative splicing isoforms of human KRAS in colorectal cancer cell lines. Oncotarget 2018; 9 (29): 20578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zhu G, Pei L, Xia H et al. Role of oncogenic KRAS in the prognosis, diagnosis and treatment of colorectal cancer. Molecular cancer 2021; 20 (1): 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Plowman SJ, Arends MJ, Brownstein DG et al. The K-Ras 4A isoform promotes apoptosis but does not affect either lifespan or spontaneous tumor incidence in aging mice. Experimental cell research 2006; 312 (1): 16–26. [DOI] [PubMed] [Google Scholar]
  • 27.Voice JK, Klemke RL, Le A et al. Four human ras homologs differ in their abilities to activate Raf-1, induce transformation, and stimulate cell motility. Journal of Biological Chemistry 1999; 274 (24): 17164–17170. [DOI] [PubMed] [Google Scholar]
  • 28.Plowman S, Berry R, Bader S et al. K-ras 4A and 4B are co-expressed widely in human tissues, and their ratio is altered in sporadic colorectal cancer. Journal of experimental & clinical cancer research 2006; 25 (2): 259–267. [PubMed] [Google Scholar]
  • 29.Yang Q, Zhao J, Zhang W et al. Aberrant alternative splicing in breast cancer. Journal of Molecular Cell Biology 2019; 11 (10): 920–929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wang Y, Bernhardy AJ, Cruz C et al. The BRCA1-Δ11q alternative splice isoform bypasses germline mutations and promotes therapeutic resistance to PARP inhibition and cisplatin. Cancer research 2016; 76 (9): 2778–2790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.DiFeo A, Martignetti JA, Narla G. The role of KLF6 and its splice variants in cancer therapy. Drug Resistance Updates 2009; 12 (1–2): 1–7. [DOI] [PubMed] [Google Scholar]
  • 32.Hatami R, Sieuwerts AM, Izadmehr S et al. KLF6-SV1 drives breast cancer metastasis and is associated with poor survival. Science translational medicine 2013; 5 (169): 169ra112–169ra112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Paschalis A, Sharp A, Welti JC et al. Alternative splicing in prostate cancer. Nature Reviews Clinical Oncology 2018; 15 (11): 663–675. [DOI] [PubMed] [Google Scholar]
  • 34.Hu R, Dunn TA, Wei S et al. Ligand-independent androgen receptor variants derived from splicing of cryptic exons signify hormone-refractory prostate cancer. Cancer research 2009; 69 (1): 16–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kohli M, Ho Y, Hillman DW et al. Androgen receptor variant AR-V9 is coexpressed with AR-V7 in prostate cancer metastases and predicts abiraterone resistance. Clinical Cancer Research 2017; 23 (16): 4704–4715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.McCrea E, Sissung TM, Price DK et al. Androgen receptor variation affects prostate cancer progression and drug resistance. Pharmacological research 2016; 114: 152–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Aljada A, Saleh AM, Al-Aqeel SM et al. Quantification of insulin receptor mRNA splice variants as a diagnostic tumor marker in breast cancer. Cancer Biomarkers 2015; 15 (5): 653–661. [DOI] [PubMed] [Google Scholar]
  • 38.Heni M, Hennenlotter J, Scharpf M et al. Insulin receptor isoforms A and B as well as insulin receptor substrates-1 and-2 are differentially expressed in prostate cancer. PLoS One 2012; 7 (12): e50953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Huang J, Morehouse C, Streicher K et al. Altered expression of insulin receptor isoforms in breast cancer. PLoS One 2011; 6 (10): e26177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Jiang L, Zhu W, Streicher K et al. Increased IR-A/IR-B ratio in non-small cell lung cancers associates with lower epithelial-mesenchymal transition signature and longer survival in squamous cell lung carcinoma. BMC cancer 2014; 14 (1): 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Santoro MA, Andres SF, Galanko JA et al. Reduced insulin-like growth factor I receptor and altered insulin receptor isoform mRNAs in normal mucosa predict colorectal adenoma risk. Cancer Epidemiology and Prevention Biomarkers 2014; 23 (10): 2093–2100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Malakar P, Chartarifsky L, Hija A et al. Insulin receptor alternative splicing is regulated by insulin signaling and modulates beta cell survival. Scientific reports 2016; 6 (1): 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Belfiore A, Malaguarnera R. The insulin receptor: a new target for cancer therapy. Frontiers in endocrinology 2011; 2: 93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Vella V, Milluzzo A, Scalisi NM et al. Insulin Receptor Isoforms in Cancer. Int J Mol Sci 2018; 19 (11): 3615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Forest A, Amatulli M, Ludwig DL et al. Intrinsic resistance to cixutumumab is conferred by distinct isoforms of the insulin receptor. Molecular Cancer Research 2015; 13 (12): 1615–1626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Belfiore A, Malaguarnera R, Vella V et al. Insulin receptor isoforms in physiology and disease: an updated view. Endocrine reviews 2017; 38 (5): 379–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Das S, Krainer AR. Emerging functions of SRSF1, splicing factor and oncoprotein, in RNA metabolism and cancer. Molecular Cancer Research 2014; 12 (9): 1195–1204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Gout S, Brambilla E, Boudria A et al. Abnormal expression of the pre-mRNA splicing regulators SRSF1, SRSF2, SRPK1 and SRPK2 in non small cell lung carcinoma. 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Karni R, de Stanchina E, Lowe SW et al. The gene encoding the splicing factor SF2/ASF is a proto-oncogene. Nature structural & molecular biology 2007; 14 (3): 185–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Yen TY, Wong R, Pizzo D et al. Over-Expression of RNA Processing, Heat Shock, and DNA Repair Proteins in Breast Tumor Compared to Normal Tissue. Proteomics 2020; 20 (15–16): 2000044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.David CJ, Chen M, Assanah M et al. HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature 2010; 463 (7279): 364–368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Frampton GM, Ali SM, Rosenzweig M et al. Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer discovery 2015; 5 (8): 850–859. [DOI] [PubMed] [Google Scholar]
  • 53.Coomer AO, Black F, Greystoke A et al. Alternative splicing in lung cancer. Biochimica et Biophysica Acta (BBA)-Gene Regulatory Mechanisms 2019; 1862 (11–12): 194388. [DOI] [PubMed] [Google Scholar]
  • 54.Garber JE, Offit K. Hereditary cancer predisposition syndromes. Journal of clinical oncology 2005; 23 (2): 276–292. [DOI] [PubMed] [Google Scholar]
  • 55.Ripperger T, Bielack SS, Borkhardt A et al. Childhood cancer predisposition syndromes—a concise review and recommendations by the Cancer Predisposition Working Group of the Society for Pediatric Oncology and Hematology. American journal of medical genetics Part A 2017; 173 (4): 1017–1037. [DOI] [PubMed] [Google Scholar]
  • 56.Peltomäki P Lynch syndrome genes. Familial cancer 2005; 4 (3): 227–232. [DOI] [PubMed] [Google Scholar]
  • 57.Clendenning M, Buchanan DD, Walsh MD et al. Mutation deep within an intron of MSH2 causes Lynch syndrome. Familial cancer 2011; 10 (2): 297–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Naruse H, Ikawa N, Yamaguchi K et al. Determination of splice-site mutations in Lynch syndrome (hereditary non-polyposis colorectal cancer) patients using functional splicing assay. Familial cancer 2009; 8 (4): 509–517. [DOI] [PubMed] [Google Scholar]
  • 59.Soukarieh O, Gaildrat P, Hamieh M et al. Exonic splicing mutations are more prevalent than currently estimated and can be predicted by using in silico tools. PLoS genetics 2016; 12 (1): e1005756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Rhine CL, Cygan KJ, Soemedi R et al. Hereditary cancer genes are highly susceptible to splicing mutations. PLoS genetics 2018; 14 (3): e1007231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Li FP, Fraumeni JF, Mulvihill JJ et al. A cancer family syndrome in twenty-four kindreds. Cancer research 1988; 48 (18): 5358–5362. [PubMed] [Google Scholar]
  • 62.Li FP, Fraumeni JF Jr. Rhabdomyosarcoma in children: epidemiologic study and identification of a familial caneer syndrome. Journal of the National Cancer Institute 1969; 43 (6): 1365–1373. [PubMed] [Google Scholar]
  • 63.Li FP, FRAUMENI JF JR. Soft-tissue sarcomas, breast cancer, and other neoplasms: a familial syndrome? Annals of internal medicine 1969; 71 (4): 747–752. [DOI] [PubMed] [Google Scholar]
  • 64.Varley J, Evans D, Birch JM. Li-Fraumeni syndrome--a molecular and clinical review. British Journal of Cancer 1997; 76 (1): 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Verselis SJ, Rheinwald JG, Fraumeni JF et al. Novel p53 splice site mutations in three families with Li-Fraumeni syndrome. Oncogene 2000; 19 (37): 4230–4235. [DOI] [PubMed] [Google Scholar]
  • 66.Frebourg T, Barbier N, Yan Y et al. Germ-line p53 mutations in 15 families with Li-Fraumeni syndrome. American journal of human genetics 1995; 56 (3): 608. [PMC free article] [PubMed] [Google Scholar]
  • 67.Kouidou S, Malousi A, Maglaveras N. Li–Fraumeni and Li–Fraumeni-like syndrome mutations in p53 are associated with exonic methylation and splicing regulatory elements. Molecular Carcinogenesis: Published in cooperation with the University of Texas MD Anderson Cancer Center 2009; 48 (10): 895–902. [DOI] [PubMed] [Google Scholar]
  • 68.Varley J, Chapman P, McGown G et al. Genetic and functional studies of a germline TP53 splicing mutation in a Li–Fraumeni-like family. Oncogene 1998; 16 (25): 3291–3298. [DOI] [PubMed] [Google Scholar]
  • 69.Nevanlinna H, Bartek J. The CHEK2 gene and inherited breast cancer susceptibility. Oncogene 2006; 25 (43): 5912–5919. [DOI] [PubMed] [Google Scholar]
  • 70.Staalesen V, Falck J, Geisler S et al. Alternative splicing and mutation status of CHEK2 in stage III breast cancer. Oncogene 2004; 23 (52): 8535–8544. [DOI] [PubMed] [Google Scholar]
  • 71.Yang HW, Kim T-M, Song SS et al. Alternative splicing of CHEK2 and codeletion with NF2 promote chromosomal instability in meningioma. Neoplasia 2012; 14 (1): 20–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Dong X, Wang L, Taniguchi K et al. Mutations in CHEK2 associated with prostate cancer risk. The American Journal of Human Genetics 2003; 72 (2): 270–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Cybulski C, Gorski B, Huzarski T et al. CHEK2 is a multiorgan cancer susceptibility gene. The American Journal of Human Genetics 2004; 75 (6): 1131–1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Black KL, Naqvi AS, Asnani M et al. Aberrant splicing in B-cell acute lymphoblastic leukemia. Nucleic Acids Res 2019; 47 (2): 1043–1043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Tang Y, Horikawa I, Ajiro M et al. Downregulation of splicing factor SRSF3 induces p53β, an alternatively spliced isoform of p53 that promotes cellular senescence. Oncogene 2013; 32 (22): 2792–2798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Li B, Brady SW, Ma X et al. Therapy-induced mutations drive the genomic landscape of relapsed acute lymphoblastic leukemia. Blood, The Journal of the American Society of Hematology 2020; 135 (1): 41–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Meyer JA, Wang J, Hogan LE et al. Relapse-specific mutations in NT5C2 in childhood acute lymphoblastic leukemia. Nat Genet 2013; 45 (3): 290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Tzoneva G, Perez-Garcia A, Carpenter Z et al. Activating mutations in the NT5C2 nucleotidase gene drive chemotherapy resistance in relapsed ALL. Nat Med 2013; 19 (3): 368–371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Brooks CL, Gu W. p53 ubiquitination: Mdm2 and beyond. Molecular cell 2006; 21 (3): 307–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Jacob AG, O’Brien D, Singh RK et al. Stress-induced isoforms of MDM2 and MDM4 correlate with high-grade disease and an altered splicing network in pediatric rhabdomyosarcoma. Neoplasia 2013; 15 (9): 1049–IN1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Comiskey DF Jr., Jacob AG, Sanford BL et al. A novel mouse model of rhabdomyosarcoma underscores the dichotomy of MDM2-ALT1 function in vivo. Oncogene 2018; 37 (1): 95–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Comiskey DF Jr, Jacob AG, Singh RK et al. Splicing factor SRSF1 negatively regulates alternative splicing of MDM2 under damage. Nucleic Acids Res 2015; 43 (8): 4202–4218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Bartel F, Harris LC, Würl P et al. MDM2 and Its Splice Variant Messenger RNAs: Expression in Tumors and Down-Regulation Using Antisense Oligonucleotides11NIH grants CA92401 and CA21765, American Lebanese Syrian Associated Charities (LCH), Deutsche Krebshilfe eV grant 2130-Ta2, Land Sachsen-Anhalt grant 3347A/0021B, and GSGT eV (FB and HT). Molecular cancer research 2004; 2 (1): 29–35. [PubMed] [Google Scholar]
  • 84.Moll UM, Petrenko O. The MDM2-p53 interaction. Molecular cancer research 2003; 1 (14): 1001–1008. [PubMed] [Google Scholar]
  • 85.Sigalas I, Calvert AH, Anderson JJ et al. Alternatively spliced mdm2 transcripts with loss of p53 binding domain sequences: transforming ability and frequent detection in human cancer. Nat Med 1996; 2 (8): 912–917. [DOI] [PubMed] [Google Scholar]
  • 86.Avnet S, Sciacca L, Salerno M et al. Insulin receptor isoform A and insulin-like growth factor II as additional treatment targets in human osteosarcoma. Cancer Res 2009; 69 (6): 2443–2452. [DOI] [PubMed] [Google Scholar]
  • 87.Sciacca L, Mineo R, Pandini G et al. In IGF-I receptor-deficient leiomyosarcoma cells autocrine IGF-II induces cell invasion and protection from apoptosis via the insulin receptor isoform A. Oncogene 2002; 21 (54): 8240–8250. [DOI] [PubMed] [Google Scholar]
  • 88.Khurshid S, Montes M, Comiskey DF et al. Splice-switching of the insulin receptor pre-mRNA alleviates tumorigenic hallmarks in rhabdomyosarcoma. NPJ Precision Oncology 2022; 6 (1): 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Gaspar N, Hawkins DS, Dirksen U et al. Ewing sarcoma: current management and future approaches through collaboration. J Clin Oncol 2015; 33 (27): 3036–3046. [DOI] [PubMed] [Google Scholar]
  • 90.Selvanathan SP, Graham GT, Erkizan HV et al. Oncogenic fusion protein EWS-FLI1 is a network hub that regulates alternative splicing. Proceedings of the National Academy of Sciences 2015; 112 (11): E1307–E1316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Palombo R, Verdile V, Paronetto MP. Poison-Exon Inclusion in DHX9 Reduces Its Expression and Sensitizes Ewing Sarcoma Cells to Chemotherapeutic Treatment. Cells 2020; 9 (2): 328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Berger M, Dirksen U, Braeuninger A et al. Genomic EWS-FLI1 fusion sequences in Ewing sarcoma resemble breakpoint characteristics of immature lymphoid malignancies. PLoS One 2013; 8 (2): e56408–e56408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Grohar PJ, Kim S, Rangel Rivera GO et al. Functional Genomic Screening Reveals Splicing of the EWS-FLI1 Fusion Transcript as a Vulnerability in Ewing Sarcoma. Cell Rep 2016; 14 (3): 598–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Neckles C, Boer RE, Aboreden N et al. HNRNPH1-dependent splicing of a fusion oncogene reveals a targetable RNA G-quadruplex interaction. RNA 2019; 25 (12): 1731–1750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lee T, Pelletier J. The biology of DHX9 and its potential as a therapeutic target. Oncotarget 2016; 7 (27): 42716–42739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Elchuri SV, Rajasekaran S, Miles WO. RNA-sequencing of primary Retinoblastoma tumors provides new insights and challenges into tumor development. Frontiers in genetics 2018; 9: 170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Rajasekaran S, Nagarajha Selvan LD, Dotts K et al. Non-coding and coding transcriptional profiles are significantly altered in pediatric retinoblastoma tumors. Frontiers in oncology 2019; 9: 221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Yandell DW, Campbell TA, Dayton SH et al. Oncogenic point mutations in the human retinoblastoma gene: their application to genetic counseling. New England Journal of Medicine 1989; 321 (25): 1689–1695. [DOI] [PubMed] [Google Scholar]
  • 99.Sweetser DA, Grabowski EF. Pediatric Malignancies: Retinoblastoma and Wilms’ Tumor. Principles of Clinical Cancer Genetics: Springer 2010; 163–180. [Google Scholar]
  • 100.Yang Z, Wang J, Zhu R. Identification of driver genes with aberrantly alternative splicing events in pediatric patients with retinoblastoma. Mathematical Biosciences and Engineering: MBE 2020; 18 (1): 328–338. [DOI] [PubMed] [Google Scholar]
  • 101.McEvoy J, Ulyanov A, Brennan R et al. Analysis of MDM2 and MDM4 single nucleotide polymorphisms, mRNA splicing and protein expression in retinoblastoma. 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Katyal S, Glubrecht DD, Li L et al. Disabled-1 alternative splicing in human fetal retina and neural tumors. PLoS One 2011; 6 (12): e28579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Lefevre S, Chauveinc L, Stoppa-Lyonnet D et al. AT to C mutation in the polypyrimidine tract of the exon 9 splicing site of the RB1 gene responsible for low penetrance hereditary retinoblastoma. Journal of medical genetics 2002; 39 (5): e21–e21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Schubert EL, Strong LC, Hansen MF. A splicing mutation in RB1 in low penetrance retinoblastoma. Human genetics 1997; 100 (5–6): 557–563. [DOI] [PubMed] [Google Scholar]
  • 105.Rodríguez-Martín C, Cidre F, Fernández-Teijeiro A et al. Familial retinoblastoma due to intronic LINE-1 insertion causes aberrant and noncanonical mRNA splicing of the RB1 gene. Journal of human genetics 2016; 61 (5): 463–466. [DOI] [PubMed] [Google Scholar]
  • 106.Dommering CJ, Mol BM, Moll AC et al. RB1 mutation spectrum in a comprehensive nationwide cohort of retinoblastoma patients. Journal of medical genetics 2014; 51 (6): 366–374. [DOI] [PubMed] [Google Scholar]
  • 107.Zhang K, Nowak I, Rushlow D et al. Patterns of missplicing caused by RB1 gene mutations in patients with retinoblastoma and association with phenotypic expression. Human mutation 2008; 29 (4): 475–484. [DOI] [PubMed] [Google Scholar]
  • 108.Klutz M, Brockmann D, Lohmann DR. A parent-of-origin effect in two families with retinoblastoma is associated with a distinct splice mutation in the RB1 gene. The American Journal of Human Genetics 2002; 71 (1): 174–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Richter S, Vandezande K, Chen N et al. Sensitive and efficient detection of RB1 gene mutations enhances care for families with retinoblastoma. The American Journal of Human Genetics 2003; 72 (2): 253–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Jia R, Li C, McCoy JP et al. SRp20 is a proto-oncogene critical for cell proliferation and tumor induction and maintenance. Int J Biol Sci 2010; 6 (7): 806–826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ajiro M, Jia R, Yang Y et al. A genome landscape of SRSF3-regulated splicing events and gene expression in human osteosarcoma U2OS cells. Nucleic Acids Res 2016; 44 (4): 1854–1870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Jia R, Ajiro M, Yu L et al. Oncogenic splicing factor SRSF3 regulates ILF3 alternative splicing to promote cancer cell proliferation and transformation. RNA 2019; 25 (5): 630–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Kohl NE, Gee CE, Alt FW. Activated expression of the N-myc gene in human neuroblastomas and related tumors. Science 1984; 226 (4680): 1335–1337. [DOI] [PubMed] [Google Scholar]
  • 114.Zhang S, Wei JS, Li SQ et al. MYCN controls an alternative RNA splicing program in high-risk metastatic neuroblastoma. Cancer Lett 2016; 371 (2): 214–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Wong N, De Melo J, Tang D. PKM2, a central point of regulation in cancer metabolism. International journal of cell biology 2013; 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Hammes A, Guo J-K, Lutsch G et al. Two splice variants of the Wilms’ tumor 1 gene have distinct functions during sex determination and nephron formation. Cell 2001; 106 (3): 319–329. [DOI] [PubMed] [Google Scholar]
  • 117.Baudry D, Hamelin M, Cabanis M-O et al. WT1 splicing alterations in Wilms’ tumors. Clinical cancer research 2000; 6 (10): 3957–3965. [PubMed] [Google Scholar]
  • 118.Huff V Wilms’ tumours: about tumour suppressor genes, an oncogene and a chameleon gene. Nature Reviews Cancer 2011; 11 (2): 111–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Lee SB, Haber DA. Wilms tumor and the WT1 gene. Experimental cell research 2001; 264 (1): 74–99. [DOI] [PubMed] [Google Scholar]
  • 120.Ullmark T, Montano G, Gullberg U. DNA and RNA binding by the Wilms’ tumour gene 1 (WT 1) protein+ KTS and− KTS isoforms—From initial observations to recent global genomic analyses. European journal of haematology 2018; 100 (3): 229–240. [DOI] [PubMed] [Google Scholar]
  • 121.Belluti S, Rigillo G, Imbriano C. Transcription factors in cancer: when alternative splicing determines opposite cell fates. Cells 2020; 9 (3): 760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Stoll R, Lee BM, Debler EW et al. Structure of the Wilms tumor suppressor protein zinc finger domain bound to DNA. Journal of molecular biology 2007; 372 (5): 1227–1245. [DOI] [PubMed] [Google Scholar]
  • 123.Laity JH, Dyson HJ, Wright PE. Molecular basis for modulation of biological function by alternate splicing of the Wilms’ tumor suppressor protein. Proceedings of the National Academy of Sciences 2000; 97 (22): 11932–11935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Larsson SH, Charlieu J-P, Miyagawa K et al. Subnuclear localization of WT1 in splicing or transcription factor domains is regulated by alternative splicing. Cell 1995; 81 (3): 391–401. [DOI] [PubMed] [Google Scholar]
  • 125.Yamanouchi K, Ohta T, Liu Z et al. The Wilms’ Tumor Gene WT1− 17AA/− KTS Splice Variant Increases Tumorigenic Activity Through Up-Regulation of Vascular Endothelial Growth Factor in an In Vivo Ovarian Cancer Model. Translational oncology 2014; 7 (5): 580–589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Barbaux S, Niaudet P, Gubler M-C et al. Donor splice-site mutations in WT1 are responsible for Frasier syndrome. Nat Genet 1997; 17 (4): 467–470. [DOI] [PubMed] [Google Scholar]
  • 127.Klamt B, Koziell A, Poulat F et al. Frasier syndrome is caused by defective alternative splicing of WT1 leading to an altered ratio of WT1+/− KTS splice isoforms. Human molecular genetics 1998; 7 (4): 709–714. [DOI] [PubMed] [Google Scholar]
  • 128.Nishikawa T, Wojciak JM, Dyson HJ et al. RNA Binding by the KTS Splice Variants of Wilms’ Tumor Suppressor Protein WT1. Biochemistry 2020; 59 (40): 3889–3901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Tate JG, Bamford S, Jubb HC et al. COSMIC: the catalogue of somatic mutations in cancer. Nucleic Acids Res 2019; 47 (D1): D941–D947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Bouaoun L, Sonkin D, Ardin M et al. TP53 variations in human cancers: new lessons from the IARC TP53 database and genomics data. Human mutation 2016; 37 (9): 865–876. [DOI] [PubMed] [Google Scholar]
  • 131.Petrovski S, Parrott RE, Roberts JL et al. Dominant splice site mutations in PIK3R1 cause hyper IgM syndrome, lymphadenopathy and short stature. Journal of clinical immunology 2016; 36 (5): 462–471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Lucas CL, Zhang Y, Venida A et al. Heterozygous splice mutation in PIK3R1 causes human immunodeficiency with lymphoproliferation due to dominant activation of PI3K. Journal of Experimental Medicine 2014; 211 (13): 2537–2547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Perry JA, Kiezun A, Tonzi P et al. Complementary genomic approaches highlight the PI3K/mTOR pathway as a common vulnerability in osteosarcoma. Proceedings of the National Academy of Sciences 2014; 111 (51): E5564–E5573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Roussel MF, Stripay JL. Epigenetic drivers in pediatric medulloblastoma. The Cerebellum 2018; 17 (1): 28–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Cerami E, Gao J, Dogrusoz U et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. AACR; 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Gao J, Aksoy BA, Dogrusoz U et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Science signaling 2013; 6 (269): pl1–pl1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Northcott PA, Buchhalter I, Morrissy AS et al. The whole-genome landscape of medulloblastoma subtypes. Nature 2017; 547 (7663): 311–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Robinson G, Parker M, Kranenburg TA et al. Novel mutations target distinct subgroups of medulloblastoma. Nature 2012; 488 (7409): 43–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Artimo P, Jonnalagedda M, Arnold K et al. ExPASy: SIB bioinformatics resource portal. Nucleic Acids Res 2012; 40 (W1): W597–W603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Fahsold R, Hoffmeyer S, Mischung C et al. Minor lesion mutational spectrum of the entire NF1 gene does not explain its high mutability but points to a functional domain upstream of the GAP-related domain. The American Journal of Human Genetics 2000; 66 (3): 790–818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Warejko JK, Schueler M, Vivante A et al. Whole exome sequencing reveals a monogenic cause of disease in≈ 43% of 35 families with midaortic syndrome. Hypertension 2018; 71 (4): 691–699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Santoro C, Di Rocco F, Kossorotoff M et al. Moyamoya syndrome in children with neurofibromatosis type 1: Italian–French experience. American journal of medical genetics Part A 2017; 173 (6): 1521–1530. [DOI] [PubMed] [Google Scholar]
  • 143.Laycock-van Spyk S, Thomas N, Cooper DN et al. Neurofibromatosis type 1-associated tumours: their somatic mutational spectrum and pathogenesis. Human genomics 2011; 5 (6): 1–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Frankiw L, Baltimore D, Li G. Alternative mRNA splicing in cancer immunotherapy. Nature Reviews Immunology 2019; 19 (11): 675–687. [DOI] [PubMed] [Google Scholar]
  • 145.Kahles A, Lehmann K-V, Toussaint NC et al. Comprehensive analysis of alternative splicing across tumors from 8,705 patients. Cancer cell 2018; 34 (2): 211–224. e216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Jayasinghe RG, Cao S, Gao Q et al. Systematic analysis of splice-site-creating mutations in cancer. Cell Rep 2018; 23 (1): 270–281. e273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Turajlic S, Litchfield K, Xu H et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. The lancet oncology 2017; 18 (8): 1009–1021. [DOI] [PubMed] [Google Scholar]
  • 148.Smart AC, Margolis CA, Pimentel H et al. Intron retention is a source of neoepitopes in cancer. Nature biotechnology 2018; 36 (11): 1056–1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Laumont CM, Vincent K, Hesnard L et al. Noncoding regions are the main source of targetable tumor-specific antigens. Science translational medicine 2018; 10 (470). [DOI] [PubMed] [Google Scholar]
  • 150.Shen L, Zhang J, Lee H et al. RNA transcription and splicing errors as a source of cancer frameshift neoantigens for vaccines. Scientific reports 2019; 9 (1): 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Oka M, Xu L, Suzuki T et al. Aberrant splicing isoforms detected by full-length transcriptome sequencing as transcripts of potential neoantigens in non-small cell lung cancer. Genome biology 2021; 22 (1): 1–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Zheng S, Naqvi AS, Bolton-Gillespie E et al. Pipeline for discovering neoepitopes generated by alternative splicing in B-ALL. American Society of Hematology; Washington, DC; 2019. [Google Scholar]
  • 153.Blaeschke F, Paul MC, Schuhmann MU et al. Low mutational load in pediatric medulloblastoma still translates into neoantigens as targets for specific T-cell immunotherapy. Cytotherapy 2019; 21 (9): 973–986. [DOI] [PubMed] [Google Scholar]
  • 154.Rivero-Hinojosa S, Grant M, Panigrahi A et al. Proteogenomic discovery of neoantigens facilitates personalized multi-antigen targeted T cell immunotherapy for brain tumors. Nature communications 2021; 12 (1): 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Lu SX, De Neef E, Thomas JD et al. Pharmacologic modulation of RNA splicing enhances anti-tumor immunity. Cell 2021; 184 (15): 4032–4047.e4031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Druker BJ, Guilhot F, O’Brien SG et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. New England Journal of Medicine 2006; 355 (23): 2408–2417. [DOI] [PubMed] [Google Scholar]
  • 157.Hochhaus A, O’brien S, Guilhot F et al. Six-year follow-up of patients receiving imatinib for the first-line treatment of chronic myeloid leukemia. Leukemia 2009; 23 (6): 1054–1061. [DOI] [PubMed] [Google Scholar]
  • 158.Berman E, Jhanwar S, Hedvat C et al. Resistance to imatinib in patients with chronic myelogenous leukemia and the splice variant BCR-ABL135INS. Leukemia research 2016; 49: 108–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Milojkovic D, Apperley J. Mechanisms of resistance to imatinib and second-generation tyrosine inhibitors in chronic myeloid leukemia. Clinical Cancer Research 2009; 15 (24): 7519–7527. [DOI] [PubMed] [Google Scholar]
  • 160.Chandrasekhar C, Kumar PS, Sarma PVGK. Novel mutations in the kinase domain of BCR-ABL gene causing imatinib resistance in chronic myeloid leukemia patients. Scientific reports 2019; 9 (1): 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Wang B-D, Lee NH. Aberrant RNA splicing in cancer and drug resistance. Cancers 2018; 10 (11): 458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Lee T-S, Ma W, Zhang X et al. BCR-ABL alternative splicing as a common mechanism for imatinib resistance: evidence from molecular dynamics simulations. Molecular cancer therapeutics 2008; 7 (12): 3834–3841. [DOI] [PubMed] [Google Scholar]
  • 163.Ng KP, Hillmer AM, Chuah CT et al. A common BIM deletion polymorphism mediates intrinsic resistance and inferior responses to tyrosine kinase inhibitors in cancer. Nat Med 2012; 18 (4): 521–528. [DOI] [PubMed] [Google Scholar]
  • 164.Liu J, Bhadra M, Sinnakannu JR et al. Overcoming imatinib resistance conferred by the BIM deletion polymorphism in chronic myeloid leukemia with splice-switching antisense oligonucleotides. Oncotarget 2017; 8 (44): 77567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Smith LD, Leme de Calais F, Raponi M et al. Novel splice-switching oligonucleotide promotes BRCA1 aberrant splicing and susceptibility to PARP inhibitor action. International Journal of Cancer 2017; 140 (7): 1564–1570. [DOI] [PubMed] [Google Scholar]
  • 166.Feins S, Kong W, Williams EF et al. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. American journal of hematology 2019; 94 (S1): S3–S9. [DOI] [PubMed] [Google Scholar]
  • 167.Maude SL, Frey N, Shaw PA et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. New England Journal of Medicine 2014; 371 (16): 1507–1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Sotillo E, Barrett DM, Black KL et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer discovery 2015; 5 (12): 1282–1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Sciarrillo R, Wojtuszkiewicz A, Kooi IE et al. Glucocorticoid resistant pediatric acute lymphoblastic leukemia samples display altered splicing profile and vulnerability to spliceosome modulation. Cancers 2020; 12 (3): 723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Shorrock HK, Gillingwater TH, Groen EJ. Overview of current drugs and molecules in development for spinal muscular atrophy therapy. Drugs 2018; 78 (3): 293–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Cartegni L, Hastings ML, Calarco JA et al. Determinants of exon 7 splicing in the spinal muscular atrophy genes, SMN1 and SMN2. The American Journal of Human Genetics 2006; 78 (1): 63–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Wood MJA, Talbot K, Bowerman M. Spinal muscular atrophy: antisense oligonucleotide therapy opens the door to an integrated therapeutic landscape. Human Molecular Genetics 2017; 26 (R2): R151–R159. [DOI] [PubMed] [Google Scholar]
  • 173.Burghes AH, McGovern VL. Antisense oligonucleotides and spinal muscular atrophy: skipping along. Genes & development 2010; 24 (15): 1574–1579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Sivanesan S, Howell M, DiDonato C et al. Antisense oligonucleotide mediated therapy of spinal muscular atrophy. Translational neuroscience 2013; 4 (1): 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Aartsma-Rus A, Krieg AM. FDA approves eteplirsen for Duchenne muscular dystrophy: the next chapter in the eteplirsen saga. Nucleic acid therapeutics 2017; 27 (1): 1–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Gurunathan S FDA Approves Sarepta Therapeutics’ Amondys 45 for Treatment of DMD Amenable to Skipping Exon 45. 2021. Muscular Dystrophy Association; 2021. [Google Scholar]
  • 177.Hoy SM. Nusinersen: first global approval. Drugs 2017; 77 (4): 473–479. [DOI] [PubMed] [Google Scholar]
  • 178.Mendell JR, Rodino-Klapac LR, Sahenk Z et al. Eteplirsen for the treatment of Duchenne muscular dystrophy. Annals of neurology 2013; 74 (5): 637–647. [DOI] [PubMed] [Google Scholar]
  • 179.Khurshid S, Montes M, Comiskey DF et al. Splice-switching of the insulin receptor in rhabdomyosarcoma: Rescuing the IR-B isoform for better treatment options. bioRxiv 2020. [Google Scholar]
  • 180.Reilley MJ, McCoon P, Cook C et al. STAT3 antisense oligonucleotide AZD9150 in a subset of patients with heavily pretreated lymphoma: results of a phase 1b trial. Journal for immunotherapy of cancer 2018; 6 (1): 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Herkt M, Thum T. Pharmacokinetics and Proceedings in Clinical Application of Nucleic Acid Therapeutics. Molecular Therapy 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Godfrey C, Desviat LR, Smedsrød B et al. Delivery is key: lessons learnt from developing splice-switching antisense therapies. EMBO Molecular Medicine 2017; 9 (5): 545–557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Gökirmak T, Nikan M, Wiechmann S et al. Overcoming the challenges of tissue delivery for oligonucleotide therapeutics. Trends in Pharmacological Sciences 2021; 42 (7): 588–604. [DOI] [PubMed] [Google Scholar]
  • 184.Hammond SM, Aartsma-Rus A, Alves S et al. Delivery of oligonucleotide-based therapeutics: challenges and opportunities. EMBO Molecular Medicine 2021; 13 (4): e13243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Cha W, Fan R, Miao Y et al. Mesoporous silica nanoparticles as carriers for intracellular delivery of nucleic acids and subsequent therapeutic applications. Molecules 2017; 22 (5): 782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Falzarano MS, Passarelli C, Ferlini A. Nanoparticle delivery of antisense oligonucleotides and their application in the exon skipping strategy for Duchenne muscular dystrophy. Nucleic acid therapeutics 2014; 24 (1): 87–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Yuan Y, Zhang L, Cao H et al. A polyethylenimine-containing and transferrin-conjugated lipid nanoparticle system for antisense oligonucleotide delivery to AML. BioMed research international 2016; 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Santiago-Ortiz JL, Schaffer DV. Adeno-associated virus (AAV) vectors in cancer gene therapy. Journal of Controlled Release 2016; 240: 287–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Aupy P, Zarrouki F, Sandro Q et al. Long-term efficacy of AAV9-U7snRNA-mediated exon 51 skipping in mdx52 Mice. Molecular Therapy-Methods & Clinical Development 2020; 17: 1037–1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Biferi MG, Cohen-Tannoudji M, Cappelletto A et al. A new AAV10-U7-mediated gene therapy prolongs survival and restores function in an ALS mouse model. Molecular Therapy 2017; 25 (9): 2038–2052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Goyenvalle A, Vulin A, Fougerousse F et al. Rescue of dystrophic muscle through U7 snRNA-mediated exon skipping. Science 2004; 306 (5702): 1796–1799. [DOI] [PubMed] [Google Scholar]
  • 192.Bonnal SC, López-Oreja I, Valcárcel J. Roles and mechanisms of alternative splicing in cancer—Implications for care. Nature Reviews Clinical Oncology 2020; 17 (8): 457–474. [DOI] [PubMed] [Google Scholar]
  • 193.Salton M, Misteli T. Small molecule modulators of pre-mRNA splicing in cancer therapy. Trends in molecular medicine 2016; 22 (1): 28–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Joshi P, Halene S, Abdel-Wahab O. How do messenger RNA splicing alterations drive myelodysplasia? Blood, The Journal of the American Society of Hematology 2017; 129 (18): 2465–2470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Kim E, Ilagan JO, Liang Y et al. SRSF2 mutations contribute to myelodysplasia by mutant-specific effects on exon recognition. Cancer cell 2015; 27 (5): 617–630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Papaemmanuil E, Cazzola M, Boultwood J et al. Somatic SF3B1 mutation in myelodysplasia with ring sideroblasts. New England Journal of Medicine 2011; 365 (15): 1384–1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Qiu J, Zhou B, Thol F et al. Distinct splicing signatures affect converged pathways in myelodysplastic syndrome patients carrying mutations in different splicing regulators. RNA 2016; 22 (10): 1535–1549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Yoshida K, Sanada M, Shiraishi Y et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature 2011; 478 (7367): 64–69. [DOI] [PubMed] [Google Scholar]
  • 199.Effenberger KA, Urabe VK, Jurica MS. Modulating splicing with small molecular inhibitors of the spliceosome. Wiley Interdisciplinary Reviews: RNA 2017; 8 (2): e1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Shi Y, Bray W, Smith AJ et al. An exon skipping screen identifies antitumor drugs that are potent modulators of pre-mRNA splicing, suggesting new therapeutic applications. PLoS One 2020; 15 (5): e0233672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Rosso M, Okoro DE, Bargonetti J. Splice variants of MDM2 in oncogenesis. Mutant p53 and MDM2 in Cancer 2014: 247–261. [DOI] [PubMed] [Google Scholar]
  • 202.Bartel F, Meye A, Würl P et al. Amplification of the MDM2 gene, but not expression of splice variants of MDM2 MRNA, is associated with prognosis in soft tissue sarcoma. International journal of cancer 2001; 95 (3): 168–175. [DOI] [PubMed] [Google Scholar]
  • 203.Bartel F, Taylor AC, Taubert H et al. SHORT COMMUNICATION Novel mdm2 Splice Variants Identified in Pediatric Rhabdomyosarcoma Tumors and Cell Lines. Oncology Research Featuring Preclinical and Clinical Cancer Therapeutics 2001; 12 (11–12): 451–457. [DOI] [PubMed] [Google Scholar]
  • 204.Dufault MR, Betz B, Wappenschmidt B et al. Limited relevance of the CHEK2 gene in hereditary breast cancer. International journal of cancer 2004; 110 (3): 320–325. [DOI] [PubMed] [Google Scholar]
  • 205.Garrido-Martín D, Palumbo E, Guigó R et al. ggsashimi: Sashimi plot revised for browser-and annotation-independent splicing visualization. PLoS computational biology 2018; 14 (8): e1006360. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Materials

RESOURCES