Abstract
The transcriptional program and RNA splicing machinery are highly and frequently dysregulated in human cancers due to genomic and epigenomic alterations during tumorigenesis. This leads to cancer-specific dependencies on components of the transcriptional program and RNA splicing machinery, providing alternative and targetable “Achilles’ heels” for cancer treatment in the clinic. To target these vulnerabilities in cancer cells, potent and specific transcriptional CDK inhibitors and chemical compounds that impair splicing have been developed and evaluated in preclinical cancer models. Several novel combination approaches with immune or targeted therapies have also been proposed for cancer treatment. More recently, inhibitors targeting transcriptional CDKs, splicing, or PRMT5 have shown promising therapeutic potential in preclinical studies, and many of them have rapidly advanced into early clinical trials for treatment of human cancer.
Keywords: Transcription cycle, RNA processing, cancer treatment
Introduction
Cancer cells exhibit accumulated genomic alterations, such as mutations, copy number changes and gene fusions, leading ultimately to uncontrolled cell growth and aberrant gene expression [1,2]. Components of the transcription program, including transcription factors, cofactors, chromatin regulators and cis-regulatory elements, are frequently altered to support the elevated level of gene expression required by the higher demands of cancer cell metabolism, proliferation, and viability compared to normal cells [3–7]. Therefore, the transcriptional machinery is always dysregulated during tumorigenesis. Increasing evidence indicates that cancer cells are highly dependent on, or “addicted to,” certain components of the transcriptional program [3–7]. Given that cancer cells are remarkably susceptible to abnormal transcription (termed “transcription addiction”) [3], targeting the transcription cycle and RNA processing have emerged as new strategies in cancer therapy [3–7]. Many tissue-specific or master transcription factors, which play central roles in regulating gene expression, are frequently and recurrently altered in human cancers; however, with the exception of nuclear receptors, targeting of transcription factors has remained a clinical challenge [3–7]. For example, MYC amplification and hyperactivation have been observed in more than 28% of tumor specimens across the TCGA sample cohort; however, most strategies directly targeting MYC have failed in preclinical development over the last two decades. Alternatively, targeting druggable proteins involved in the transcription cycle or RNA processing has been reported as a potential therapeutic strategy in cancer treatment [3–7].
Targeting transcriptional cyclin-dependent kinases (CDKs)
The family of cyclin-dependent kinases (CDKs) is composed of serine-threonine kinases that play critical roles in controlling the cell cycle and regulating transcription [8,9]. CDKs are classified as cell cycle-related CDKs or transcriptional CDKs that involved in regulation of basal transcription. The activity of a CDK depends on its regulatory partner cyclin. Upon the binding of cyclin, a cyclin-CDK complex transmits active signaling to multiple downstream proteins through phosphorylation [8,9]. Over the past decades, the biology of CDKs and their roles in diseases, including cancer, have been extensively studied [8,9]. Cell cycle-related CDKs directly regulate the progress of cell cycle. Transcriptional CDKs regulate phosphorylation of the C-terminal domain (CTD) of RNA polymerase II (Pol II), therefore controlling gene transcriptions. Given that cancer cells are always undergoing uncontrolled proliferation and dysregulated transcription, it is unsurprising that CDKs have been considered attractive targets for cancer drug development [3–5,10,11]. Development of selective CDK inhibitors to target certain CDKs is key for the application of CDKis in the clinic. During recent years, an increasing number of selective inhibitors targeting cell cycle-regulated CDKs, for example CDK4/6, have been approved for the clinical care of patients with ER+ and HER2− advanced metastatic breast cancer [10,11]. More recently, selective inhibitors targeting transcriptional CDKs have also emerged as a new class of anti-cancer agents [3–5]. For example, CDK7, CDK8/19, CDK9, CDK11 and CDK12/13 inhibitors have shown promising therapeutic potential in preclinical cancer models as well as in early-stage clinical trials [12–17] (Table 1). However, given that more than ten transcriptional CDKs have been identified in the human genome [8,9], identification and prioritization of appropriate candidates for cancer treatment remain challenging. Recently, by analyzing recurrent copy number alterations, mutations, and fusions of 21 CDK genes across more than 10,000 human tumor specimens, we found that CDKs are pervasively subject to recurrent genomic alterations across cancers [18]. Somatic copy number alteration is dominant among the genomic changes identified in CDK genes, and copy number status is positively correlated with mRNA expression levels. Notably, the copy number alterations in cell cycle-related CDKs are distinct from those in transcriptional CDKs. Cell cycle-related CDKs tend to be recurrently amplified, while transcriptional CDKs show recurrent copy number loss across cancers (Figure 1). Although transcription addiction in cancer is well recognized, the role of copy number losses in transcriptional CDKs in tumors is still largely unknown. More intriguingly, homogenous deletion is very rare whereas hemizygous loss is the dominant form of copy number deletions in transcriptional CDKs, suggesting their important roles in maintaining cancer cell growth and survival [18]. Hemizygous loss of an essential gene in cancer could create vulnerabilities which can be targeted for cancer treatment [19–24]. Some of these CYCLOPS genes (copy number alterations yielding cancer liabilities owing to partial loss [20]) have been identified and evaluated in preclinical models [20–23]. Excitingly, the very first successful example of this concept is the approval of lenalidomide (Revlimid), which targets CK1α (located in 5q32) to treat myelodysplastic syndrome with loss of chromosome 5q [24]. Given that transcriptional CDKs are essential for cell growth and survival, targeting transcriptional CDKs would be a promising therapeutic strategy for tumor cells with hemizygous loss of transcriptional CDKs [18]. For example, recurrent and focal CDK7 losses were found in multiple cancer types, and 27.3% (2,991/10,950) of TCGA tumors showed hemizygous CDK7 copy number loss [18]. The high frequency of CDK7 hemizygous loss in adult solid tumors suggests that CDK7 inhibition may trigger treatment-induced “haploinsufficiency”. Thus, low-dose CDK7 inhibitor treatment may be sufficient to kill tumor cells which are subject to transcription addiction and show hemizygous loss of CDK7. In this setting, the treatment may thus be tolerable to normal cells harboring two copies of the CDK7 gene. Taken together, targeting transcriptional CDKs with recurrent genomic losses could be a novel cancer therapeutic strategy, and such genomic alterations may also serve as biomarkers to stratify patient populations that will benefit from the treatment [18]. Additionally, inhibition of transcriptional CDKs may preferentially suppress the expression of the genes regulated by super enhancers [3–5], such as MYC, MCL1 and RUNX1 in cancer cells, providing additional therapeutic windows in patients with tumors that depend on hyperactivation of these oncogenes.
Table 1.
List of small molecule compounds targeting transcriptional cycle or splicing process.
| Compound | Target | CID | PMID | Clinical Identifier |
|---|---|---|---|---|
| BS-181 | CDK7 | 49867929 | 19638587 | |
| LDC4297 | CDK7 | 78161839 | 25047832 | |
| THZ1 | CDK7 | 73602827 | 25043025 | |
| THZ2 | CDK7 | 78357763 | 26406377 | |
| CT7001 | CDK7 | 91844733 | 29545334 | NCT03363893 |
| YKL-5–124 | CDK7 | 121443990 | 30905681 | |
| SY-1365 | CDK7 | 118426108 | 31064851 | NCT03134638 |
| LY3405105 | CDK7 | NCT03770494 | ||
| SY-5609 | CDK7 | NCT04247126 | ||
| LY2857785 | CDK9 | 78357764 | 24688048 | |
| BAY-1251152 | CDK9 | 74767009 | NCT02635672, NCT02745743 | |
| Atuveciclib | CDK9 | 71618220 | 28961375 | NCT01938638, NCT02345382 |
| MC180295 | CDK9 | 137333456 | 30454645 | |
| NVP-2 | CDK9 | 66937006 | 29251720 | |
| AZD4573 | CDK9 | 124155204 | 31699827 | NCT04630756 |
| CCT251545 | CDK8/19 | 77050682 | 26502155 | |
| SEL-120 | CDK8/19 | 73776232 | NCT04021368 | |
| AS2863619 | CDK8/19 | 139600293 | 31653719 | |
| BI-1347 | CDK8/19 | 132180482 | 32024684 | |
| OTS964* | CDK11 | 89675898 | 31511426 | |
| THZ531 | CDK12/13 | 118025540 | 27571479 | |
| SR-4835 | CDK12/13 | 139600338 | 31668947 | |
| FR901463 | SF3B1 | 70678766 | 9031664, 9031665 | |
| FR901464 | SF3B1 | 10553647 | 9031664, 9031665 | |
| FR901465 | SF3B1 | 10673616 | 9031664, 9031665 | |
| Herboxidiene | SF3B1 | 6438496 | 12523818 | |
| Pladienolides B | SF3B1 | 16202130 | 15152802, 15152803, 15152804 | |
| Pladienolides D | SF3B1 | 52944386 | 15152802, 15152803, 15152804 | |
| spliceostatin A | SF3B1 | 10673568 | 17643111 | |
| E7107 | SF3B1 | 16202132 | 17643112 | NCT00459823, NCT00499499 |
| Meayamycin | SF3B1 | 16111971 | 17279752 | |
| Sudemycins | SF3B1 | 21344922 | ||
| H3B-8800 | SF3B1 | 92135969 | 29457796 | NCT02841540 |
| TG003 | CLK1, CLK2, CLK4 | 1893668 | 15010457 | |
| SRPIN340 | SRPK1, SRPK2 | 2797577 | 16840555 | |
| cpd-1/2/3 | SRPK1, SRPK2, CLK1, CLK2 | 25581376 | ||
| Indisulam | RBM39 | 216468 | 10508428 |
NCT00060567, NCT00165594, NCT01692197, NCT00059735, NCT00080197, NCT00165854, NCT00165880, NCT00003976, NCT00003981, NCT00014625, NCT00165867, |
| C220 | PRMT5 | 32669286 | ||
| PRT811 | PRMT5 | NCT04089449 | ||
| PRT543 | PRMT5 | NCT03886831 | ||
| GSK3368715 | PRMT1 | 90462880 | 31257072 | NCT03666988 |
| JNJ-64619178 | PRMT5 | 126637809 | NCT03573310 | |
| GSK3326595 | PRMT5 | 90241742 | 29946150 | NCT02783300, NCT03614728, NCT04676516 |
| PF-06939999 | PRMT5 | NCT03854227 | ||
CID = PubChem Compound CID; https://pubchem.ncbi.nlm.nih.gov/
PMID = PubMed Reference PMID; https://pubmed.ncbi.nlm.nih.gov/
Clinical Identifier = ClinicalTrials.gov Identifier; https://www.clinicaltrials.gov/ct2/home
OTS964 was originally identified as a TOPK (T–lymphokine-activated killer cell–originated protein kinase) inhibitor. Recent study indicated that OTS964 is potent CDK11 inhibitor.
Figure 1. Genomic alterations of CDKs/cyclins are dominantly driven by somatic copy number aberrations in cancer.

Somatic copy number aberrations of the genes encoding CDKs/cyclins were characterized across the TCGA sample cohort (33 cancer types and more than 10,000 tumor specimens) by SNP arrays. Summary of the overall G-scores, which consider both the amplitudes of the aberrations and the frequencies of their occurrence across samples, of the function-defined CDKs/cyclins in cell cycle (left panel) and transcription (right panel) pathways. The size of the bubble: overall G-score; red: gain; blue: loss.
Targeting splicing dysregulations
To maintain uncontrolled cell growth and aberrant gene expression, the RNA processing machinery is also frequently altered during tumorigenesis [6,7]. RNA splicing is one of the essential steps for gene expression, and is necessary for producing diverse transcripts required by multiple cellular functions including proliferation, survival, and differentiation [6,7]. Dysregulation and mutations of core components of the spliceosome complex and splicing regulatory proteins, as well as mutations in regulatory sequences of the pre-mRNA of cancer-associated genes, have been identified in human cancers [1,2]. Consistent with this, transcriptome-wide aberrant splicing in tumor cells has been confirmed in large-scale pan-cancer analyses. For example, significant amounts of mis-spliced transcripts were found in tumor cells compared to their normal counterparts, generating cancer-specific gene isoforms that support cancer growth and causing the cells to develop splicing dependency [25–27]. Finally, in addition to splicing-disrupting mutations, splice-site-creating mutations that generate novel splicing sites have also been identified across cancers [28]. Taken together, dysregulated splicing emerges as a targetable vulnerability for cancers, especially for cancers bearing spliceosome mutations [29–33]. In this regard, small molecules targeting abnormal splicing have been identified during the past decades (Table 1). For example, inhibitors targeting the core spliceosome, such as E7107 (analogue of pladienolide B), have been developed and tested in clinical trials [34]. However, unexpected toxicity was observed in early-stage clinical studies of splicing inhibitors, leading to the early termination of these trials. Excitingly, novel derivatives of pladienolide B compounds (e.g., H3B-8800) with lower cytotoxicity have been developed and are under clinical evaluation [32]. In addition to directly targeting the core spliceosome, recent studies reported that inhibition of PRMT5 has anti-tumor potency in vitro and in pre-clinical models in multiple cancer types [35–40]. Several PRMT5 inhibitors are currently under evaluation in clinical trials for both hematologic cancers and solid tumors (Table 1). PRMT5 is an arginine methyltransferase which methylates Sm proteins of U2 snRNP, which are crucial in maintaining splicing fidelity in cells. Knocking down PRMT5 leads to abnormal splicing, such as exon skipping and intron retention on genes related to cell survival. Notably, tumors with splicing defects (e.g., splicing factor mutations) are more sensitive to PRMT5 inhibition [35,41]. Moreover, studies showed that MYC-driven cancers are also preferentially vulnerable to spliceosome inhibition, including treatment with PRMT5 inhibitors, because many splicing regulators and snRNA assembly factors are direct transcription targets of MYC [35,42,43]. Upregulation of these factors contributes to oncogenic transformation. For example, MYC-upregulated SRSF1 promotes mammary epithelial cell transformation together with MYC [43,44]. hnRNP1 and hnRNP2 induced by MYC lead to isoform switching of target gene; they regulate alternative splicing of pyruvate kinase and increase expression of its cancer-associated PKM2 isoform [42]. In addition, MYC also directly upregulates PRMT5 transcription; the increased expression of PRMT5 may be in response to an increased demand for accurate gene splicing in cells [35,45]. Depletion of PRMT5 results in mis-splicing of genes required for the growth and survival of MYC-driven lymphomas [35]. Furthermore, by leveraging the enhanced sensitivity of MYC-driven tumors to splicing perturbations, researchers identified synthetic lethal genes for MYC such as BUD31, which is a component of the core spliceosome [46]. Depletion of BUD31 expression in MYC-driven breast cancer cells resulted in reduced cell viability and increased apoptosis. Finally, RNA-based therapy has also been explored as an approach to treat dysregulated splicing in cancer. Antisense oligonucleotides (ASOs) modulating splicing (termed splice-switching oligonucleotides, SSOs) have been effective in correcting abnormal splicing events in vivo, and were approved for the treatment of Duchenne muscular dystrophy and spinal muscular atrophy in the clinic [47,48]. Sequence-specific SSO binds to corresponding pre-mRNA at spice sites or splicing regulatory motifs, preventing the recruitment of splicing machinery. The binding of SSOs to splicing enhancers or silencers promotes exon exclusion or inclusion, respectively. Despite the success of SSOs in treating non-oncology diseases, their application in cancer treatment remains challenging. The global dysregulation of splicing in cancer makes it uncertain whether modulating only a single splicing event in cancer would have an anti-tumor effect. However, recent results have demonstrated that correcting mis-spliced BRD9 by SSO in uveal melanoma inhibited cancer cell growth in vivo and in vitro, showing the potential of SSOs in cancer therapy [49]. Taken together, targeting dysregulated splicing in cancer has been proposed as a promising approach; however, more efforts are still needed for further understanding of the underlying mechanism in the context of tumorigenesis, as well as development of more specific and tolerable splicing inhibitors.
Combination with immune and targeted therapies
Although single-agent treatment of drugs targeting dysregulated transcription and RNA processing have shown promising results in cancers, recent studies revealed the possibility of advances from a combination of CDK inhibitors or splicing inhibitors with immune and targeted therapies. For example, the combination of CDK7 inhibitor (CDK7i) with p53 activators has been reported in cancer cell in vitro [50], and the combination of CDK7i or CDK12 inhibitor (CDK12i) with targeted drugs, such as PARP inhibitors (PARPis) [18,51,52], have been evaluated in animal models and early stage of clinical trials. The PARP (Poly(ADP-ribose) polymerase) family is a group of enzymes that post-translationally modify its target proteins by conjugating polymeric chains of ADP-ribose. PARPs have been found to be involved in multiple cellular processes including DNA repair. PARPs, especially PARP1, are important in recruiting homologous recombination (HR) proteins such as BRCA1 to DNA breaks. Cells with PARP inhibition show hyper-recombinogenic phenotypes with increasing HR frequency. Multiple PARPis have been successfully applied in the treatment of HR-deficient tumors; however, their effects were limited in cancers with primary or acquired HR-proficiency. Therefore, strategies suppressing expression of HR genes (chemically induced BRCAness) have been combined with PARPis or other DNA damage agents as a novel strategy to treat HR-proficient tumors [53]. Both CDK7is and CDK12is preferentially repress expression of genes in the DNA damage repair pathways, thereby impairing the activity of HR [18,51,54]. Loss of CDK7 has been reported to be correlated with elevated sensitivities to PARPis in ovarian and breast cancer cells [18]. Decreased CDK7 activity by CDK7i treatment represses the expression of BRCA1, BRCA2 and RAD51, subsequently sensitizing HR-proficient tumors to PARP inhibition. Low-dose CDK7i treatment is sufficient to act synergistically with PARPis in preclinical models of ovarian and breast cancers. Additionally, CDK7i also represses the expression of mutant BRCA1 with restored function after secondary mutations, or blocks the expression of HR genes in the context of other resistance mechanisms. CDK12 has been reported to regulate HR gene expression by suppressing intronic polyadenylation, subsequently leading to HR-deficiency [54]. By such depletion of HR gene expression, inhibition of CDK12 sensitizes or re-sensitizes cancer cells with primary or acquired PARPi resistance to PARPi therapy; this has been demonstrated in BRCA-mutant and wildtype triple negative breast cancers [51]. Thus, combination of transcriptional CDK inhibitors and PARPis may not only benefit cancer patients with primary HR-proficient tumors, but may also overcome PARPi resistance in patients with acquired HR-proficiency.
In human cancer, altered splicing has been connected with the formation of neoantigens, which are proteins that are absent in normal tissues but specifically present on the surface of cancer cells. Multiple studies have demonstrated that peptides derived from cancer-specific splicing events serve as neoepitopes by binding to histocompatibility complex class I (MHC I) molecules [26,28,55]. These studies also revealed that peptides derived from cancer-specific neojunctions significantly increase the target space for immunotherapy [26], and splice-site-creating mutations in cancer generate more than two times the number of neoepitopes per mutation compared with non-synonymous mutations. Moreover, a higher neoantigen burden is associated with elevated immune response, and splice-site-creating mutations increase the overall immunogenicity of cancers, suggesting the potential for application of immunotherapy in these cancers. Recently, PRMT5i has been reported to enhance the tumor immune response [56]. Inhibition of PRMT5 increased expression levels of interferon and chemokines, as well as abundance of MHC I molecules through regulation of target genes, therefore modulating antigen presentation of melanoma cells. Combination of PRMT5i with immune checkpoint therapy successfully inhibited tumor growth in a melanoma mouse model, showing enhanced efficacy compared to monotherapy alone. This evidence strongly suggests the potential of combining PRMT5is and immunotherapy for further clinical development.
Conclusion
There is an urgent unmet medical need to develop novel and alternative therapeutic strategies for patients with cancer. Given that the transcriptional program and RNA splicing machinery are highly dysregulated in human cancers, resulting in cancer dependencies on specific components of the transcription and splicing machinery, targeting transcriptional CDKs and splicing are emerging as new strategies for cancer treatment. Potent and selective transcriptional CDK inhibitors and chemical compounds that impair splicing have been developed and evaluated in preclinical models of cancer. Many of them have been advanced into early stage of clinical trials as a single therapeutic agent or in combination with immune and targeted therapeutic drugs.
Acknowledgements
This work was supported, in whole or in part, by the Harry Fields Professorship. L. Zhang was supported by the US National Institutes of Health (NIH) grants (R01CA142776, R01CA190415, R01CA225929, R01CA262070, P50CA083638, and P50CA174523). X. Hu was supported by the Ovarian Cancer Research Alliance. X. Hu and Y. Zhang were supported by the Foundation for Women’s Cancer.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of interest statement
Drs. Lin Zhang and Xiaowen Hu report having received research funding from Prelude Therapeutics, AstraZeneca, and Bristol-Myers Squibb/Celgene.
References and recommended reading
- 1.Garraway LA, Lander ES: Lessons from the cancer genome. Cell 2013, 153:17–37. [DOI] [PubMed] [Google Scholar]
- 2.Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr., Kinzler KW: Cancer genome landscapes. Science 2013, 339:1546–1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Bradner JE, Hnisz D, Young RA: Transcriptional Addiction in Cancer. Cell 2017, 168:629–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Chou J, Quigley DA, Robinson TM, Feng FY, Ashworth A: Transcription-Associated Cyclin- Dependent Kinases as Targets and Biomarkers for Cancer Therapy. Cancer Discov 2020, 10:351–370. [DOI] [PubMed] [Google Scholar]
- 5.Parua PK, Fisher RP: Dissecting the Pol II transcription cycle and derailing cancer with CDK inhibitors. Nat Chem Biol 2020, 16:716–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Obeng EA, Stewart C, Abdel-Wahab O: Altered RNA Processing in Cancer Pathogenesis and Therapy. Cancer Discov 2019, 9:1493–1510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Escobar-Hoyos L, Knorr K, Abdel-Wahab O: Aberrant RNA Splicing in Cancer. Annu Rev Cancer Biol 2019, 3:167–185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Morgan DO: Principles of CDK regulation. Nature 1995, 374:131–134. [DOI] [PubMed] [Google Scholar]
- 9.Malumbres M: Cyclin-dependent kinases. Genome Biol 2014, 15:122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.O’Leary B, Finn RS, Turner NC: Treating cancer with selective CDK4/6 inhibitors. Nature Reviews Clinical Oncology 2016, 13:417–430. [DOI] [PubMed] [Google Scholar]
- 11.Sherr CJ, Beach D, Shapiro GI: Targeting CDK4 and CDK6: From Discovery to Therapy. Cancer Discov 2016, 6:353–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ali S, Heathcote DA, Kroll SH, Jogalekar AS, Scheiper B, Patel H, Brackow J, Siwicka A, Fuchter MJ, Periyasamy M, et al. : The development of a selective cyclin-dependent kinase inhibitor that shows antitumor activity. Cancer Res 2009, 69:6208–6215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kwiatkowski N, Zhang T, Rahl PB, Abraham BJ, Reddy J, Ficarro SB, Dastur A, Amzallag A, Ramaswamy S, Tesar B, et al. : Targeting transcription regulation in cancer with a covalent CDK7 inhibitor. Nature 2014, 511:616–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Cidado J, Boiko S, Proia T, Ferguson D, Criscione SW, San Martin M, Pop-Damkov P, Su N, Roamio Franklin VN, Sekhar Reddy Chilamakuri C, et al. : AZD4573 Is a Highly Selective CDK9 Inhibitor That Suppresses MCL-1 and Induces Apoptosis in Hematologic Cancer Cells. Clin Cancer Res 2020, 26:922–934. [DOI] [PubMed] [Google Scholar]
- 15.Lin A, Giuliano CJ, Palladino A, John KM, Abramowicz C, Yuan ML, Sausville EL, Lukow DA, Liu L, Chait AR, et al. : Off-target toxicity is a common mechanism of action of cancer drugs undergoing clinical trials. Sci Transl Med 2019, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Zhang T, Kwiatkowski N, Olson CM, Dixon-Clarke SE, Abraham BJ, Greifenberg AK, Ficarro SB, Elkins JM, Liang Y, Hannett NM, et al. : Covalent targeting of remote cysteine residues to develop CDK12 and CDK13 inhibitors. Nat Chem Biol 2016, 12:876–884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Dale T, Clarke PA, Esdar C, Waalboer D, Adeniji-Popoola O, Ortiz-Ruiz MJ, Mallinger A, Samant RS, Czodrowski P, Musil D, et al. : A selective chemical probe for exploring the role of CDK8 and CDK19 in human disease. Nat Chem Biol 2015, 11:973–980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Shan W, Yuan J, Hu Z, Jiang J, Wang Y, Loo N, Fan L, Tang Z, Zhang T, Xu M, et al. : Systematic Characterization of Recurrent Genomic Alterations in Cyclin-Dependent Kinases Reveals Potential Therapeutic Strategies for Cancer Treatment. Cell Rep 2020, 32:107884. [DOI] [PMC free article] [PubMed] [Google Scholar]; • This study comprehensively analyzed recurrent genomic alterations of CDK/cyclin genes in more than 10,000 TCGA tumor specimens. It provides genomic information for identifying and prioritizing the targets for CDK-targeting drug development.
- 19.Frei E, 3rd: Gene deletion: a new target for cancer chemotherapy. Lancet 1993, 342:662–664. [DOI] [PubMed] [Google Scholar]
- 20.Nijhawan D, Zack TI, Ren Y, Strickland MR, Lamothe R, Schumacher SE, Tsherniak A, Besche HC, Rosenbluh J, Shehata S, et al. : Cancer vulnerabilities unveiled by genomic loss. Cell 2012, 150:842–854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Paolella BR, Gibson WJ, Urbanski LM, Alberta JA, Zack TI, Bandopadhayay P, Nichols CA, Agarwalla PK, Brown MS, Lamothe R, et al. : Copy-number and gene dependency analysis reveals partial copy loss of wild-type SF3B1 as a novel cancer vulnerability. Elife 2017, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Nichols CA, Gibson WJ, Brown MS, Kosmicki JA, Busanovich JP, Wei H, Urbanski LM, Curimjee N, Berger AC, Gao GF, et al. : Loss of heterozygosity of essential genes represents a widespread class of potential cancer vulnerabilities. Nat Commun 2020, 11:2517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Rendo V, Stoimenov I, Mateus A, Sjoberg E, Svensson R, Gustavsson AL, Johansson L, Ng A, O’Brien C, Giannakis M, et al. : Exploiting loss of heterozygosity for allele-selective colorectal cancer chemotherapy. Nat Commun 2020, 11:1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kronke J, Fink EC, Hollenbach PW, MacBeth KJ, Hurst SN, Udeshi ND, Chamberlain PP, Mani DR, Man HW, Gandhi AK, et al. : Lenalidomide induces ubiquitination and degradation of CK1alpha in del(5q) MDS. Nature 2015, 523:183–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Dvinge H, Bradley RK: Widespread intron retention diversifies most cancer transcriptomes. Genome Med 2015, 7:45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kahles A, Lehmann KV, Toussaint NC, Huser M, Stark SG, Sachsenberg T, Stegle O, Kohlbacher O, Sander C, Cancer Genome Atlas Research N, et al.: Comprehensive Analysis of Alternative Splicing Across Tumors from 8,705 Patients. Cancer Cell 2018, 34:211–224 e216. [DOI] [PMC free article] [PubMed] [Google Scholar]; • This study described landscape of alternative splicing across 8,705 cancer patients. The drivers of abnormal splicing and neojunctions that specific in cancer were identified by this study.
- 27.Group PTC, Calabrese C, Davidson NR, Demircioglu D, Fonseca NA, He Y, Kahles A, Lehmann KV, Liu F, Shiraishi Y, et al. : Genomic basis for RNA alterations in cancer. Nature 2020, 578:129–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Jayasinghe RG, Cao S, Gao Q, Wendl MC, Vo NS, Reynolds SM, Zhao Y, Climente-Gonzalez H, Chai S, Wang F, et al. : Systematic Analysis of Splice-Site-Creating Mutations in Cancer. Cell Rep 2018, 23:270–281 e273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lee SC, Dvinge H, Kim E, Cho H, Micol JB, Chung YR, Durham BH, Yoshimi A, Kim YJ, Thomas M, et al. : Modulation of splicing catalysis for therapeutic targeting of leukemia with mutations in genes encoding spliceosomal proteins. Nat Med 2016, 22:672–678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Obeng EA, Chappell RJ, Seiler M, Chen MC, Campagna DR, Schmidt PJ, Schneider RK, Lord AM, Wang L, Gambe RG, et al. : Physiologic Expression of Sf3b1(K700E) Causes Impaired Erythropoiesis, Aberrant Splicing, and Sensitivity to Therapeutic Spliceosome Modulation. Cancer Cell 2016, 30:404–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Shirai CL, White BS, Tripathi M, Tapia R, Ley JN, Ndonwi M, Kim S, Shao J, Carver A, Saez B, et al. : Mutant U2AF1-expressing cells are sensitive to pharmacological modulation of the spliceosome. Nat Commun 2017, 8:14060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Seiler M, Yoshimi A, Darman R, Chan B, Keaney G, Thomas M, Agrawal AA, Caleb B, Csibi A, Sean E, et al. : H3B-8800, an orally available small-molecule splicing modulator, induces lethality in spliceosome-mutant cancers. Nat Med 2018, 24:497–504. [DOI] [PMC free article] [PubMed] [Google Scholar]; • This study identified a novel small molecule H3B-8800 (a modulator of SF3B complex) which preferentilly targets cancer cells haboring spliceosome mutations. H3B-8800 is currently under early stage of clinical testing.
- 33.Wang E, Lu SX, Pastore A, Chen X, Imig J, Chun-Wei Lee S, Hockemeyer K, Ghebrechristos YE, Yoshimi A, Inoue D, et al. : Targeting an RNA-Binding Protein Network in Acute Myeloid Leukemia. Cancer Cell 2019, 35:369–384 e367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Eskens FA, Ramos FJ, Burger H, O’Brien JP, Piera A, de Jonge MJ, Mizui Y, Wiemer EA, Carreras MJ, Baselga J, et al. : Phase I pharmacokinetic and pharmacodynamic study of the first-in-class spliceosome inhibitor E7107 in patients with advanced solid tumors. Clin Cancer Res 2013, 19:6296–6304. [DOI] [PubMed] [Google Scholar]
- 35.Koh CM, Bezzi M, Low DH, Ang WX, Teo SX, Gay FP, Al-Haddawi M, Tan SY, Osato M, Sabo A, et al. : MYC regulates the core pre-mRNA splicing machinery as an essential step in lymphomagenesis. Nature 2015, 523:96–100. [DOI] [PubMed] [Google Scholar]
- 36.Chan-Penebre E, Kuplast KG, Majer CR, Boriack-Sjodin PA, Wigle TJ, Johnston LD, Rioux N, Munchhof MJ, Jin L, Jacques SL, et al. : A selective inhibitor of PRMT5 with in vivo and in vitro potency in MCL models. Nat Chem Biol 2015, 11:432–437. [DOI] [PubMed] [Google Scholar]
- 37.Braun CJ, Stanciu M, Boutz PL, Patterson JC, Calligaris D, Higuchi F, Neupane R, Fenoglio S, Cahill DP, Wakimoto H, et al. : Coordinated Splicing of Regulatory Detained Introns within Oncogenic Transcripts Creates an Exploitable Vulnerability in Malignant Glioma. Cancer Cell 2017, 32:411–426 e411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Fedoriw A, Rajapurkar SR, O’Brien S, Gerhart SV, Mitchell LH, Adams ND, Rioux N, Lingaraj T, Ribich SA, Pappalardi MB, et al. : Anti-tumor Activity of the Type I PRMT Inhibitor, GSK3368715, Synergizes with PRMT5 Inhibition through MTAP Loss. Cancer Cell 2019, 36:100–114 e125. [DOI] [PubMed] [Google Scholar]
- 39.Radzisheuskaya A, Shliaha PV, Grinev V, Lorenzini E, Kovalchuk S, Shlyueva D, Gorshkov V, Hendrickson RC, Jensen ON, Helin K: PRMT5 methylome profiling uncovers a direct link to splicing regulation in acute myeloid leukemia. Nat Struct Mol Biol 2019, 26:999–1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Fong JY, Pignata L, Goy PA, Kawabata KC, Lee SC, Koh CM, Musiani D, Massignani E, Kotini AG, Penson A, et al. : Therapeutic Targeting of RNA Splicing Catalysis through Inhibition of Protein Arginine Methylation. Cancer Cell 2019, 36:194–209 e199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hamard PJ, Santiago GE, Liu F, Karl DL, Martinez C, Man N, Mookhtiar AK, Duffort S, Greenblatt S, Verdun RE, et al. : PRMT5 Regulates DNA Repair by Controlling the Alternative Splicing of Histone-Modifying Enzymes. Cell Rep 2018, 24:2643–2657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.David CJ, Chen M, Assanah M, Canoll P, Manley JL: HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature 2010, 463:364–368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Das S, Anczukow O, Akerman M, Krainer AR: Oncogenic splicing factor SRSF1 is a critical transcriptional target of MYC. Cell Rep 2012, 1:110–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Anczukow O, Rosenberg AZ, Akerman M, Das S, Zhan L, Karni R, Muthuswamy SK, Krainer AR: The splicing factor SRSF1 regulates apoptosis and proliferation to promote mammary epithelial cell transformation. Nat Struct Mol Biol 2012, 19:220–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Lee SC, Abdel-Wahab O: Therapeutic targeting of splicing in cancer. Nat Med 2016, 22:976–986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Hsu TY, Simon LM, Neill NJ, Marcotte R, Sayad A, Bland CS, Echeverria GV, Sun T, Kurley SJ, Tyagi S, et al. : The spliceosome is a therapeutic vulnerability in MYC-driven cancer. Nature 2015, 525:384–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Finkel RS, Mercuri E, Darras BT, Connolly AM, Kuntz NL, Kirschner J, Chiriboga CA, Saito K, Servais L, Tizzano E, et al. : Nusinersen versus Sham Control in Infantile-Onset Spinal Muscular Atrophy. N Engl J Med 2017, 377:1723–1732. [DOI] [PubMed] [Google Scholar]
- 48.Mendell JR, Rodino-Klapac LR, Sahenk Z, Roush K, Bird L, Lowes LP, Alfano L, Gomez AM, Lewis S, Kota J, et al. : Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol 2013, 74:637–647. [DOI] [PubMed] [Google Scholar]
- 49.Inoue D, Chew GL, Liu B, Michel BC, Pangallo J, D’Avino AR, Hitchman T, North K, Lee SC, Bitner L, et al. : Spliceosomal disruption of the non-canonical BAF complex in cancer. Nature 2019, 574:432–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kalan S, Amat R, Schachter MM, Kwiatkowski N, Abraham BJ, Liang Y, Zhang T, Olson CM, Larochelle S, Young RA, et al. : Activation of the p53 Transcriptional Program Sensitizes Cancer Cells to Cdk7 Inhibitors. Cell Rep 2017, 21:467–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Johnson SF, Cruz C, Greifenberg AK, Dust S, Stover DG, Chi D, Primack B, Cao S, Bernhardy AJ, Coulson R, et al. : CDK12 Inhibition Reverses De Novo and Acquired PARP Inhibitor Resistance in BRCA Wild-Type and Mutated Models of Triple-Negative Breast Cancer. Cell Rep 2016, 17:2367–2381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Iniguez AB, Stolte B, Wang EJ, Conway AS, Alexe G, Dharia NV, Kwiatkowski N, Zhang TH, Abraham BJ, Mora J, et al. : EWS/FLI Confers Tumor Cell Synthetic Lethality to CDK12 Inhibition in Ewing Sarcoma. Cancer Cell 2018, 33:202–+. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Pilie PG, Tang C, Mills GB, Yap TA: State-of-the-art strategies for targeting the DNA damage response in cancer. Nat Rev Clin Oncol 2019, 16:81–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Dubbury SJ, Boutz PL, Sharp PA: CDK12 regulates DNA repair genes by suppressing intronic polyadenylation. Nature 2018, 564(7734):141–145. [DOI] [PMC free article] [PubMed] [Google Scholar]; • This study demontrated molecular mechanism by which CDK12 regulates HR genes through modulation of intronic polyadenylation. They reported that the intronic polyadenylation sites in HR genes are sensitive to CDK12 inhibition.
- 55.Smart AC, Margolis CA, Pimentel H, He MX, Miao D, Adeegbe D, Fugmann T, Wong KK, Van Allen EM: Intron retention is a source of neoepitopes in cancer. Nat Biotechnol 2018, 36:1056–1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kim H, Kim H, Feng Y, Li Y, Tamiya H, Tocci S, Ronai ZA: PRMT5 control of cGAS/STING and NLRC5 pathways defines melanoma response to antitumor immunity. Sci Transl Med 2020, 12. [DOI] [PMC free article] [PubMed] [Google Scholar]; • This study showed that PRMT5 inhibitor enhances anti-tumor immune response, indicating a potential combination of PRMT5 inhibition and immunotherapy in cancer treatment.
