Options for the development of colorectal cancer immunotherapy

Cover Page


Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

In colorectal cancer immunotherapy, the use of antibodies against the PD-1/PD-L1 checkpoints showed low efficacy and the development of a number of side effects with damage to the liver, lung, and thyroid gland. For this reason, to stimulate the antitumor immune response, it is necessary to search for other targets, which can be used as retroelements. Epigenetic activation of their expression with inhibitors of histone methyltransferases and deoxyribonucleic acids (DNA) leads to the formation of double-stranded ribonucleic acids (RNA) that stimulate the antiviral response of interferon, which causes apoptosis of tumor cells. This method of viral mimicry shows an objective response in colorectal cancer and other malignant neoplasms. However, activation of retrotransposons is an inducer of carcinogenesis and a necessary condition for clonal evolution and the development of chemoresistance. Therefore, the most rational combination of the method of viral mimicry is with selective inhibition of retroelements involved in the pathogenesis of colorectal cancer. For this purpose, specific miRNAs, that recruit DNA methyltransferases to the loci of the location of retroelements due to the complementarity of nucleotide sequences, which is due to their evolutionary relationship, can be used. An analysis of the scientific literature revealed 28 miRNAs derived from transposons and associated with colorectal cancer, some of which exhibit oncosuppressive activity, while others exhibit oncogenic activity. These miRNAs can be used as guides for epigenetic effects on retroelements involved in colorectal cancer carcinogenesis.

Full Text

Introduction

According to the International Agency for Research on Cancer (https://gco.iarc.fr), colorectal cancer (CRC) is the third most common malignant neoplasm (MNP) worldwide. In 2020, CRC affects 24.8 per 100,000 people. In the Russian Federation, colon, rectal, rectosigmoid junction, and anal cancers accounted for 12.3% of all MNPs [1]. Therefore, effective methods for treating CRC are urgently needed.

Immunotherapy is a promising direction for cancer treatment. The progression of carcinogenesis is caused by the immune response being eluded because of the activation of negative regulatory pathways, known as control points. Two known control points are programed cell death (PD-1) and cytotoxic T-lymphocyte protein 4 (CTLA4).

Upon activation, T cells express the cell surface receptor PD-1, which binds to either PD-L1 or PD-L2 on tumor cells, resulting in the suppression of T-lymphocyte activity. Antibodies that block the interaction between PD-L1 and PD-1 activate the immune antiviral response to MNP cells. However, because PD-L1 and PD-L2 ligands are also present in normal cells [2], autoimmune reactions provoked by anti-PD-1/PD-L1 drugs cannot be ruled out.

In addition, meta-analyses have shown that anti-PD-1/PD-L1 medications can cause liver damage, immune-related pneumonitis, and thyroid dysfunction (particularly hypothyroidism) [3–5]. When coadministered with BRAF and MEK inhibitors, anti-PD-L1/PD-1 significantly increases the risk of fever, asthenia, myalgia, arthralgia, hypothyroidism, and liver damage, as evidenced by changes in the levels of aspartate aminotransferase and alanine aminotransferase [6].

The effectiveness of anti-PD-L1/PD-1 monotherapy is limited. These treatments are generally ineffective for patients with CRC and only affect tumors with high microsatellite instability (which accounts for approximately 15% of all CRC cases) and total mutational load. Objective response was observed in only 31% of patients within 12 months of therapy [7].

Resistance to anti-PD-L1/PD-1 develops because of several factors, including severe T-cell depletion, activation of alternative immune checkpoints, suppression of major histocompatibility complex class I molecules, and production of metabolites such as PEG2, ROS, and IDO that suppress T-cell antitumor response by MNP cells. In addition, various immunosuppressive factors, such as interleukin-6 and interleukin-10, transforming growth factor-β, vascular endothelial growth factor, and ROS, contribute to this resistance [8]. To improve immunotherapy for MNPs, new ways that target specific aspects involved in carcinogenesis are needed. A promising approach is the use of retroelements (RE) found in the human genome, which are dysregulated in CRC.

REs are mobile genetic elements that move within the host genome by reverse transcription of their ribonucleic acids (RNA) and integration of the resulting complementary deoxyribonucleic acid (DNA) into new loci. Autonomous REs encode reverse transcriptase and integrase. They include long terminal repeat (LTR)-containing endogenous retroviruses (ERV) and long interspersed elements (LINE) that do not contain LTRs. Nonautonomous REs, such as short interspersed elements (SINE)— including the most common Alu elements v and complex SVAs (SINE-R, VNTR, and Alu), play a role in carcinogenesis by inserting into oncosuppressor genes, inhibiting them, and activating oncogenes [9].

Transposons and their sequence residues comprise over two-thirds of the human genome [10]. They help in controlling gene expression through cis- and trans-regulation and influence epigenetic factors, such as DNA methylation, histone modifications, and RNA interference by noncoding RNAs [9]. LINE-1 (L1) accounts for 17% of all DNA sequences and contributes to somatic movements of nonautonomous Alu elements, SVA, and processed pseudogenes in addition to retrotranspositions of their copies. ERVs account for 8% of the human genome.

Role of REs in the pathogenesis of CRC

CRC is characterized by high expression levels of REs [13] because of epigenetic dysregulation. In a 2012 study of CRC tissues, RE insertions were found in 69 of 107 samples [14]. Whole-genome sequencing of CRC samples showed the presence of multiple L1 retrotranspositions in a single tumor (14–15) with new insertions (25 events) in subclones, suggesting a role of REs in the clonal evolution of CRC [15]. Under the influence of antitumor drugs, such as etoposide, RE expression contributes to the development of chemoresistance in CRC. This was demonstrated in relation to the activation of human endogenous retrovirus (HERV)-WE1, HERV-FRD1, HERV-31, and HERV-V1 [16].

In 2019, a study analyzed 7769 samples of various MNPs and found that 70% of CRC samples had RE insertions with the activation of proto-oncogenes under the influence of integrated HERV and LINE-1 promoters [17]. The severity of RE expression affects the survival of patients with MNPs, indicating the significance of REs in the mechanisms of tumor progression [18].

Insertional mutagenesis in CRC can lead to the inactivation of oncosuppressor genes BRAF, TP53, and APC, contributing to the progression of carcinogenesis [19]. In addition, APC mutation causes hereditary adenomatous polyposis of the colon (Gardner syndrome) and contains mutagenesis hotspots for L1 insertions, which can initiate CRC development [20].

Mutagenesis hotspots for AluYa5 insertions have been identified in hereditary CRC (Lynch syndrome), which accounts for 3% of all cases [21]. A 2020 study of somatic retrotranspositions in the Pan-Cancer Analysis of Whole Genomes project showed observed events in 70% of CRC samples [11].

Studies of patients with metastatic CRC have shown high levels of ERVs because of increased expression of the TET2 DNA demethylase gene. This was accompanied by an immune and antiviral response [12]. REs are upregulated with long noncoding RNAs, which can be used for epigenetic therapy of CRC. For instance, in 22, 53% of CRC samples exhibited high expression levels of the long noncoding RNA endogenous retroviral-associated adenocarcinoma RNA (EDAVR) associated with ERV MER48. The promoter of ERV MER48 acts as a driver of EDAVR transcription.

Prospects for RE inhibition during tumor treatment

Given that hyperactivated REs play a crucial role in CRC pathogenesis, the use of nucleoside reverse-transcriptase inhibitors (NRTIs), typically employed in viral infection therapy, could represent a promising direction for treating this malignancy. Clinical trials have demonstrated the significant efficacy of NRTIs in the treatment of CRC. In addition to eliminating RE-induced genomic instability, NRTIs induce DNA damage and an interferon response to the tumor [13].

In 2015, experimental studies have demonstrated the efficacy of antiviral medications such as amantadine, ribavirin, and pleconaril in overcoming chemoresistance in CRC caused by HERV activation because of antitumor drug influence [16]. In addition, reverse-transcriptase inhibitors can enhance the effect of immune checkpoint inhibitors by suppressing the expression of telomerase reverse transcriptase [23].

The antitumor efficacy of NRTIs was determined in hormone-resistant prostate cancer [24]. A significant increase in the number and rate of cell death, as well as the inhibition of cell migration ability, was observed in breast cancer cell lines treated with the NRTIs abacavir and stavudine, particularly when used in combination with paclitaxel [25]. A meta-analysis study showed that patients with chronic viral hepatitis B taking NRTI tenofovir have a reduced risk of developing hepatocellular carcinoma [26].

Non-NRTIs, such as efavirenz, have demonstrated antitumor efficacy on pancreatic cancer cells [27]. In vitro, etravirine causes the degradation of AGR2, an endoplasmic reticulum protein secreted by the tumor microenvironment, and suppresses the proliferation, migration, and invasion of tumor cells. In mouse models, the combination of paclitaxel and etravirine more effectively inhibited the progression of ovarian cancer [28].

Histone demethylase inhibitors, such as KDM1A, show promise in suppressing RE activity in CRC by promoting RE silencing. KDM1A demethylates H3K9 and enhances the acetylation of H3K27 and methylation of H3K4, which suppresses the expression of HERV and genes containing LTR in their promoters [29].

Various tumor types exhibit high levels of histone demethylase subfamily members KDM5A/B/C/D of the JARID1 family, which demethylate H3K4me2 and H3K4me3. Selective inhibitors of these members include CPI-455 and 1,7-naphthyridines, which are effective against chemotherapy-resistant MNPs [30].

However, RE silencing does not always exert antitumor effects. For instance, the DNA methyltransferase (DMT) SETDB1 (also known as KMT1E or ESET), which forms repressive tags on H3K9, represses RE expression; however, it is an oncogene because it simultaneously inhibits the TP53 gene. Therefore, SETDB1 expression plays a crucial role in the survival of most MNPs, including CRC [31]. When considering targets for epigenetic MNP therapy, certain properties must be considered to prevent tumor growth.

Epigenetic activation of REs for CRC treatment

Although RE activation plays an important role in tumor progression and its inhibition is effective in treating CRC and other MNPs, CRC immunotherapy takes a diametrically opposite approach by stimulating antiviral immunity directed at double-stranded RNAs formed from RE transcripts. This method, known as viral mimicry, is associated with the artificial epigenetic activation of RE by inhibiting DMT and histone methyltransferase [32].

Upon the enhanced RE expression and double-stranded RNA formation, the interferon response is activated, leading to the apoptosis of MNP cells. Double-stranded RNAs can be sensed by mitochondrial antiviral signaling proteins MAVS and the Toll-like receptor TLR3 [33]. In addition, T-killers recognize HERV transcription products, leading to the destruction of MNP cells [34]. This mechanism can be used for DNA vaccination based on viral vectors [35].

The use of the DMT inhibitors 5-aza-2-deoxycytidine and 5-azacytidine (5AS) for viral mimicry was first explored in 1979 during the treatment of myeloleukemia [36]. Preclinical studies conducted in 2015 on CRC [37] and breast cancer [38] cells have also reported viral mimicry under the influence of 5-aza-2-deoxycytidine [37, 38].

In 2017, clinical trials demonstrated the efficacy of guadecitabine (SGI-110), a non-nucleoside DMT inhibitor, in patients with acute myeloblastic leukemia [39]. Tazemostat inhibits EZH, a histone methyltransferase that establishes H3K27me tags in RE loci. The efficacy of Tazemostat in clinical trials for mesothelioma, epithelioid sarcoma, and large B-cell lymphoma [40] served as the foundation for the use of EZH2 inhibitors in treating chemotherapy-resistant breast cancer [41] and prostate cancer in combination with anti-PD-1 [42]. Histone methyltransferase G9a inhibitors were found to be effective against ovarian cancer [43].

SUV39H1 histone methyltransferase, which is recruited by FBXO44 to the RE loci in the genome, may be a target for viral mimicry in antitumor therapy [44]. However, REs not only stimulate the antiviral response but also have immunosuppressive effects, which explain their key role in carcinogenesis. For instance, injecting metastatic CRC cells with activated HERV expression into Danio fish embryos significantly decreased interleukin-1 and myeloperoxidase levels [45]. This favors a selective approach for viral mimicry aimed at stimulating specific REs that have a stimulatory antiviral response and are not involved in CRC pathogenesis.

Furthermore, the search for drugs with additional antitumor properties is promising. For instance, the DMT inhibitor procaine activates PAX9, which promotes the differentiation of oral carcinoma cells, leading to their subsequent apoptosis [46].

DMT inhibitors can demethylate the promoter regions of hypermethylated tumor-suppressor genes such as MGMT, MLH1, and RASSF1A, resulting in additional antitumor effects [47].

Prospects for targeted immunotherapy for CRC

To achieve the best antitumor effect in treating CRC, specific REs involved in carcinogenesis must be inhibited, and the expression of the most immunogenic REs that do not contribute to the pathogenesis of CRC must be stimulated. Identifying these REs as targets can be performed by analyzing their interactions with microRNAs, which play a crucial role in MNP development [48].

The expression pattern of specific microRNAs in tumors may reflect the participation of mobile genetic elements in MNP development. In addition, the evolutionary relatedness of microRNAs with these elements implies the possibility of using microRNAs as guides for DMT recruitment to the region of mobile genetic element location for their inhibition. Antisense oligonucleotides, RNA sequences that are 12–25 nucleotides long that inhibit gene expression, show promise in targeting microRNAs derived from transposons with oncogenic properties [49].

Since 2008, several publications have presented data on the origin of microRNA genes from REs [50–55]. In addition, OncoMIR, an online resource, stores data on microRNAs involved in the pathogenesis of MNP-specific microRNAs [48]. By analyzing this resource and data from the scientific literature, 28 microRNAs originating from REs (Table 1) have altered expression in CRC. Of these, 14 miRNAs (miR-1249, miR-1266, miR-1271, miR-1296, miR-28, miR-320b, miR-320c, miR-326, miR-342, miR-378a, miR-502, miR-652, miR-769, and miR-942) exhibited oncosuppressor activity. They could guide the silencing of REs involved in CRC carcinogenesis because of RNA-directed DNA methylation [56]. Because 14 of these microRNAs (miR-151a, miR-192, miR-197b, miR-2355, miR-335, miR-340, miR-374a, miR-374b, miR-450b, miR-495, miR-577, miR-582, miR-708, and miR-95) are oncogenic (high expression in CRC), antisense oligonucleotides can be used as tools to inhibit them for treating CRC.

 

Table 1. Pattern of changes in the expression of microRNAs originating from transposons in colorectal adenocarcinoma

MicroRNA

Transposon, a source of microRNAs [authors]

Changes in microRNA expression [48]

miR-1249

LINE/L2 [51–53]

Decrease

miR-1266

SINE/MIR [52, 53]

Decrease

miR-1271

LINE/L2 [51–52]

Decrease

miR-1296

LINE/L2 [54]

Decrease

miR-151a

LINE/L2 [50–52, 55]

Increase

miR-192

LINE/L2 [54]

Increase

miR-1976

SINE/MIR [54]

Increase

miR-2355

LINE/RTE-BovB [52, 53, 55]

Increase

miR-28

LINE/L2 [50–52, 55]

Decrease

miR-320b

DNA/hAT-Charlie, LINE/L2 [54]

Decrease

miR-320c

LINE/L1, LINE/L2 [54]

Decrease

miR-326

DNA-TE/hAT-Tip100 [52, 53]

Decrease

miR-335

SINE/MIR [52, 53, 55]

Increase

miR-340

DNA-TE/TcMar [51–53, 55]

Increase

miR-342

SINE/tRNA-RTE [51–53, 55]

Decrease

miR-374a

LINE/L2 [52, 53, 55]

Increase

miR-374b

LINE/L2 [50, 52]

Increase

miR-378a

SINE/MIR [50, 52, 55]

Decrease

miR-450b

LINE/L1 [51–53, 55]

Increase

miR-495

ERV-L/MaLR [54]

Increase

miR-502

LINE/L2 [54]

Decrease

miR-577

LINE/L2 [52, 53]

Increase

miR-582

LINE/CR1 [50–52]

Increase

miR-652

DNA/hAT-Tip100 [50–52, 55]

Decrease

miR-708

LINE/L2 [51–53]

Increase

miR-769

LINE/CR1 [54]

Decrease

miR-942

LINE/L2 [54]

Decrease

miR-95

LINE/L2 [50, 52, 55]

Increase

Note: RNA, ribonucleic acid.

 

In addition to microRNAs, long noncoding RNAs can be used as guides in CRC immunotherapy. These RNAs, whose genes evolved from REs [57] and are involved in carcinogenesis, include the long noncoding RNA TROJAN, which evolved from HERV and is involved in the progression of triple-negative breast cancer [58]. HERVs are sources of long noncoding RNAs involved in carcinogenesis, such as HCP5 [59], PRLH1 [60], and lncMER52A [61]. In addition, specific long noncoding RNAs serve as guides for histone modification (ANRASSF1, ANRIL, BORDERLINE, Kcnq1ot1, NeST, and PINT) and DNA methylation (Airn, ecCEBP, H19, Kcnq1ot1, PAPAS, pRNA, PTENpa1-AS, TARID, and Xist) [62]. This suggests the possibility of using them for epigenetic influence on RE activity in CRC antitumor therapy.

Conclusions

The search for new immunotherapy methods for the treatment of CRC is promising because of the low efficacy and dangerous side effects of checkpoint inhibitors. Targets such as REs, whose double-chain transcription products stimulate interferon synthesis and tumor cell apoptosis, have shown clinical efficacy in CRC and other MNPs. Because RE activation is a key factor in CRC development, the safest approach is to target highly immunogenic REs that are not involved in CRC pathogenesis. RE activation drives tumorigenesis in CRC, leading to clonal evolution and chemoresistance. Therefore, selective inhibition of REs involved in CRC carcinogenesis shows promise as a complex therapy in conjunction with viral mimicry. Reverse-transcriptase inhibitors may be included in the complex treatment of preventing RE insertions. MicroRNAs that are involved in carcinogenesis and originated from REs during evolution can guide the targeting of specific REs.

 

Funding source. This study was conducted without sponsorship.
Conflict of interest. The author declares that there are no conflicts of interest in the presented article.

×

About the authors

Rustam N. Mustafin

Bashkir State Medical University

Author for correspondence.
Email: ruji79@mail.ru
ORCID iD: 0000-0002-4091-382X
SPIN-code: 4810-2534
Scopus Author ID: 56603137500
ResearcherId: S-2194-2018

Cand. Sci. (Biol.), Assoc. Prof., Depart. of Medical Genetics and Fundamental Medicine

Russian Federation, Ufa, Russia

References

  1. Zlokachestvennye novoobrazovaniya v Rossii v 2020 godu. (Malignant neoplasms in Russia in 2020.) AD Kaprin, VV Starinskiy, AO Shakhzadov, editors. Moscow: MNIOI im PA Gertsena — filial FGBU “NMITs radiologii” Minzdrava Rossii; 2021. 252 p. (In Russ.)
  2. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541(7637):321–330. doi: 10.1038/nature21349.
  3. Zhang X, Ran Y, Wang K, Zhu Y, Li J. Incidence and risk of hepatic toxicities with PD-1 inhibitors in cancer patients: A meta-analysis. Drug Des Devel Ther. 2016;10:3153–3161. doi: 10.2147/DDDT.S115493.
  4. Xu D, Liu H, Xiang M, Feng A, Tian M, Li D, Mao Y, Zhang L, Zhang S, Tian Y. The relationship between pneumonitis and programmed cell death-1/programmed cell death ligand 1 inhibitors among cancer patients: A systematic review and meta-analysis. Medicine (Baltimore). 2020;99(41):e22567. doi: 10.1097/MD.0000000000022567.
  5. Tian Y, Li R, Liu Y, Li M, Song Y, Zheng Y, Gao A, Went Q, Su G, Sun Y. The risk of immune-related thyroid dysfunction induced by PD-1/PD-L1 inhibitors in cancer patients: An updated systematic review and meta-analysis. Front Oncol. 2021;11:667650. doi: 10.3389/fonc.2021.667650.
  6. Liu Y, Zhang X, Wang G, Cui X. Triple combination therapy with PD-1/PD-L1, BRAF, and MEK inhibitor for stage III–IV melanoma: A systematic review and meta-analysis. Front Oncol. 2021;11:693655. doi: 10.3389/fonc.2021.693655.
  7. Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, Diaz Jr LA. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat Rev Gastroenterol Hepatol. 2019;16:361–375. doi: 10.1038/s41575-019-0126-x.
  8. Payandeh Z, Khalili S, Somi MH, Mard-Soltani M, Baghbanzadeh A, Hajiasgharzadeh K, Samad N, Baradaran B. PD-1/PD-L1-dependent immune response in colorectal cancer. J Cell Physiol. 2020;235(7–8):5461–5475. doi: 10.1002/jcp.29494.
  9. Mustafin RN. Influence of retroelements on oncogenes and tumor suppressors in carcinogenesis: A review. Journal of Modern Oncology. 2021;23(4):666–673. (In Russ.) doi: 10.26442/18151434.2021.4.201199.
  10. De Koning AP, Gu W, Castoe TA, Batzer MA, Pollock DD. Repetitive elements may comprise over two-thirds of the human genome. PLOS Genetics. 2011;7(12):e1002384. doi: 10.1371/journal.pgen.1002384.
  11. Rodriguez-Martin B, Alvarez EG, Baez-Ortega A, Zamora J, Supek F, Demeulemeester J, Santamarina M, Ju YS, Temes J, Garcia-Souto D, Detering H, Li Y, Rodriguez-Castro J, Dueso-Barroso A, Bruzos AL, Dentro SC, Blanco MG, Contino G, Ardeljan D, Tojo M, Roberts ND, Zumalave S, Edwards PAW, Weischenfeldt J, Puiggros M, Chong Z, Chen K, Lee EA, Wala JA, Raine K, Butler A, Waszak SM, Navarro FCP, Schumacher SE, Monlong J, Maura F, Bolli N, Bourque G, Gerstein M, Park PJ, Wedge DC, Berokhim R, Torrents D, Korbel JO, Martincorena I, Fitzgerald RC, Loo PV, Kazazian HH, Burns KH; PCAWG SVW Group; Campbell PJ, Tubio JMC; PCAWG Consortium. Pan-cancer analysis of whole genomes identifies driver rearrangements promoted by LINE-1 retrotransposition. Nat Genet. 2020;52:306–319. doi: 10.1038/s41588-019-0562-0.
  12. Topham JT, Titmuss E, Pleasance ED, Williamson LM, Karasinska JM, Culibrk L, Lee MKC, Mendis S, Denroche RE, Jang GH, Kalloger SE, Wong HL, Moore RA, Mungall AJ, O’Kane GM, Knox JJ, Gallinger S, Loree JM, Mager DL, Laskin J, Marra MA, Jones SJM, Schaeffer DF, Renouf DJ. Endogenous retrovirus transcript levels are associated with immunogenic signatures in multiple metastatic cancer types. Mol Cancer Ther. 2020;19(9):1889–1897. doi: 10.1158/1535-7163.MCT-20-0094.
  13. Rajurkar M, Parikh AR, Solovyov A, You E, Kulkarni AS, Chu C, Xu KH, Jaicks C, Taylor MS, Wu C, Aryee MJ, Hong TS, Berger SL, Walt DR, Burns KH, Park PJ, Greenbaum BD, Ting DT. Reverse transcriptase inhibition disrupts repeat element life cycle in colorectal cancer. Cancer Discov. 2022;12(6):1462–1481. doi: 10.1158/2159-8290.CD-21-1117.
  14. Solyom S, Ewing AD, Rahrmann EP, Doucet T, Nelson HH, Burns MB, Harris RS, Sigmon DF, Casella A, Erlanger B, Wheelan S, Upton KR, Shukla R, Faulkner GJ, Largaespada DA, Kazazian Jr HH. Extensive somatic L1 retrotransposition in colorectal tumors. Genome Res. 2012;22(12):2328–2338. doi: 10.1101/gr.145235.112.
  15. Pradhan B, Cajuso T, Katainen R, Sulo P, Tanskanen T, Kilpivaara O, Pitkanen E, Aaltonen LA, Kauppi L, Palin K. Detection of subclonal L1 transductions in colorectal cancer by long-distance inverse-PCR and Nanopore sequencing. Sci Rep. 2017;7(1):14521. doi: 10.1038/s41598-017-15076-3.
  16. Diaz-Carballo D, Acikelli AH, Klein J, Jastrow H, Dammann P, Wyganowski T, Guemues C, Gustmann S, Bardenheuer W, Malak S, Tefett NS, Khosrawipour V, Giger-Pabst U, Tannapfel A, Strumberg D. Therapeutic potential of antiviral drugs targeting chemorefractory colorectal adenocarcinoma cells overexpressing endogenous retroviral elements. J Exp Clin Cancer Res. 2015;34(1):81. doi: 10.1186/s13046-015-0199-5.
  17. Jang HS, Shah NM, Du AY, Dailey ZZ, Pehrsson EC, Godoy PM, Zhang D, Li D, Xing X, Kim S, O’Donnell D, Gordon JI, Wang T. Transposable elements drive widespread expression of oncogenes in human cancer. Nat Genet. 2019;51:611–617. doi: 10.1038/s41588-019-0373-3.
  18. Ye D, Jiang D, Li Y, Jin M, Chen K. The role of LINE-1 methylation in predicting survival among colorectal cancer patients: A meta-analysis. Int J Clin Oncol. 2017;22(4):749–757. doi: 10.1007/s10147-017-1106-1.
  19. Cajuso T, Sulo P, Tanskenen T, Katainen R, Taira A, Hanninen UA, Kondelin J, Forsstrom L, Valimaki N, Aavikko M, Kaasinen E, Ristimaki A, Koskensalo S, Lepisto A, Renkonen-Sinisalo L, Seppala T, Kuopio T, Bohm J, Mecklin J, Kilpivaara O, Pitkanen E, Palin K, Aaltonen LA. Retrotransposon insertions can initiate colorectal cancer and are associated with poor survival. Nat Commun. 2019;10(1):4022. doi: 10.1038/s41467-019-11770-0.
  20. Scott EC, Gardner EJ, Masood A, Chuang NT, Vertino PM, Devine SE. A hot L1 retrotransposon evades somatic repression and initiates human colorectal cancer. Genome Res. 2016;26(6):745–755. doi: 10.1101/gr.201814.115.
  21. Solassol J, Larrieux M, Leclerc J, Ducros V, Corsini C, Chiesa J, Pujol P, Rey J. Alu element insertion in the MLH1 exon 6 coding sequence as a mutation predisposing to Lynch syndrome. Hum Mutat. 2019;40(6):716–720. doi: 10.1002/humu.23725.
  22. Gibb EA, Warren RL, Wilson GW, Brown SD, Robertson GA, Morin GB, Holt RA. Activation of an endogenous retrovirus-associated long non-coding RNA in human adenocarcinoma. Genome Med. 2015;7(1):22. doi: 10.1186/s13073-015-0142-6.
  23. Bao H, Bai T, Takata K, Yokobori T, Ohnaga T, Hisada T, Maeno T, Bao P, Yoshida T, Kumakura Y, Honjo H, Sakai M, Sohda M, Fukuchi M, Altan B, Handa T, Ide M, Miyazaki T, Ogata K, Oyama T, Shimizu K, Mogi A, Asao T, Shirabe K, Kuwano H, Kaira K. High expression of carcinoembryonic antigen and telomerase reverse transcriptase in circulating tumor cells is associated with poor clinical response to the immune checkpoint inhibitor nivolumab. Oncol Lett. 2018;15(3):3061–3067. doi: 10.3892/ol.2017.7671.
  24. Sciamanna I, Sinibaldi-Vallebona P, Serafino A, Spadafora C. LINE-1-encoded reverse transcriptase as a target in cancer therapy. Front Biosci (Landmark Ed). 2018;23(7):1360–1369. doi: 10.2741/4648.
  25. Sekeroglu ZA, Sekeroglu V, Kucuk N. Effects of reverse transcriptase inhibitors on proliferation, apoptosis, and migration in breast carcinoma cells. Int J Toxicol. 2021;40(1):52–61. doi: 10.1177/1091581820961498.
  26. Choi WM, Choi J, Lim YS. Effects of Tenofovir vs Entecavir on risk of hepatocellular carcinoma in patients with chronic HBV infection: A systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2021;19(2):246–258.e9. doi: 10.1016/j.cgh.2020.05.008.
  27. Hecht M, Erber S, Harrer T, Klinker H, Roth T, Parsch H, Fiebig N, Fietkau R, Distel LV. Efavirenz has the highest anti-proliferative effect of non-nucleoside reverse transcriptase inhibitors against pancreatic cancer cells. PLoS One. 2015;10(6):e0130277. doi: 10.1371/journal.pone.0130277.
  28. Ly TTG, Yun J, Ha J, Kim Y, Jang W, Van Le TH, Rethineswaran VK, Choi J, Kim J, Min S, Lee D, Yang J, Chung J, Kwon S. Inhibitory effect of Etravirine, a non-nucleoside reverse transcriptase inhibitor, via anterior gradient protein 2 homolog degradation against ovarian cancer metastasis. Int J Mol Sci. 2022;23(2):944. doi: 10.3390/ijms23020944.
  29. Macfarla TS, Gifford WD, Agarwal S, Driscoll S, Lettieri K, Wang J, Andrews SE, Franco L, Rosenfeld MG, Ren B, Pfaff SL. Endogenous retroviruses and neighboring genes are coordinately repressed by LSD1/KDM1A. Genes Dev. 2011;25:594–607. doi: 10.1101/gad.2008511.
  30. Harmeyer KM, Facompre ND, Herlyn M, Basu D. JARID1 histone demethylases: Emerging targets in cancer. Trends Cancer. 2017;3:713–725. doi: 10.1016/j.trecan.2017.08.004.
  31. Lazaro-Camp VJ, Salari K, Meng X, Yang S. SETDB1 in cancer: overexpression and its therapeutic implications. Am J Cancer Res. 2021;11(5):1803–1827. PMID: 34094655.
  32. Chen R, Ishak CA, De Carvalho DD. Endogenous retroelements and the viral mimicry response in cancer therapy and cellular homeostasis. Cancer Discov. 2021;11(11):2707–2725. doi: 10.1158/2159-8290.
  33. Chiappinell KB, Strissel PL, Desrichard A, Li H, Henke C, Akman B, Hein A, Rote NS, Cope LM, Snyder A, Makarov V, Budhu S, Slamon DJ, Wolchok JD, Pardoll DM, Beckmann MW, Zahnow CA, Merghoub T, Chan TA, Baylin SB, Strick R. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell. 2015;162:974–86. doi: 10.1016/j.cell.2017.03.036.
  34. Attermann AS, Bjerregaard AM, Saini SK, Gronbaek K, Hadrup SR. Human endogenous retroviruses and their implication for immunotherapeutics of cancer. Ann Oncol. 2018;29:2183–2191.
  35. Bermejo AV, Ragonnaud E, Daradoumis J, Holst P. Cancer associated endogenous retroviruses: Ideal immune target for adenovirus-based immunotherapy. Int J Mol Sci. 2020;21:4843. doi: 10.3390/ijms21144843.
  36. Von Hoff DD, Schilsky R, Reichert CM, Reddick RL, Rozencweig M, Young RC, Muggia FM. Toxic effects of cis-dichlorodiammineplatinum (II) in man. Cancer Treat Rep. 1979;63(9–10):1527–1531. PMID: 387223.
  37. Roulois D, Loo Yau H, Singhania R, Wang Y, Danesh A, Shen SY, Han H, Liang G, Jones PA, Pugh TJ, O’Brien C, De Carvalho DD. DNA-demethylating agents target colorectal cancer cells by inducing viral mimicry by endogenous transcripts. Cell. 2015;162:961–973. doi: 10.1016/j.cell.2015.07.056.
  38. Chiappinelli KB, Strissel PL, Desrichard A, Li H, Henke C, Akman B, Hein A, Rote NS, Cope LM, Snyder A, Makarov V, Budhu S, Slamon DJ, Wolchok JD, Pardoll DM, Beckmann MW, Zahnow CA, Merghoub T, Chan TA, Baylin SB, Strick R. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell. 2015;162(5):974–986. doi: 10.1016/j.cell.2015.07.011.
  39. Kantarjian HM, Roboz GJ, Kropf PL, Yee KWL, O’Connell CL, Tibes R, Walsh KJ, Podoltsev NA, Griffiths EA, Jabbour E, Garcia-Manero G, Rizzieri D, Stock W, Savona MR, Rosenblat TL, Berdeja JG, Ravandi F, Rock EP, Hao Y, Azab M, Issa JJ. Guadecitabine (SGI-110) in treatment-naive patients with acute myeloid leukaemia: Phase 2 results from a multicentre, randomised, phase /2 trial. Lancet Oncol. 2017;18(10):1317–1326. doi: 10.1016/S1470-2045(17)30576-4.
  40. Hoy SM. Tazemetostat: first approval. Drugs. 2020;80:513–521. doi: 10.1007/s40265-020-01288-x.
  41. Deblois G, Tonekaboni SM, Grillo G, Martinez C, Kao YI, Tai F, Ettayebi I, Fortier A, Savage P, Fedor AN, Liu X, Guilhamon P, Lima-Fernandes E, Murison A, Kuasne H, Ba-Alawi W, Cescon DW, Arrowsmith CH, De Carvalho DD, Haibe-Kains B, Locasale JW, Park M, Lupien M. Epigenetic switch-induced viral mimicry evasion in chemotherapy-resistant breast cancer. Cancer Discov. 2020;10:1312–1329. doi: 10.1158/2159-8290.CD-19-1493.
  42. Morel KL, Sheahan AV, Burkhart DL, Baca SC, Boufaied N, Liu Y, Qiu X, Canadas I, Roehle K, Heckler M, Calagua C, Ye H, Pantelidou C, Galbo P, Panja S, Balk SP, Loda M, Labbe DP, Olson BM, Ellis L. EZH2 inhibition activates a dsRNA-STING-interferon stress axis that potentiates response to PD-1 checkpoint blockade in prostate cancer. Nat Cancer. 2021;2(4):444–456. doi: 10.1038/s43018-021-00185-w.
  43. Liu M, Thomas SL, DeWitt AK, Zhou W, Madaj ZB, Ohtani H, Baylin SB, Liang G, Jones PA. Dual inhibition of DNA and histone methyltransferases increases viral mimicry in ovarian cancer cells. Cancer Res. 2018;78:5754–5766. doi: 10.1158/0008-5472.CAN-17-3953.
  44. Shen JZ, Qiu Z, Wu Q, Finlay D, Garcia G, Sun D, Rantala J, Barshop W, Hope JL, Gimple RC, Sangfelt O, Bradley LM, Wohlschlegel J, Rich JN, Spruck C. FBXO44 promotes DNA replication-coupled repetitive element silencing in cancer cells. Cell. 2021;184:352–369. doi: 10.1016/j.cell.2020.11.042.
  45. Ferrari L, Cafora M, Rota F. Extracellular vesicles released by colorectal cancer cell lines modulate innate immune response in Zebrafish model: The possible role of human endogenous retroviruses. Int J Mol Sci. 2019;20(15):3669. doi: 10.3390/ijms20153669.
  46. Bhol CS, Mishra SR, Patil S, Sahu SK, Kirtana R, Manna S, Shanmugam MK, Sethi G, Patra SK, Bhutia SK. PAX9 reactivation by inhibiting DNA methyltransferase triggers antitumor effect in oral squamous cell carcinoma. Biochim Biophys Acta Mol Basis Dis. 2022;1868(9):166428. doi: 10.1016/j.bbadis.2022.166428.
  47. Koutsimpelas D, Pongsapich W, Heinrich U, Mann S, Mann WJ, Brieger J. Promoter methylation of MGMT, MLH1 and RASSF1A tumor suppressor genes in head and neck squamous cell carcinoma: Pharmacological genome demethylation reduces proliferation of head and neck squamous carcinoma cells. Oncol Rep. 2012;27(4):1135–1141. doi: 10.3892/or.2012.1624.
  48. Wong NW, Chen Y, Chen S, Wang X. OncomiR: And online resource for exploring pan-cancer microRNA dysregulation. Bioinformatics. 2018;34:713–715. doi: 10.1093/bioinformatics/btx627.
  49. Quemener AM, Bachelot L, Forestier A, Donnou-Fournet E, Gilot D, Galibert M. The powerful world of antisense oligonucleotides: From bench to bedside. Wiley Interdiscip Rev RNA. 2020;11(5):e1594. doi: 10.1002/wrna.1594.
  50. Piriyapongsa J, Marino-Ramirez L, Jordan IK. Origin and evolution of human microRNAs from transposable elements. Genetics. 2007;176:1323–1337.
  51. Filshtein TJ, Mackenzie CO, Dale MD, Dela-Cruz PS, Ernst DM, Frankenberger EA, He C, Heath KL, Jones AS, Jones DK, King ER, Maher MB, Mitchell TJ, Morgan RR, Sirobhushanam S, Halkyard SD, Tiwari KB, Rubin DA, Borchert GM, Larson ED. Orbid: Origin-based identification of microRNA targets. Mobile Genetic Elements. 2012;2:184–192. doi: 10.4161/mge.21617.
  52. Tempel S, Pollet N, Tahi F. NcRNAclassifier: a tool for detection and classification of transposable element sequences in RNA hairpins. BMC Bioinformatics. 2012;13: 246–258. doi: 10.1186/1471-2105-13-246.
  53. Qin S, Jin P, Zhou X, Chen L, Ma F. The role of transposable elements in the origin and evolution of microRNAs in human. PLoS One. 2015;10:e0131365. doi: 10.1371/journal.pone.0131365.
  54. Wei G, Qin S, Li W, Chen L, Ma F. MDTE DB: a database for microRNAs derived from Transposable element. IEEE/ACM Trans Comput Biol Bioinform. 2016;13:1155–1160. doi: 10.1109/TCBB.2015.2511767.
  55. Lee HE, Huh JW, Kim HS. Bioinformatics analysis of evolution and human disease related transposable element-derived microRNAs. Life (Basel). 2020;10:95. doi: 10.3390/life10060095.
  56. Watcharanurak P, Mutirangura A. Human RNA-directed DNA-methylation methylates high-mobility group box 1 protein-produced DNA gaps. Epigenomics. 2022;14(12):741–756. doi: 10.2217/epi-2022-0022.
  57. Johnson R, Guigo R. The RIDL hypothesis: transposable elements as functional domains of long noncoding RNAs. RNA. 2014;20:959–976. doi: 10.1261/rna.044560.114.
  58. Jin X, Xu XE, Jiang YZ, Liu YR, Sun W, Guo Y, Ren Y, Zuo W, Hu X, Huang S, Shen H, Lan F, He Y, Hu G, Di G, He X, Li D, Liu S, Yu K, Shao Z. The endogenous retrovirus-derived long noncoding RNA TROJAN promotes triple-negative breast cancer progression via ZMYND8 degradation. Sci Adv. 2019;5(3):eaat9820. doi: 10.1126/sciadv.aat9820.
  59. Kulski JK. Long noncoding RNA HCP5, a hybrid HLA class I endogenous retroviral gene: Structure, expression, and disease associations. Cells. 2019;8(5):480. doi: 10.3390/cells8050480.
  60. Deng B, Xu W, Wang Z, Liu C, Lin P, Li B, Huang Q, Yang J, Zhou H, Qu L. An LTR retrotransposon-derived lncRNA interacts with RNF169 to promote homologous recombination. EMBO Rep. 2019;20(11):e47650. doi: 10.15252/embr.201847650.
  61. Wu Y, Zhao Y, Huan L, Zhao J, Zhou Y, Xu L, Hu Z, Liu Y, Chen Z, Wang L, Huang S, He X, Liang L. An LTR retrotransposon-derived long noncoding RNA lncMER52A promotes hepatocellular carcinoma progression by binding p120-catenin. Cancer Res. 2020;80(5):976–987. doi: 10.1158/0008-5472.CAN-19-2115.
  62. Grammatikakis I, Panda AC, Abdelmohsen K, Gorospe M. Long noncoding RNAs (lncRNAs) and the molecular hallmarks of aging. Aging (Albany NY). 2014;6:992–1009. doi: 10.18632/aging.100710.

Supplementary files

Supplementary Files
Action
1. JATS XML

© 2023 Eco-Vector





This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies