Abstract

Telomerase is expressed in more than 85% of cancer cells. Tumor cells with metastatic potential may have a high telomerase activity, allowing cells to escape from the inhibition of cell proliferation due to shortened telomeres. Human telomerase primarily consists of two main components: hTERT, a catalytic subunit, and hTR, an RNA template whose sequence is complimentary to the telomeric 5′-dTTAGGG-3′ repeat. In humans, telomerase activity is typically restricted to renewing tissues, such as germ cells and stem cells, and is generally absent in normal cells. While hTR is constitutively expressed in most tissue types, hTERT expression levels are low enough that telomere length cannot be maintained, which sets a proliferative lifespan on normal cells. However, in the majority of cancers, telomerase maintains stable telomere length, thereby conferring cell immortality. Levels of hTERT mRNA are directly related to telomerase activity, thereby making it a more suitable therapeutic target than hTR. Recent data suggests that stabilization of telomeric G-quadruplexes may act to indirectly inhibit telomerase action by blocking hTR binding. Telomeric DNA has the propensity to spontaneously form intramolecular G-quadruplexes, four-stranded DNA secondary structures that are stabilized by the stacking of guanine residues in a planar arrangement. The functional roles of telomeric G-quadruplexes are not completely understood, but recent evidence suggests that they can stall the replication fork during DNA synthesis and inhibit telomere replication by preventing telomerase and related proteins from binding to the telomere. Long-term treatment with G-quadruplex stabilizers induces a gradual reduction in the length of the G-rich 3’ end of the telomere without a reduction of the total telomere length, suggesting that telomerase activity is inhibited. However, inhibition of telomerase, either directly or indirectly, has shown only moderate success in cancer patients. Another promising approach of targeting the telomere is the use of guanine-rich oligonucleotides (GROs) homologous to the 3’ telomere overhang sequence (T-oligos). T-oligos, particularly a specific 11-base oligonucleotide (5’-dGTTAGGGTTAG-3’) called T11, have been shown to induce DNA damage responses (DDRs) such as senescence, apoptosis, and cell cycle arrest in numerous cancer cell types with minimal or no cytostatic effects in normal, non-transformed cells. As a result, T-oligos and other GROs are being investigated as prospective anticancer therapeutics. Interestingly, the DDRs induced by T-oligos in cancer cells are similar to the effects seen after progressive telomere degradation in normal cells. The loss of telomeres is an important tumor suppressor mechanism that is commonly absent in transformed malignant cells, and hence, T-oligos have garnered significant interest as a novel strategy to combat cancer. However, little is known about their mechanism of action. In this review, we discuss the current understanding of how T-oligos exert their antiproliferative effects in cancer cells and their role in inhibition of telomerase. We also discuss the current understanding of telomerase in cancer and various therapeutic targets related to the telomeres and telomerase.

Highlights

  • Telomerase is expressed in more than 85% of cancer cells

  • We discuss the current understanding of telomerase in cancer and various therapeutic targets related to the telomeres and telomerase

  • We suggest that a human the reverse transcriptase catalytic subunit (TERT) inhibitor may be a more efficient therapeutic than current hTR inhibitors since, it would prohibit telomere elongation in addition to halting TERT’s non-canonical effects

Read more

Summary

Current Therapies Related to Telomeres and Telomerase

Due to its over-expression in the majority of cancers, and minimal or nonexistent expression in most somatic cells, telomerase is a unique cancer biomarker. The use of G-quadruplex–stabilizing ligands has potential applications for the development of treatments for malignant and progressive cancers These agents bind with high affinity to the 3’ single-stranded region of the telomere, facilitating formation and stabilization of the. Some of the most promising G-quadruplex–stabilizing ligands include telomestatin, BRACO-19, and RHPS4, and studies utilizing these agents have demonstrated increased G-quadruplex stability and upregulation of DDRs in cancer cells [33,35,36,37,38]. It has been known that TERT is a major component and limiting factor for telomerase activity [41,47] Based on these findings, we suggest that a human TERT (hTERT) inhibitor may be a more efficient therapeutic than current hTR inhibitors since, it would prohibit telomere elongation in addition to halting TERT’s non-canonical effects. A potentially promising telomere-related therapy is the use of guanine-rich oligonucleotides (GROs), those that are homologous to the telomere, to disrupt the telomere [1]

Telomere Homolog Oligonucleotides
Findings
Conclusions
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call