What TERT inhibitors are in clinical trials currently?

11 March 2025
Introduction to TERT and Its Role in Cancer

Function of TERT
Telomerase reverse transcriptase (TERT) is the catalytic subunit of the enzyme telomerase, which is responsible for maintaining telomere length at the ends of chromosomes. Telomeres are repetitive nucleotide sequences that protect chromosomes from erosion during successive rounds of cell division. Under normal conditions, most somatic cells have little or no telomerase activity, which leads to progressive telomere shortening and ultimately senescence or apoptosis. However, in stem cells and in highly proliferative cells, low‐level telomerase activity is maintained to secure genomic stability. In addition to its classical role in telomere elongation, TERT has been shown to have extra‐telomeric functions that include the regulation of transcription factors, protection against oxidative stress, and modulation of cell survival pathways (e.g., NF‐κB and MYC regulation) . These additional roles help explain why TERT reactivation is nearly ubiquitous in the majority of human cancers.

TERT in Cancer Progression
In more than 90% of cancers, TERT is reactivated, an event that has wide-reaching implications for both cellular immortalization and tumor progression . The reactivation of TERT allows cancer cells to bypass the Hayflick limit by maintaining telomere length, thereby providing them with unlimited replicative potential. Moreover, TERT’s extra‐telomeric functions can stimulate pro‐survival and proliferative pathways such as NF‐κB signaling and c‐Myc regulation. These activities contribute to aggressive tumor behavior by preventing cell cycle arrest and apoptosis. The oncogenic potential of TERT reactivation has made it an attractive target for the development of anti‐cancer therapies, as inhibiting telomerase may thwart the ability of cancer cells to proliferate indefinitely .

Overview of TERT Inhibitors

Mechanism of Action
TERT inhibitors are designed to counteract the reactivation of telomerase in cancer cells. Given TERT’s dual role in telomere maintenance and the regulation of key pathways involved in cell survival, therapeutic agents have been developed with two main mechanistic approaches:

1. Direct inhibition of telomerase catalytic activity through small-molecule compounds or oligonucleotides that bind to the RNA template or active site of TERT. One of the best-known examples is imetelstat (also known as GRN163L), an oligonucleotide that binds to the RNA component of telomerase, competitively inhibiting its enzymatic function . Imetelstat is designed to prevent the addition of telomeric repeats, gradually driving cancer cells into replicative crisis. However, its clinical development, particularly in solid tumors, has been challenged by dose-limiting hematologic and hepatotoxic effects, though promising results have been observed in hematological malignancies such as myelofibrosis, essential thrombocythemia, myelodysplastic syndromes, and acute myeloid leukemia .

2. Immunotherapeutic approaches in which a peptide derived from the active site of TERT is used to “vaccinate” patients and stimulate a T-cell mediated immune reaction against TERT-expressing tumor cells. GV1001 is the most prominent example in this class. It is a 16–amino acid peptide vaccine derived from human TERT that, when administered, is intended to induce both CD4+ and CD8+ T-cell responses targeting cancer cells expressing TERT . Through this mechanism, the immune system learns to recognize and attack cancer cells that rely on telomerase activity for survival.

Other preclinical agents, such as BIBR1532, have been studied extensively for their ability to interact directly with the TERT catalytic site. Although BIBR1532 exhibits potent inhibitory activity in vitro and in animal models, it has yet to progress into clinical trials due mainly to solubility and cellular uptake issues . Thus, in the clinical realm the predominant candidates have remained imetelstat and various TERT peptide vaccines such as GV1001.

Potential Benefits and Risks
The therapeutic rationale behind inhibiting TERT is compelling because of its nearly universal expression in cancer cells and its pivotal role in enabling unlimited proliferation. The potential benefits of TERT inhibitors include:

• Inducing replicative senescence or apoptosis in cancer cells by preventing telomere elongation, which would eventually lead to tumor regression.
• Interfering with the extra‐telomeric functions of TERT that promote cell survival, such as downregulation of pro‐apoptotic signals and activation of oncogenic transcription factors (e.g., MYC and NF‐κB).
• Possibly sensitizing cancer cells to conventional chemotherapy and radiation therapy when used in combination regimens.

However, there are several risks and challenges:

• A time delay is inherent with strategies aiming to deplete telomere length—many cell divisions may be necessary before a measurable effect on tumor cell viability is observed.
• Toxicities can arise because certain normal cells, particularly those in the bone marrow and other regenerative tissues, require some level of telomerase activity. Imetelstat, for example, has demonstrated significant hematologic and hepatic toxicity in clinical studies .
• The immune approach (GV1001) runs the risk of off-target immunological effects, though current clinical data have suggested it is well tolerated in a variety of patient populations.
• Heterogeneity among tumors in terms of TERT expression and the mechanism of its reactivation may make a one-size-fits-all approach difficult to achieve.

Thus, while TERT inhibitors hold notable promise, their benefits must be balanced carefully against potential systemic toxicities and the challenge of long-term clinical efficacy.

Current Clinical Trials of TERT Inhibitors

Active Trials
At present, clinical development of TERT-targeting therapies encompasses mainly two classes of agents: direct telomerase inhibitors and immunotherapeutic vaccines that target TERT-derived epitopes. Two examples currently in clinical trials are imetelstat and GV1001.

• Imetelstat (GRN163L):
Imetelstat is a nucleotide-based inhibitor that binds competitively to the RNA template region of telomerase. It has been evaluated in several clinical studies, mostly in hematologic malignancies. One phase II trial investigated its activity in pediatric patients with recurrent central nervous system tumors, where dose-limiting hematologic toxicities (e.g., thrombocytopenia, lymphopenia, and neutropenia) were a significant concern . Although imetelstat demonstrated in vitro activity with cell cycle arrest and apoptosis in TERT-positive cells, its translation into solid tumor efficacy in adults has been modest . Nevertheless, its clinical investigations in diseases such as myelofibrosis and other blood cancers continue, demonstrating that the focus for direct telomerase inhibition remains within hematologic contexts. These trials are aimed at balancing the antitumor activity with manageable safety profiles, and they have provided valuable pharmacokinetic and pharmacodynamic data that guide subsequent dose escalation and combination strategies.

• GV1001:
GV1001, a peptide vaccine generated from the TERT sequence, is under investigation in a variety of clinical settings and represents a different format of targeting TERT—namely, via immunotherapy. Multiple studies have evaluated GV1001 in clinical trials for both oncologic and non-oncologic conditions, reflecting its diverse potential applications. Several clinical trial entries from the synapse database report investigations including:

A Phase I clinical trial assessing the safety, tolerability, and pharmacokinetic characteristics of GV1001 in healthy subjects . This trial aims to determine the maximum tolerated dose and gather preliminary safety data as a prerequisite to subsequent clinical studies.

A Phase II clinical study to evaluate the efficacy and safety of GV1001 in patients with benign prostatic hyperplasia (BPH) . Although BPH is a non-cancer condition, the trial is evaluating the biologic activity of GV1001 in modulating pathological processes, possibly leveraging its immunomodulatory or anti-Telomerase properties.

A Phase II trial to evaluate the efficacy and safety of GV1001 in Alzheimer patients . While at first glance Alzheimer’s disease may seem unrelated to cancer therapy, this trial explores the concept that telomerase activity and cellular aging play roles in neurodegeneration; herein, GV1001 might serve as a neuroprotective agent by affecting cellular senescence—and its mechanisms are being explored concurrently with the assessment of antitumor effects.

A Phase 2 clinical study to evaluate the efficacy and safety of GV1001 administered subcutaneously for the treatment of mild to moderate Alzheimer’s disease (AD) and a Phase 3 clinical trial for moderate to severe AD . These studies focus on different stages of Alzheimer’s disease, with endpoints evaluating cognitive function, long-term safety, and clinical benefits.

A Phase IIa exploratory clinical trial assessing the efficacy and safety of subcutaneous administration of GV1001 in patients with progressive supranuclear palsy (PSP) . This trial examines GV1001′s potential to slow neurodegenerative progression, again leveraging its non-canonical effects beyond telomere maintenance.

Another Phase IIa multicenter trial is investigating the safety and efficacy of GV1001 in patients with moderate Alzheimer’s disease . This adds to the body of evidence regarding the potential repurposing of telomerase-based immunotherapies beyond oncology.

In the oncology sphere, a Phase III study titled “A Phase III Study to Assess the Efficacy and Safety of GV1001 Concurrent With Gemcitabine/Capecitabine Versus Gemcitabine/Capecitabine Alone in Treating Locally Advanced and Metastatic Pancreatic Cancer Patients” is evaluating whether combining GV1001 with established chemotherapeutic regimens will provide superior clinical outcomes compared to chemotherapy alone. This study represents a strategic shift: rather than using a TERT inhibitor as a monotherapy, combining it with cytotoxic agents may exploit synergistic effects—potentially targeting both the proliferative drive and survival signaling mechanisms in cancer cells.

In summary, GV1001 is the most widely explored TERT-targeting agent in current clinical trials, and its applications span both cancer and non-cancer indications. Through these trials, investigators aim to elucidate whether immune-mediated targeting of TERT can improve survival, delay disease progression, or complement standard-of-care treatments. Other agents, such as imetelstat, continue to be evaluated, particularly in hematological malignancies, where the balance between efficacy and toxicity can be more tightly controlled.

Phases and Objectives
Clinical trials of TERT-targeting agents have been designed following standard phase I–III models, with each phase having distinct goals:

Phase I trials for TERT inhibitors primarily focus on safety, tolerability, pharmacokinetics, and establishing the maximum tolerated dose (MTD). For example, the Phase I trial of GV1001 in healthy subjects is structured to monitor immediate adverse events, determine appropriate dosing regimens, and define pharmacodynamic markers that could later be used in patient trials. Similarly, early trials of imetelstat assessed dose-escalation, hematologic parameters, and biochemical changes in telomerase activity, although much of its clinical data arise from hematological malignancy studies .

Phase II trials aim to gather preliminary evidence of efficacy while continuing to assess safety in patient populations. In the case of GV1001, multiple phase II studies are being conducted to evaluate its immunomodulatory and therapeutic effects in conditions such as Alzheimer’s disease and pancreatic cancer . The objectives in these trials include evaluating clinical endpoints (e.g., improvement in cognitive scores in AD, reduction in tumor burden and survival metrics in pancreatic cancer), as well as immunological endpoints such as T-cell responses specific to TERT epitopes. Additionally, for imetelstat, phase II trials in hematologic malignancies assess endpoints such as progression-free survival, overall response rates, and alterations in telomere length as surrogate markers of drug activity .

Phase III trials are designed to confirm efficacy in a larger set of patients and to compare the new therapy with standard-of-care treatments. For instance, the pancreatic cancer trial evaluating GV1001 in a Phase III setting is intended to validate the improved efficacy when combined with gemcitabine and capecitabine. Upon successful completion of these trials, the data gathered would support applications for regulatory approval.

These multi-phase approaches reflect the complexity of targeting TERT. Given that telomere shortening as a therapeutic endpoint may require extended treatment durations, many of the trials—especially those employing a vaccine-based strategy—employ surrogate biomarkers to provide early evidence of efficacy . Moreover, the dual role of TERT in both telomere maintenance and cell survival pathways necessitates robust analysis of pharmacodynamic markers (such as changes in NF-κB and MYC downstream signaling) in parallel with traditional endpoints like overall survival or disease progression .

Challenges and Future Prospects

Current Challenges
Despite the promise of TERT inhibition as an anti-cancer strategy, several challenges complicate clinical translation:

Delayed Onset of Action: In many cases, direct telomere erosion requires numerous cell divisions to reach a critically short state, meaning that the antitumor effects of traditional telomerase inhibitors such as imetelstat may not be immediately apparent. This delay complicates the design of clinical endpoints and may necessitate prolonged treatment durations to observe measurable benefits .

Toxicity and Off-Target Effects: The reliance of normal regenerative tissues (e.g., hematopoietic progenitors) on low levels of telomerase means that inhibitors like imetelstat can cause hematologic toxicities. Early-phase trials have reported significant dose-limiting toxicities—including thrombocytopenia, lymphopenia, and neutropenia—that restrict the maximum deliverable dose, particularly in adult solid tumor settings .

Heterogeneity Among Tumors: TERT reactivation in tumors occurs via different mechanisms (such as promoter mutations versus gene amplification), and the level of dependency on telomerase may vary between cancer types. This heterogeneity may require tailored therapeutic approaches and complicates the interpretation of trial outcomes if patient selection is not optimal .

Immunological Complexity: For TERT-based vaccines like GV1001, achieving a robust immune response without triggering autoimmunity or off-target cytotoxicity is challenging. While early results suggest that GV1001 is well tolerated, the durability of the immune response and its translation into clinical benefit remain under active investigation .

Combination Therapy Considerations: Enhancing the efficacy of TERT-targeting agents by combining them with other cytotoxic or targeted therapies is an attractive strategy. However, the combinatorial regimens add layers of complexity to trial design, as interactions between agents can lead to unexpected toxicities or alter pharmacokinetic profiles .

Future Directions in Research
Looking forward, several avenues are being actively pursued to overcome these challenges and further the clinical translation of TERT inhibitors:

Optimization of Direct Inhibitors: Researchers are working on next-generation direct inhibitors of TERT that have improved pharmacological properties relative to first-generation compounds like imetelstat. Efforts are focused on improving water solubility, cellular uptake, and specificity to reduce off-target toxicities. For example, structure-guided drug design may help identify small-molecule inhibitors with better selectivity profiles that disrupt the TERT active site or its interactions with other telomerase components without significantly impacting normal stem cells .

Enhancing Immunotherapy Approaches: For peptide vaccines such as GV1001, future research will likely aim to optimize adjuvants, dosing schedules, and delivery systems to sustain a strong and durable T-cell response. In addition, combination immunotherapies—for instance, pairing GV1001 with checkpoint inhibitors—may further potentiate the antitumor immune response while mitigating immune escape mechanisms .

Biomarker Development for Patient Selection: Given the heterogeneity in telomerase activation mechanisms among tumors, identifying reliable biomarkers (such as the presence of TERT promoter mutations, telomere length status, and expression of key downstream targets like NF‐κB and MYC) is critical. Future clinical trials may focus on stratified patient populations based on these biomarkers to ensure that TERT-targeted therapies are administered to those who are most likely to benefit .

Combination Treatments: Research into rational combination regimens is underway. The combination of TERT inhibitors with conventional chemotherapeutic agents (as seen in the pancreatic cancer trial combining GV1001 with gemcitabine and capecitabine ) is one strategy. It is expected that inhibiting telomerase may sensitize cancer cells to DNA-damaging agents, thereby enhancing overall therapeutic efficacy. Additionally, combinations with other targeted therapies (such as inhibitors of pathways downstream of TERT that influence cell cycle progression or apoptosis) represent a promising area of investigation .

Addressing Resistance Mechanisms: As with most targeted therapies, cancer cells may develop resistance mechanisms to TERT inhibition. Further research is needed to understand the molecular adaptations that confer resistance, as well as to develop strategies that either prevent or overcome such resistance. This could include designing agents that target both the canonical and extra-telomeric functions of TERT simultaneously .

Expanding the Indications: Beyond oncology, there is growing interest in studying telomerase inhibitors or activators in other disease contexts. For instance, GV1001 has been explored in neurodegenerative conditions like Alzheimer’s disease, where aging and cellular senescence play significant roles . Further exploration of TERT modulation in non-oncologic diseases may broaden the therapeutic impact of these agents.

Innovative Delivery Systems: The development of innovative drug delivery methods—such as nanoparticle encapsulation, targeted liposomes, or dendritic cell-based platforms—is another avenue to enhance the clinical efficacy of TERT inhibitors while minimizing toxicity. These delivery systems may allow for more precise targeting of tumor cells while protecting normal tissues from exposure to the inhibitor .

Detailed Conclusion
In summary, TERT inhibitors currently in clinical trials are being investigated through two primary therapeutic approaches. On one hand, direct telomerase inhibitors such as imetelstat (GRN163L) are being evaluated predominantly in hematologic malignancies where there is a relatively controllable safety profile despite notable hematologic toxicities. Imetelstat’s mechanism involves direct inhibition of the telomerase RNA template, leading to gradual telomere shortening and eventual cell death . On the other hand, the immunotherapeutic approach exemplified by GV1001 is under extensive clinical evaluation in a broad range of indications—including Alzheimer’s disease, progressive supranuclear palsy, benign prostatic hyperplasia, and metastatic pancreatic cancer. GV1001 functions as a peptide vaccine derived from the TERT sequence that aims to elicit a targeted immune response against TERT-expressing cells .

Clinical trial data gathered from early-phase studies have provided valuable insights into the safety, tolerability, and preliminary efficacy of these agents. In particular, Phase I trials for GV1001 have focused on establishing safety and proper dosing , while Phase II studies in neurodegenerative conditions and combination cancer therapies aim to refine patient selection, evaluate biomarkers of immunological response, and assess clinical endpoints relevant to each disease state. The design of these trials takes into account the delay inherent in telomere erosion therapies, the need for surrogate biomarker endpoints, and the potential for combination regimens that may synergistically enhance the antitumor effect.

Despite the clinical promise, several challenges remain. The long duration needed to observe meaningful telomere shortening, the risk of toxicity in normal regenerative tissues, and the immunological variability among patients necessitate further refinement of these approaches. Future research is focusing on next-generation inhibitors with better pharmacological profiles, improved drug delivery systems, and combination regimens designed to overcome resistance and delay tumor progression. Moreover, the development of reliable biomarkers for patient selection remains a critical factor in maximizing the therapeutic benefits of TERT inhibition.

From a broader perspective, the continued advancement in our understanding of both the canonical telomere-maintenance and extra-telomeric functions of TERT provides a solid rationale for innovative treatment strategies. The research community is actively pursuing approaches that not only target the enzyme’s activity but also harness the immune system’s capability to recognize and destroy TERT-positive cells. The convergence of immunotherapy and direct inhibition represents a holistic strategy to dismantle one of cancer’s fundamental survival mechanisms.

In conclusion, the current landscape of TERT inhibitors in clinical trials centers primarily on imetelstat and GV1001, each representing a distinct pathway to counteract TERT reactivation in cancer. Imetelstat’s direct inhibitory actions have shown potential in hematologic cancers despite significant toxicity issues, whereas GV1001 is being explored across a spectrum of indications through its vaccine-mediated activation of antitumor immune responses. As clinical data continue to accrue, these agents are poised to refine our approach to targeting cellular immortality, potentially transforming the therapeutic landscape for cancers and even extending into non-oncological indications where cellular aging is a contributing factor. The future of TERT inhibition will rely on overcoming current challenges with innovative drug design, optimized patient selection, and rational combination therapies that together promise a more effective and safer intervention for patients battling cancer and age-related diseases .

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成