What are the therapeutic candidates targeting HDACs?

11 March 2025
Introduction to HDACsHistone deacetylases (HDACs)s) are a family of zinc‐dependent enzymes that remove acetyl groups from lysine residues on histone proteins and a wide range of non‐histone substrates. This deacetylation has a profound impact on chromatin structure, thereby controlling the “opening” or “closing” of genomic regions for transcription. HDACs are also involved in post‐translational modifications of transcription factors, chaperones, and other regulatory proteins, exerting pleiotropic effects on cell proliferation, differentiation, apoptosis, and stress responses.

Role of HDACs in Cellular Processes

HDACs assist in the dynamic regulation of gene expression by removing acetyl groups that normally decrease the positive charge on histone tails. When histones are deacetylated, the chromatin becomes more compact, leading to transcriptional suppression. This regulation of the epigenome is central to many biological processes including:
• Cell cycle progression – by controlling the transcription of cyclin-dependent kinase inhibitors such as p21 and p27.
• Apoptosis and differentiation – by altering gene expression profiles that determine cell survival and terminal differentiation programs.
• Stress response and metabolic regulation – since HDACs also act on non-histone proteins, their activity influences metabolic enzymes and pathways in a tissue‐dependent manner.

Importance of HDACs in Disease Pathology

Aberrant HDAC activity has been linked to the development and progression of many diseases. In oncology, hyperactivity or overexpression of certain HDAC isoforms leads to the repression of tumor-suppressor genes and unbalanced expression of genes involved in cell division and apoptosis. Conditions such as cutaneous T-cell lymphoma (CTCL), peripheral T-cell lymphoma (PTCL), multiple myeloma, and even a variety of solid tumors have demonstrated a dependency on HDAC-mediated epigenetic silencing. Beyond cancer, deregulated HDAC activity plays roles in neurodegenerative disorders, inflammatory conditions, and even fungal infections, thus underscoring the widespread importance of these enzymes in pathophysiology.

Therapeutic Candidates Targeting HDACs

A major avenue of drug discovery in the past two decades has focused on developing small molecule inhibitors that target HDACs. These drugs aim to reverse abnormal deacetylation, reactivate silenced tumor suppressor genes, and trigger apoptosis in cancer cells. The therapeutic candidates that target HDACs can be examined from several perspectives, ranging from the currently approved inhibitors to experimental compounds emerging from recent research.

Overview of Current HDAC Inhibitors

Currently, several HDAC inhibitors (HDACi) have received regulatory approval for specific indications, particularly hematological malignancies. For example:
• Vorinostat (also known as suberoylanilide hydroxamic acid, SAHA) was the first HDAC inhibitor approved by the FDA in 2006 for the treatment of CTCL.
• Romidepsin (FK228) emerged later as an inhibitor with a cyclic peptide structure and is approved for CTCL and PTCL.
• Belinostat, a hydroxamic acid derivative, is used in the treatment of relapsed or refractory PTCL and has demonstrated efficacy with a manageable safety profile.
• Panobinostat, approved in 2015 for the treatment of multiple myeloma, is recognized as a pan-HDAC inhibitor that targets multiple isoforms simultaneously.
• Tucidinostat (also known as chidamide) is approved in China for PTCL and may also be explored in other cancers.
In addition to these, other candidates such as entinostat, which shows selectivity for HDAC1 and HDAC3, and other HDAC inhibitors undergoing phase I/II trials have broadened the therapeutic landscape. The candidates are not only investigated as stand-alone agents but are frequently tested in combinatorial regimens with other therapies (chemotherapy, immunotherapies, or other targeted agents) to overcome resistance and enhance efficacy.

Classification of HDAC Inhibitors

Therapeutic candidates targeting HDACs are classified based on chemical structure, isoform-selectivity, and the types of enzymes they inhibit. The major classes include:

• Hydroxamic acids – This class is the most widely used, with compounds such as vorinostat, belinostat, and panobinostat. Their hydroxamic acid moiety chelates the zinc ion at the HDAC active site and provides potent, broad-spectrum inhibition by targeting class I, II, and IV enzymes.

• Cyclic peptides – Romidepsin is a representative of this class. It is a prodrug that undergoes reduction to release a free thiol group, which then binds to the zinc ion in HDAC active sites. Cyclic peptides tend to have improved potency and may offer a different toxicity profile compared to hydroxamic acids.

• Benzamides – These inhibitors such as chidamide offer more subtype selectivity, particularly toward class I HDACs, and have been shown to have a favorable safety profile in clinical studies. Benzamides can target specific isoforms and are being actively developed to reduce off-target side effects.

• Short-chain fatty acids – Naturally occurring compounds such as sodium butyrate fall into this category. Although weaker in potency compared to other HDACi, they have been studied for their chemopreventive properties and for reversing epigenetic silencing in various models.

• Sirtuin inhibitors – Although sirtuins (class III HDACs) are not zinc-dependent, there are also compounds that target these enzymes (such as nicotinamide derivatives). However, many therapeutic efforts have so far concentrated on the zinc-dependent HDAC families (classes I, II, IV) because of their involvement in cancer.

The classification reflects both the pharmacophore – a zinc-binding group, a linker that spans the tubular channel of the enzyme, and a “cap” group that interacts with the enzyme’s surface – and the unique structure of the candidate. Many ongoing studies try to improve isoform selectivity with dual or multi-targeting compounds and even explore nanoparticle-based delivery to achieve more precise targeting.

Mechanisms of Action

HDAC inhibitors function through a number of biochemical and molecular mechanisms that ultimately result in altered gene expression and cellular processes. Their mechanisms also determine how they may contribute to anticancer effects and other therapeutic benefits.

How HDAC Inhibitors Work

All HDAC inhibitors primarily work by binding to the catalytic domain of HDAC enzymes. This binding is usually mediated by a metal-chelating group (for zinc-dependent HDACs) that interferes with the enzyme’s ability to hydrolyze acetyl groups. By blocking HDAC activity, histone acetylation levels increase, leading to a more open chromatin configuration. This not only promotes the transcription of previously silenced genes but also influences non-histone protein acetylation, thereby affecting various cellular pathways. For example:

• Inhibition of HDAC enzymes results in upregulation of cell cycle inhibitors (such as p21^WAF1/CIP1), leading to cell cycle arrest at the G1/S or G2/M phases.
• HDAC inhibitors can trigger apoptosis by reactivating pro-apoptotic genes, inducing the intrinsic (mitochondrial) or extrinsic (death receptor) apoptotic pathways.
• HDAC inhibition is also associated with inhibition of angiogenesis, migration, and metastasis as inhibitors reduce the expression of genes coding for factors like VEGF and suppress epithelial-mesenchymal transition (EMT) processes.
• Beyond histones, they increase the acetylation of transcription factors (e.g., p53) and other regulators, which can stabilize these proteins, enhance their function, or even modify their interactions with other cellular partners.

This multi-level impact on the acetylome explains why HDACi can elicit a broad spectrum of biological responses. The range of responses, however, is highly dependent on the particular HDAC isoform targeted and the selected dosing schedules.

Impact on Gene Expression and Cellular Function

The consequence of HDAC inhibition is profound modulation of gene expression. This modulation can be described from both a general and specific perspective:

• General Impact: Increases in histone acetylation relax the chromatin structure. This results in the reactivation of silenced tumor suppressor genes, restoration of normal gene expression patterns, and reversal of the epigenetic changes that allow tumor cells to proliferate unchecked. A typical outcome is the upregulation of genes that cause cell cycle arrest, apoptosis, and differentiation.

• Specific Impact: Some HDAC inhibitors show selectivity toward certain isoforms, which results in a more targeted effect on gene expression. For instance, entinostat’s preferential inhibition of HDAC1 and HDAC3 selectively affects the expression of a subset of genes critical in leukemogenic and solid tumor pathways without broadly impacting normal cellular gene expression. In contrast, pan-HDAC inhibitors target multiple isoforms resulting in more widespread changes, which can include both the upregulation of a host of genes and the downregulation of others through indirect mechanisms mediated by altered transcription factor dynamics.

The alteration of gene expression is accompanied by changes in cell proliferation, apoptosis, differentiation, and DNA repair. In cancer cells, this means that cells can be forced out of a malignant state and pushed toward cell death, while in other diseases, correcting abnormal repression or activation of genes can help restore cellular homeostasis. Moreover, studies have shown that HDAC inhibition may sometimes lead to unexpected effects such as the downregulation of genes despite increased histone acetylation due to concomitant changes in the recruitment of transcriptional co-regulators.

Clinical Applications

HDAC inhibitors have made their way into clinical practice, predominantly as anticancer agents. Their approval and use in the clinical setting illustrate how targeting epigenetic regulators can lead to significant improvements in disease outcomes, particularly when conventional chemotherapy options are limited.

Approved Drugs and Their Indications

At present, the regulator-approved HDAC inhibitors primarily target hematological malignancies – a testament to the clear epigenetic underpinnings of these diseases:

• Vorinostat (SAHA) – Approved for the treatment of CTCL, it works as a broad-spectrum pan-HDAC inhibitor that modulates the acetylation state of histones and non-histone proteins, thereby affecting cell cycle, apoptosis, and differentiation.
• Romidepsin (FK228) – Its approval for CTCL and PTCL highlights its efficacy in tumors that heavily rely on HDAC-driven transcriptional repression. Its cyclic peptide structure gives it a unique profile compared to other small molecule HDAC inhibitors.
• Belinostat – Approved for relapsed or refractory PTCL, it is a hydroxamic acid derivative that has shown robust activity in lymphoma patients with manageable toxicity.
• Panobinostat – Used in combination with other therapies in multiple myeloma, this inhibitor targets a broad range of HDAC isoforms and has opened up additional paradigms for combination approaches to overcome drug resistance.
• Tucidinostat (Chidamide) – Approved in China for PTCL, this benzamide HDAC inhibitor is distinctive in its more selective profile, reducing off-target effects and enabling chronic dosing.

Other candidates such as entinostat are undergoing clinical evaluation and have been tested in various settings, including breast cancer and acute myeloid leukemia, with data suggesting that selective inhibition of HDAC1/HDAC3 can enhance the antitumor efficacy, especially in combination with immunotherapeutic agents. Clinical trials also explore HDAC inhibitors in combination with other epigenetic drugs (like DNA methyltransferase inhibitors), signal transduction modulators, and immunotherapies, which points to the evolving nature of treatment strategies involving the HDAC target.

Ongoing Clinical Trials

An important aspect of HDAC inhibitor research is the extensive range of ongoing clinical trials that test these agents as monotherapies or in combination:

• Multiple trials are evaluating pan-HDAC inhibitors like panobinostat and vorinostat in solid tumors as well as hematological malignancies.
• Selective inhibitors (e.g., chidamide, entinostat) have appeared in phase I/II studies, exploring more refined endpoints such as progression-free survival and overall response rates in cancers that have shown resistance to standard therapies.
• There is growing interest in combination strategies involving HDAC inhibitors plus immunotherapy. Early studies have shown that HDAC inhibition may upregulate immune checkpoint ligands like PD-L1 on tumor cells, making them more amenable to treatments such as anti-PD-1 therapies.
• In addition, trials examining combination regimens of HDAC inhibitors with traditional cytotoxic agents such as doxorubicin or novel targeted agents (e.g., kinase inhibitors) are underway. The aim is to exploit synergistic mechanisms where the epigenetic reversion provided by HDAC inhibition makes tumor cells more sensitive to other drugs.

The clinical pipeline is dynamic, with numerous studies registered on databases such as clinicaltrials.gov. These trials span indications including refractory cancers, neurodegenerative diseases and inflammatory conditions—and collectively they are attempting to address dosage, safety, and the biological underpinnings that may distinguish responders from non-responders.

Challenges and Future Directions

Despite the promising therapeutic candidates targeting HDACs, there remain significant challenges and opportunities for future research.

Limitations of Current HDAC Inhibitors

One of the most pressing issues with current HDAC inhibitors is their limited isoform selectivity. Pan-HDAC inhibitors such as vorinostat and panobinostat—although effective—often cause off-target effects due to inhibition of multiple HDAC isoforms. Such non-selectivity can lead to side effects including cytopenias, gastrointestinal disturbances, and even cardiac toxicity. Other limitations include:

• Toxicity concerns: High doses required for efficacy in some solid tumors result in significant toxicity and limit long-term administration.
• Drug resistance: Tumor cells can develop resistance through mechanisms such as upregulation of drug efflux pumps and compensatory signaling pathways that bypass the need for HDAC activity.
• Limited potency in some solid tumors: While many HDAC inhibitors have shown excellent responses in hematologic cancers, their efficacy in solid tumors has been modest.
• Pharmacokinetic challenges: Issues such as bioavailability, tissue penetration (especially in dense fibrotic or immune‑privileged sites), and metabolic instability remain hurdles for clinical translation.

Improving selectivity is a major goal, and strategies include developing HDAC inhibitors with dual or multi-targeting functions (such as combining a kinase inhibitor pharmacophore with an HDAC inhibitory motif) or designing inhibitors that can be delivered selectively to tumor tissue using nanoparticle or antibody‑drug conjugate strategies.

Emerging Research and New Targets

Ongoing research is aimed at addressing these challenges and expanding the repertoire of HDAC-targeting therapies:

• Isoform-selective inhibitors: There is an increasing focus on developing inhibitors that selectively target HDAC isoforms associated with tumor progression, such as HDAC1, HDAC2, or HDAC3. Selective inhibition may confer enhanced efficacy with a reduced side-effect profile. For example, entinostat’s ability to target HDAC1/3 selectively has shown promise in both preclinical and clinical studies.
• Combination therapies: Preclinical findings and early-phase clinical trials are increasingly exploring HDAC inhibitors in combination with other anticancer agents, immunotherapies, and even radiotherapies. This approach aims to provide synergistic effects that overcome resistance mechanisms while allowing lower doses of each agent.
• Dual/multi-target inhibitors: As single-target therapies can be limited by tumor heterogeneity and the complex interplay of signaling pathways, research is being directed at dual inhibitors that combine HDAC inhibitory function with activity against other cancer-relevant targets (for example, kinase inhibitors).
• Nanoparticle-based delivery systems: Innovations in drug delivery such as nanoparticles and liposomal formulations are being investigated to improve the tissue specificity of HDAC inhibitors. Such approaches can increase bioavailability and reduce systemic toxicity.
• Repurposing for non-cancer indications: Beyond oncology, HDAC inhibitors are being studied for neurodegenerative diseases, inflammatory disorders, and even some infectious diseases. This repurposing is driven by the observation that epigenetic dysregulation is a common mechanism in chronic diseases.
• Biomarker-driven patient selection: Research into biomarkers that predict response to HDAC inhibitors is critical. Integration of genomic, proteomic, and epigenomic data may allow clinicians to identify patients who are most likely to benefit from HDAC-targeted therapies, as well as monitor efficacy using quantitative assays developed from blood-based biomarkers.

The current research landscape advocates for a more personalized approach to HDAC inhibition, whereby a tumor’s specific epigenetic signature, expression profile of HDAC isoforms, and concurrent signaling pathway activation determine the choice of inhibitor and combination regimen.

Detailed Conclusion

In summary, therapeutic candidates targeting HDACs span a broad chemical and pharmacological spectrum from first‑generation pan‑HDAC inhibitors to next‑generation isoform‑selective and multi‑target candidates. HDAC inhibitors work by binding to the zinc‑dependent catalytic site of HDAC enzymes, leading to increased histone acetylation and a cascade of downstream effects including cell cycle arrest, apoptosis, anti‑angiogenesis, and immunomodulation. These cellular effects translate into clinical benefits primarily in hematological malignancies (as exemplified by vorinostat, romidepsin, belinostat, panobinostat, and tucidinostat), with ongoing clinical trials aiming to extend their use to solid tumors and non‑oncological indications.

The inhibition of HDACs rewires the transcription program of tumor cells by reactivating silenced tumor suppressor genes and modifying non‑histone protein function. However, non‑selective inhibition can lead to wide‑ranging gene expression changes and result in adverse effects. As a result, one of the major challenges is to develop inhibitors that are more selective for the tumor-specific isoforms while reducing toxicity. Strategies for future research include the development of dual‑targeting compounds, the incorporation of novel drug delivery systems (such as nanoparticle carriers), and the use of biomarkers to guide patient selection and treatment monitoring. Innovations in these areas are expected to overcome the drawbacks of current compounds and further elevate the therapeutic potential of HDAC inhibitors.

From a general perspective, HDAC inhibitors represent a conceptually novel class of therapies that have the potential to be applied across multiple diseases due to their central role in epigenetic regulation. More specifically, in oncology, the fact that epigenetic abnormalities contribute directly to tumorigenesis has made HDAC inhibition an attractive strategy. Yet, the precise and balanced modulation of the acetylome remains challenging. Future directions must focus on achieving high isoform selectivity to maximize efficacy and reduce adverse reactions. Researchers are now turning to advanced medicinal chemistry and delivery techniques to achieve these goals while also studying synergistic combinations with immunotherapy or DNA repair inhibitors for more durable responses.

In conclusion, the therapeutic candidates targeting HDACs have already revolutionized the treatment of certain hematological malignancies and hold great promise for further expansion into multiple therapeutic areas. Continued innovation in the design, delivery, and combination of HDAC inhibitors—complemented by biomarker development and careful clinical trial interpretation—is essential to fully harness the potential of epigenetic therapies. This robust, multi-perspective approach to HDAC inhibition underscores the significant progress made so far and the exciting road ahead in precision medicine and targeted therapeutics.

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