HDAC6 regulates signaling pathways which are involved in tumor cell growth, survival, and invasiveness and are often overexpressed in the majority of malignancies, including CRC. The increased expression of HDAC6 was reported in colon cancer tissue compared to the adjacent noncancerous tissue and is often associated with unfavorable disease prognosis [
7,
59]. The oncogenic potential of HDAC6 has been well established as its inactivation by genetic manipulations reduced oncogenic transformation and tumor growth
in vitro as well as in
in vivo models. More importantly, data obtained on HDAC6 null mice show that HDAC6 is not an essential gene for the development of an adult organism and that physiological functions of normal cells are not affected with deletion of HDAC6 gene [
22,
133]. Furthermore, it has been shown that unlike the other HDACs, selective inhibition of HDAC6 impaired tumor growth and progression without inducing major adverse events in experimental animals [
22,
61,
110,
134]. Altogether, these characteristics make HDAC6 a highly desirable target for cancer treatment. [
135,
136]. In experimental models of CRC, the HDAC6 gene knockdown and pharmacological HDAC6 inhibition reduced cell viability and migration of tumor cells by inhibiting the MAPK/ERK pathway [
59,
137]. It has been reported that patients with CRC show significantly lower expression of SET7 Lys methyltransferase in cancer tissue than in adjacent tissue. SET7 catalyzes the methylation of histone H3K4 that affects chromatin remodeling and regulates genes that are involved in cell cycle regulation, differentiation, DNA damage response and thereby plays a significant role in tumorigenesis. Moreover, SET7 functions as a tumor suppressor by inhibiting deacetylating activity of HDAC6, partially through ERK signaling pathway in colon cancer cells [
137]. Downregulation of SET7 expression was closely correlated with poor prognosis in CRC [
138] which is relevant for the oncogenic potential of HDAC6 in CRC. Furthermore, high expression of HDAC6 in CRC tissue was reported to be associated with reduced levels of acetylation at the 12
th Lys residue of the histone H4 protein (H4K12ac). This histone residue was also reported to be highly sensitive to HDAC6 inhibition in several cancer types that subsequently induced chromatin relaxation [
7].
HDAC inhibitors (HDACi)s can be classified according to their chemical structure and based on their ability to inhibit the specific HDAC isoform or distinct HDAC classes. In this sense, HDAC6 activity can be inhibited by unselective or pan-HDACis that inhibit the majority of HDAC classes, and selective inhibitors that target specifically HDAC6.
Ricolinostat (ACY-1215) is the first effective selective HDAC6i that became orally available, and it was shown to be ten times more selective against HDAC6 than other HDACs [
151,
152]. Since its first application in MM [
151], ACY-1215 has shown some efficacy in various tumors [
153]. By increasing the level of acetylated tubulin, cortactin, and Hsp90, ACY-1215 inhibits cell cycle progression, motility and invasiveness, processing of misfolded proteins, ubiquitin proteasome pathway, and autophagy. Furthermore, it renders GRP78 in acetylated form thus preventing it from transporting misfolded proteins from ER lumen, hence increasing ER stress. This effect, together with increased accumulation of protein aggregates, polyubiquitinated and misfolded proteins, induced tumor cell death by apoptosis [
154]. Furthermore, ACY-1215 exerts its antitumor effect by inhibition of proliferation and induction of apoptosis by inhibiting MAPK/ERK, PI3K/AKT cell signaling pathways in CRC [
59,
155].
3.1. HDAC6 inhibition in combination with other therapeutic modalities
In order to explore the options for the most optimal antitumor activity while minimizing the side effects of the maximally tolerated doses, the selective HDAC6is have been further evaluated in combination with different anticancer agents such as proteasome inhibitors, tyrosine kinase inhibitors, radiotherapy, and immunotherapy.
The afore mentioned ACY-1215 has been reported to act more effectively in combination with the protease inhibitor bortezomid which is currently used in the treatment of MM and Hodgkin and non-Hodgkin lymphoma [
151]. In CRC, ACY1215 showed improved antitumor effect in combination with carfilzomib (proteasome inhibitor) by inducing accumulation of protein aggregates, ER stress, and subsequently apoptosis of treated CRC cell lines harboring BRAFV600E mutation in
in vitro and in xenograft murine models [
165]. Another selective HDAC6 inhibitor C1A also showed potential antitumor activity against KRAS-mutated CRC in murine xenograft models [
150] in combination with bortezomib that was characterized with inhibited degradation of misfolded proteins in proteasomes and decreased autophagy [
166].
BRAFV600E mutations that lead to constitutive activation of BRAF kinase and increased RAS/RAF/MEK/ERK signaling have been reported in 10% of patients with CRC [
167]. However, in CRC harboring BRAFV600E mutations, response rates to BRAF inhibitors vemurafenib or dabrafenib and MEK1/2 inhibition with trametinib were low due to adaptive feedback reactivation of upstream RTKs and RAS. In these tumors, BRAF inhibition enhances RAF dimerization and thus results in adaptive feedback re-activation of MAPK signaling, often mediated by EGFR activation [
168]. HDAC inhibitors have been tested as potential agents to reduce the resistance to BRAF/MEK inhibition in CRC. Accordingly, one study in mouse xenograft CRC model and cell lines showed the ability of vorinostat to overcome resistance to treatment with MEK1/2 inhibitor trametinib. The study also identified a novel resistance mechanism that is mediated via STAT3 and the anti-apoptotic protein c-FLIPL [
169]. Increasing experimental evidence shows enhanced antitumor effects of simultaneous application of HDACis and RAF/MEK/ERK targeting therapeutic agents, in other tumors with BRAF mutation. Synergistic antitumor effect of vorinostat in combination with PLX4720 BRAF inhibitor [
170] and the ability of vorinostat to eliminate BRAF inhibitor resistant and senescencent cells was reported in BRAF mutated melanoma cells [
171]. Similarly, panobinostat in combination with MAPK and BRAF inhibitor dabrafenib has shown synergistic antitumor effect in BRAF mutated thyroid carcinoma cells [
172].
As HDAC inhibition often induces cell cycle arrest and inhibits DNA repair, the ability of HDAC inhibitors to improve the therapeutic outcome of radiotherapy and to sensitize the tumor cells to ionizing radiation has been investigated in experimental settings [
173]. In this sense pan-HDAC inhibitors panobinostat and vorinostat have shown enhanced antitumor effect when used in combination with radiation therapy in clinical trials in prostate and gastrointestinal tract carcinoma [
174]. Furthermore, one study reported that treatment with the selective HDAC6i SP-2-225 resulted in decreased tumor growth and increased infiltration of M1 macrophages within tumors. These finding support further investigation of the use of selective HDAC6is to improve antitumor immune responses and prevent post-radiation therapy tumor relapse [
175].
3.2. Modulation of antitumor immunity with HDAC6 inhibition
HDAC inhibitors have shown considerable immunomodulatory effects by influencing many aspects of the immune response in tumors. However, some differences were reported depending on weather pan-HDAC or selective HDAC6 inhibitors were used and on the experimental model used in a study (
Table 4) [
117].
Pan-HDAC inhibitors have been reported to decrease the expression of costimulatory molecules (CD40, CD80, CD83 and CD86) on DCs and secretion of Th1 polarizing cytokines (IL-6, IL-12, TNF) after stimulation of TLR on DCs that led to inhibition of T cell activity. Panobinostat reduced the expression of costimulatory molecules on DCs and impaired IFN-γ production by T cells [
176]. Similarly, pan-HDACi vorinostat was reported to inhibit T cell functions by inducing the transcription of enzyme indoleamine 2,3-dioxygenase (IDO) in DCs which regulates the catabolism of tryptophan which is essential for T cell activation [
177]. HDAC inhibition by vorinostat was shown to promote the transcription of IDO through acetylation and activation of STAT3 and hence to inhibit T cell functions [
178]. Contrary to pan-HDACis, selective HDAC6 inhibitors have shown immunostimulatory effects on DCs. It has been reported that ACY241 increased the expression of costimulatory and MHC molecules on DCs [
179]. Furthermore, it was shown that tubastatin A impairs the production of immunosuppressive cytokine IL-10 by DCs and macrophages by disrupting the complex between HDAC6 and STAT3 and by impairing STAT3 signaling that in turn increased the production of IFN-γ by CD4 T cells [
118,
163]. All these factors indicate that HDACis by affecting APCs may regulate innate and adaptive immune response and inflammation in TME. Moreover, tubastatin A showed antiinflammatory effect by inhibiting IL-6 synthesis, nitric oxide (NO) secretion, cell viability and motility in human macrophages stimulated with LPS [
180]. This effect is also relevant for antitumor effect of tubastatin A, as IL-6 represents a potent EMT triggering factor in the TME involved in tumor progression, metastatic invasion, and chemoresistance [
99].
The effects of pan-HDACis on T cell activation differ from the effects of specific HDAC6is. Pan-HDACis trichostatin A and rodempsin inhibit activation-induced proliferation of naïve T cells and IL-2 production [
181], unfavorably affect metabolic reprograming of recently activated T cells, impair T cell receptor signaling, and induce T cell apoptosis [
117,
182,
183]. However, reports on the impact of pan-HDACis on previously activated effector T cells show that when applied after initial activation of CD4 T cells, pan-HDACi trichostatin A prevents FasL-driven activation-induced cell death, increases infiltration of CD4 T cells into tumor and reduces tumor growth in lymphoma and melanoma murine models [
117,
184]. Regarding selective HDAC6is, Laino et al. found that peripheral blood T cells of melanoma patients
in vitro treated with selective HDAC6is ACY-1215 and ACY-241 showed decreased production of immunosuppressive Th2 cytokines (IL-4, IL-5, IL-6, IL-10 and IL-13) with concomitant downregulation of the Th2 transcription factor GATA3, upregulation of the Th1 transcription factor T-BET, and favorized accumulation of central memory phenotype T cells. This report indicated the immunostimulating potential of selective HDAC6is on T cell cells [
185].
CD8 T cells directly kill tumor cells by secretion of perforin and granzymes and facilitating antitumor immune responses by production of IFN-γ and TNF that activate local APCs and increase immunogenicity of tumor cells by inducing MHC expression, and activation of immunoproteasomes. It was reported that treatment with pan-HDACi vorinostat increased the proliferation and function of CD8 T cells, particularly the frequency of IFN-γ or perforin-producing T cells in mammary tumor bearing mice [
186]. Contrary to pan-HDAC inhibitor vorinostat, selective inhibition of HDAC6 may impair the cytotoxic capacity of CD8 T cells, as tumor-specific CD8 T cells from mice treated with HDAC6-specific inhibitor tubastatin A and the HDAC6 deficient mice showed reduced lytic capacity of CD8 T cells, probably due to interrupted intracellular trafficking and exocytosis of perforin [
125]. However, one study reported that another HDAC6 selective inhibitor, ACY241 increased perforin and IFN-γ production in CD8 T cells [
179]. Evidently, due to the conflicting evidence on the effect of different selective HDAC6is on T cell activity, further investigations are needed [
187].
Treatment with selective HDAC6is has been shown to affect the susceptibility of tumor cells to T and NK cell mediated killing, as they affect the expression of MHC class I molecules on tumor cells. In this sense, HADC6 inhibitor tubastatin A has been reported to induce expression of MHC class I molecules on melanoma cells and increase their susceptibility to CD8 T cell mediated-lysis [
132]. Moreover, the treatment of colon cancer cells with pan-HDACi vorinostat increased the expression of death receptor Fas that led to enhanced Fas-dependent cytotoxicity activity of T cells [
149]. Furthermore, it has been reported that treatment with pan-HDACi panobinostat increased the expression of genes involved in cell adhesion and junction and formation of conjugates between NK and tumor cells, and modulated the expression of NK cell-activating receptors and ligands on tumor cells, thus contributing to the increased cytolysis of tumor [
188]. Regarding NK cell antitumor activity, it was reported that pan-HDACis vorinostat and trichostatin A induced the expression of MHC class I-related chain A (MICA) and B ligands for activating NK cell receptor NKG2D in hepatocellular carcinoma and Ewing sarcoma and thereby increased the susceptibility of treated tumor cells to NK cell-mediated lysis [
189,
190]. Furthermore, it was reported that HDAC6i Nex A increased tumor infiltration with NK cells, an effect that has been associated with improved prognosis and survival in tumors [
121,
187].
Moreover, some HDACis enhance T cell migration to tumor site. Pan-HDACi rodempsin was reported to increase the expression of chemokines CCL5, CXCL9, 10 by tumor and stromal cells, increase tumor infiltration with T cells and thereby improve antitumor immune response [
191].
Selective HDAC6 inhibition contributes to antitumor immunity by inhibiting differentiation and influx of suppressive immune cells into TME. Ricolinostat was reported to inhibit the activity of myeloid-derived suppressor cells (MDSC) and inhibit tumor growth [
192] while Nex A treatment has been shown to induce differentiation of immunostimulating M1 macrophages [
121] similarly to the pan-HDACi trichostatin A [
193].
In summary, the HDAC6is have shown potential inherent to immunomodulatory agents as they have the ability to improve antitumor immunity by stimulating the immunogenicity of tumors and activity of immune cells.
The expression of immune checkpoint (IC) molecules is often upregulated in immune cells in TME as a consequence of antitumor immune responses and due to elevated levels of immunosuppressive mediators produced by tumor cells, suppressive immune cells, and stromal cells [
194]. In this sense in TME, T and NK cells may express programed cell death receptor (PD)-1, cytotoxic T lymphocyte antigen (CTL)-4, T cell immunoglobulin and mucin domain-containing protein 3 (TIM3), lymphocyte activation gene-3 (LAG-3), and TIGIT, that inhibit antitumor immune responses [
104,
195]. On the other hand, tumor cells often express ligands for ICs that can be induced with oncogenic pathways and extrinsic factors in TME such as cytokines IFN-γ, IL-6, TNF-α [
196,
197] that contribute to upregulation of PD-1 ligands (L)1 (B7H1), and L2 (PD-L2, B7DC) on tumor cells. In CRC, MAPK, PI3K, JAK/STAT3, and phospholipase Cγ signaling pathways have been related to the upregulation of PD-L1 expression [
198].
It has been well established that metastases of CRC show increased expression of PD-L1 compared to primary tumors [
199] that is associated with unfavorable disease prognosis [
200]. Moreover, the expression of PD-L2 in tumor cells, which is more inherent to immune cells, was recently associated with poor patient survival in CRC [
200,
201]. Therapeutic blockade of PD-1 and its ligand with anti-PD-1 and anti-PD-L1 antibodies has shown a considerable clinical benefit in some metastatic CRC [
199,
202]. However, blockade of PD-1 enhances T cell function and the subsequent production of inflammatory cytokines, most notably IFN-γ that enhances PD-L1 and PD-L2 expression on tumor cells thereby inducing negative feedback as well as other immunosuppressive pathways [
194,
196,
203].
As tumors during their evolution develop mechanisms allowing them to evade immune responses, it is of importance to identify treatments that can increase immunogenicity, minimize immune-related adverse events, and maximize therapeutic benefits of IC inhibition [
104]. Among the emerging new therapeutic targets HDACs have raised great interest, especially HDAC6 since it is involved in the control of immunomodulatory pathways and expression of IC molecules. It has been shown recently that HDAC6 may induce the expression of PD-L1 in cancer cells via activation and recruitment of STAT3 transcription factor, as it was shown in the experimental models of pharmacological impairment of HDAC6 or by its genetic abrogation in melanoma [
204] and breast cancer [
163,
205]. However, there are differences in existing published data between the effects of some selective pharmacological HDAC6 inhibitors on PD-L1 expression. It was reported that
in vitro treatment with Nex A decreased the expression of PD-L1 in melanoma [
121] and breast [
163] animal tumor models. Similar effect was reported for the
in vitro treatment with the novel selective HDAC6i MPTOG612 in CRC cells [
164]. However, some publications report the opposing effect for certain selective HDAC6is. One study reported that the treatment with novel small molecule HDAC6i A452 and ACY-1215 increased PD-L1 expression in CRC tumor cells [
157]. Another study reported that ACY-1215 alone and in combination with alkylating agents upregulated PD-L1 in CRC cells
in vitro and that was achieved irrespective of p-STAT3 status [
155]. However, ACY-1215 and A452 induced PD-L1 expression may increase susceptibility of tumor cells to PD-1/PD-L1 axis IC blockade therapy.
The use of nonspecific HDACi, such as panobinostat [
206] as well as low doses of trichostatin A [
193] was reported to increase the expression of PD-L1 and PD-L2 on cell surface of tumor cells in murine models of melanoma and breast cancer. Moreover, panobinostat was shown to synergize with PD-L1 blockade by different mechanisms such as promotion of NK cell-target cell conjugation formation by increasing the expression of cell adhesion and tight junction-related genes and by increasing the expression of CD80, CD86 (ligands for CD28), and CD112 (PVRL2/nectin-2 ligand for activating DNAM-1 NK cell receptor) on tumor cells [
188]. These results also indicate the antitumor potential of HDAC inhibition in the context of NK cell-based immunotherapy. It was also reported that panobinostat augmented the expression of MHC I and costimulatory molecules (CD40, CD80) on melanoma cells
in vitro that led to increased activation of antigen-specific T cells [
207].
Aside from the immune cells, PD-1 is also expressed on malignant cells including colon cancer [
208]. Moreover, tumor cells expressing PD-1 exhibit higher ability of proliferation and tumorigenicity [
209]. Recently it has been reported that the transcription of PD-1 gene in cancer cells is regulated via acetylation of p53 tumor suppressor by HATs p300, CBP and Tip60 in a manner that acetylated p53 recruits the acetyltransferase cofactors to interact with PD-1 promotor and induce the expression of PD-1 [
210]. Although the validation on larger sample size is needed, the expression of PD-1 on tumor cells versus its expression on immune cells may have some relevance for IC blockade selection in patients with CRC [
208]. However, there are scarce studies on the effect of HDACis on PD-1 compared to PD-L1 in tumor cells [
208].
Selective HDAC6 inhibition also affects IC expression on immune cells. In this sense, one study reported that ACY-1215 and ACY-241 decreased the expression of LAG-3, TIM-3, and PD-1 on peripheral blood T cells of melanoma patients. These findings indicate that inhibition of HDAC6 activity may be effective in alleviating T cell suppression and enhancement of the cytotoxic function of T cells and may provide theoretical basis for further evaluation of potential clinical efficacy of joint HDAC6 and IC inhibition [
185]. These effects of HDAC6 inhibition may be relevant for CRC cells as TIM-3 [
211], along with PDL-1/2 [
186,
200], has been identified on tumor cells and has been designated as a negative prognostic biomarker in colon cancer.
3.3. Combination therapies with HDAC6 and IC inhibitors
Although the IC blockade therapies targeting PD-1 and CTLA-4 have shown considerable therapeutic benefit, they have proved ineffective for some patients presumably due to the development of resistance to therapy, immunosuppressive nature of TME, and lack of antitumor T cell response prior to therapy. As selective HDAC6 inhibition has shown in preclinical settings the ability to alter the expression of PD-L1 and PD-L2 on tumor cells, increase immunogenicity of tumor cells, induce more effective antigen presentation, relieve T cell suppression by downregulating the expression of ICs (PD-1, TIM-3, LAG-3) on immune cells [
121,
184], and reverse TME by increasing infiltration of immunostimulating and inhibiting differentiation of suppressive immune cells, the combined application of HDAC6i and immunotherapy has recently emerged as significant approach in the field of cancer treatment [
212]. Therefore, the use of HDAC inhibitors to augment antitumor T cell responses following the application of IC inhibitors may increase the number of patients that respond to IC blockade therapy.
Experimental data obtained on HDAC6 inhibition in combination with immunotherapeutic agents in the settings of tumor cell lines and animal models have shown improved antitumor effect of this drug combination compared to each agent alone. The examples that support this are investigations of ACY1215 in combination with anti-PD-L1 therapy in ovarian carcinoma [
213], ACY-241 in combination with anti-PD-L1 antibody in MM [
214], Nex A in combination with anti-PD-1 antibodies in melanoma [
121], etc. Based on the similar and growing experimental data a considerable number of clinical studies were conducted in advanced solid tumors. The synergistic effect of pan-HDACi vorinostat with anti-PD-1 agent pembrolizumab was shown in metastatic non-small cell lung cancer (NSCLC) patients with good tolerance of both agents in investigated patients [
215]. Another phase 2 clinical trial showed that combined application of vorinostat with pembrolizumab and ER antagonist tamoxifen in ER-positive breast cancer patients resulted in prolonged progression free survival of treated patients [
216]. Phase 2 trial involving combined application of pembrolizumab and vorinostat in recurrent metastatic head and neck showed significant efficacy although with a relatively higher grade of toxicity compared with anti-PD-1 monotherapy [
217]. Regarding the studies on the selective HDAC6i in solid tumors, some therapeutic benefit was shown in patients with previously treated advanced NSCLC treated with ACY-241 in combination with PD-1 blocking drug nivolumab [
218].
Regarding CRC, in patients with microsatellite stable (MSS) CRC that do not respond to IC inhibitors, anti-tumor activity of HDACi CXD101 and nivolumab (anti-PD-1) was assessed in phase II clinical and showed good tolerance and efficacy in the treatment of advanced or metastatic microsatellite-stable CRC [
219]. Furthermore, there is an ongoing phase I clinical study on joint application of pan-HDACi romidepsin in combination with anti-PD-1 agent pembrolizumab in mismatch repair proficient CRC [
220].
Therefore, there is a need for broadening of the research on the role of HDACi and IC inhibitors in antitumor treatment to better evaluate their therapeutic potential and broaden synergistic application in a wider range of tumors.
3.4. Novel HDAC6 based therapeutical approaches
The use of drug combinations can provide efficacy by targeting different signaling pathways and may reverse drug resistance. Although preclinical studies showed that HDACis in combination with other anticancer agents have a better antitumor effect, results obtained in clinical trials have not been in accordance with this. Simultaneous disruption of different signaling pathways and processes aims to reduce tumor cell proliferation and induce tumor cell death but unfortunately some interactions between simultaneously applied drugs cannot be predicted. Desirable additive or synergistic effects of drug combinations may thus impose a risk of inducing adverse effects due to unwanted drug to drug interactions [
221,
222].
Given the advantages and disadvantages of combinational therapies, a novel approach in pharmacology is the design of a drug that can interact with two different targets that are common in a certain disease. Therefore, new treatments have been developed by conjugating two distinct therapeutic compounds in a single molecule for dual- targeting strategies. This may provide increased efficacy of the drug by targeting additional disease-related pathways. In this sense, dual HDAC and kinase inhibitors have been tested in preclinical and clinical settings [
222,
223]. At preclinical level the research on HDACis has evolved and compounds that HDACs interact with receptor tyrosine kinases including PI3K, Src, CDKs, JAKs as well as HDAC have been developed [
221,
223] Class II HDAC6 selective inhibitors that simultaneously target JAK via conjugation of JAK2 selective inhibitor pacritinib with SAHA, has shown improved selectivity for JAK2 [
224].
As in CRC harboring BRAF-V600E mutations, the response rate to BRAF inhibitors is low due to the emergence of resistance. With the aim to effectively target BRAF mutated CRC, novel series of hydroxamate acid and 2-aminopyridinyl-containing compounds such as BRAF and HDAC dual-targeted inhibitors were synthetized. The compounds exerted enzymatic inhibitory activities against BRAFV600E, HDAC1/6 and suppressed the proliferation of CRC cells harboring both wild-type BRAF and mutated BRAFV600E [
225]. Therefore, in theory, the optimal pathway blockade can be achieved by simultaneously targeting multiple steps of the pathway.
Other combinations in development include dual inhibitors that contain DNA targeting agents such as DNA-alkylating agents temozolomide [
221]. Furthermore, dual HDAC6 inhibitor that contained Hsp90 inhibitor, the compound identified as compound 12 (dual HDAC6 Hsp90) displayed inhibitory effects towards the HDAC6 isoform and 246-fold higher selectivity for HDAC6 over HDAC 1, 3 and 8 isoforms and was endowed with significant cytotoxic effects against CRC cell lines [
159].
As was stated before, multi-target drug design approach is aiming to enhance drug activity, selectivity, and overcome drug resistance. Recently, the proteolysis targeting chimera (PROTAC) has become a revolutionary technology in modern drug discovery. PROTACs are bifunctional small molecules consisting of an E3 ubiquitin ligase recognition motif and a ligand for the target protein of interest connected by a suitable linker. PROTACS regulate the expression of the target protein of interest at the posttranslational level by inducing its degradation in proteasomes. The advantages of this catalytic mode of action are enhanced selectivity and improved potency. Moreover, this mode of action results in acute post-translational depletion of pathological protein (target) and eliminates the risk of therapeutic resistance due to physiological feedback mechanisms that upregulate the target protein. In the field of HDAC targeting PROTACs, HDAC6 was designated as the promising target. Several examples in the literature have demonstrated that HDAC6 could be selectively degraded by converting either HDAC6-specific or even pan-HDAC inhibitors into PROTACs. The first HDAC6 degrader was generated based on nonselective HDAC inhibitor and pomalidomide as a ligand for the E3 ubiquitin ligase cereblon (CRBN). CRBN ligands rely on the structure of the anticancer drug thalidomide and its derivatives and have proved to be efficient and selective degraders of HDAC6 [
226]. This may be due to cellular localization of HDAC6 or to formation of more efficient ternary complex [
221,
226,
227]. Until recently, most research on HDAC PROTACs was focused on hematological malignancies that are more sensitive to HDAC6 degraders regarding degradation of HDAC6 [
227] although some encouraging results were observed in tumor cell lines derived from solid tumors [
228]. However, since this research area has very recently emerged it is too early to draw any conclusions.