Open Access Article
Elisa Tassinari and
Andrea Milelli
*
Department for Life Quality Studies, Alma Mater Studiorum - University of Bologna, Corso d'Augusto 237, 47921 Rimini, Italy. E-mail: andrea.milelli3@unibo.it
First published on 30th September 2025
Despite recent advances, Alzheimer's disease (AD) remains largely a mystery more than a century after its discovery. Protein kinases are among the new targets under investigation, which is not surprising given their crucial role in maintaining cellular homeostasis and in the development of various diseases. Several protein kinase inhibitors have shown remarkable therapeutic efficacy in the context of AD, although none of them have yet received approval by regulatory agencies. Alongside the use of classic inhibitors, a new therapeutic approach has emerged in recent years, shifting the focus from modulation to targeted degradation of the protein. The purpose of this review is to highlight and discuss novel series of proteolysis-targeting chimeras (PROTACs) directed against protein kinases relevant to the development of AD.
In addition, despite decades of substantial investment by for-profit and non-profit organizations, AD is still an unsolved question and only a few pieces of its pathogenesis are clear today.4 Indeed, an objective understanding of its emergence and development is still far from being achieved, and many theories have been proposed in recent years by eminent scientists to explain what remains, for all intents and purposes, an enigma.5 Due to the many theories developed, countless possible drug targets have emerged and have been pursued in numerous drug discovery programs accompanied, unfortunately, by dramatic high-level failures.6 Up to 2021 only four molecules were used in clinical practice, three acetylcholinesterase inhibitors and one NMDA antagonist. Nevertheless, none of these drugs decrease neuronal loss or reverse cognitive impairments or act as a truly disease-modifying drug.7 Among the various theories proposed, a central focus in the agendas of many research groups is the β-amyloid (Aβ) protein, which represents one of the two hallmarks of the disease, together with neurofibrillary tangles (NFT) consisting of hyperphosphorylated tau protein.8 Over the years the highest efforts have been directed towards the Aβ protein and to reduce Aβ-induced toxicity. Recently, these efforts have resulted in three Aβ-directed monoclonal antibodies that have received FDA approval: aducanumab approved in 2021, albeit with furious controversy,9 and discontinued at the beginning of 2024,10 lecanemab11 and donanemab.12 Unfortunately, along with justifiable public enthusiasm, there is an animated debate in the scientific community about the efficacy, safety, and costs of these agents.13 Indeed, these antibodies, although inducing a substantial Aβ reduction in AD patients, produce only a modest cognitive and clinical benefit compared to placebo.14,15 In addition, numerous adverse effects were observed in the lecanemab and donanemab trials. In particular, a significant increase in cases of symptomatic amyloid-related imaging abnormalities was found during phase 3 studies.16
Given this, the drug discovery efforts to uncover effective disease-modifying agents for the treatment of AD are highly dynamic. According to Cummings et al.'s annual analysis of the AD pipeline, currently there are 182 trials and 138 novel drugs across the 2025 AD pipeline.17 A closer look reveals that these drugs are directed towards a wide array of targets, the most common include modulators of neurotransmitter receptors, inflammation, amyloid-induced toxicity, and synaptic plasticity. Interestingly, there is an increasing number of protein kinase inhibitors in the AD pipeline,18 which are mainly known as drugs at the forefront of the anticancer therapy.19 Indeed, masitinib and nilotinib BE are in clinical phase 3, while baricitinib and MW150 are in clinical phase 2.17 However, the appearance of kinase inhibitors in the AD pipeline is not surprising considering that these proteins are responsible for the intracellular signaling and, therefore, they regulate multiple downstream effects. Several kinases play a key role in the development and progression of AD.20 For instance, NFTs result from an overactivation of protein kinases, such as glycogen synthase kinase 3β (GSK-3β), cyclin-dependent kinase 5 (CDK5), extracellular signal-regulated kinase 2 (ERK2), microtubule-affinity regulating kinase (MARK), protein kinase A (PKA), death-associated protein kinase 1 (DAPK1), Rho-associated protein 1 (ROCK1), and Fyn, which cause hyperphosphorylation of the tau protein and its subsequent aggregation into NFTs.21 Besides this crucial role, kinases are also involved in other processes such as Aβ aggregation, neuroinflammation, synaptic plasticity, memory formation and overactivation of astrocytes and microglia.20 Therefore, it is not surprising that many drug discovery programs are aimed at obtaining protein kinase inhibitors as disease-modifying agents for the treatment of AD. However, the use of kinase inhibitors for the treatment of AD is still in its infancy and, although some drugs have been or are still in the clinical study phase, there are no inhibitors that have received regulatory agency approval. Unfortunately, the development of such molecules is by no means straightforward, due to a variety of reasons, including the inherent difficulty of specifically targeting protein kinases. Off-target side effects due to poor selectivity are the main problem associated with the development of a kinase inhibitor since there are approximately 518 kinases in the human kinome. Furthermore, achieving effective therapeutic concentration in the brain is a major challenge for CNS-directed kinase inhibitors since, due to a combination of physicochemical and pharmacokinetic factors, they do not cross the blood brain barrier (BBB). Indeed, molecular weight (MW), lipophilicity (log
P) and polar surface area (PSA) influence the BBB penetration ability of a molecule. An analysis by Chico et al. reported that kinase inhibitor drugs tend to have higher mean values for these parameters, when compared to other known CNS-penetrating compounds.22 For instance, imatinib showed limited efficacy in glioblastoma clinical trial since it did not reach therapeutic levels in the brain.23 Lastly, since kinase networks are highly redundant and interconnected, their inhibition causes compensatory changes, thus, reducing effectiveness and causing adverse effects.
Recently, the drug design process has been disrupted by the appearance of a new strategy for modulating the activity of proteins, known as targeted protein degradation (TPD).24 In particular, the use of proteolysis targeting chimeras (PROTACs) may offer several advantages over traditional kinase inhibitors in modulating kinase activity in AD.25
In the current review, to avoid unnecessary overlap with the prolific recent literature, in the following paragraph, we will briefly discuss PROTAC characteristics. Following this, we will examine the role that different kinases play in the pathogenesis of AD and highlight selected case studies of kinase-targeted PROTACs in the context of AD.
Further, LRRK2 has been the target of a successful medicinal chemistry campaign that led to the discovery of XL01126, a promising PROTAC exhibiting remarkable oral bioavailability and BBB penetration.33
Although no degraders for AD have entered the clinical trials yet, the development of protein degraders is extremely dynamic, as shown in Fig. 1a.34
![]() | ||
| Fig. 1 a) Number of articles per year featuring “PROTACs” and “PROTAC” and “Alzheimer's disease” in the title, abstract, or keywords (Scopus search, June 2025); b) mechanism of action of PROTACs. | ||
Although the MoA of PROTACs is still being investigated, it is widely accepted that PROTACs initiate the degradation cascade by recruiting POI and forming a ternary complex with E3 ligase. The induced proximity between POI and E3 ubiquitin ligase causes ectopic ubiquitination of the POI, which is then degraded via the ubiquitin-proteasome system (UPS).
One of the main challenges in producing a successful kinase inhibitor lies in the identification of a potent and selective molecule able to modulate a given target. This is even more complex when dealing with protein lacking a specific binding site. Moreover, when considering classic inhibitors, it is required to maintain a suitable drug concentration for a specific amount of time. Due to their peculiar mechanism of action, PROTACs offer numerous advantages over classical small molecule inhibitors, especially regarding the treatment of brain-related diseases. Target occupancy is a crucial factor when considering classic inhibitors. To achieve sufficient level of target-occupancy, high drug concentrations are required, which are frequently associated to off-target effect(s). On the other hand, protein degradation represents an event-driven pharmacology, where a low level of target engagement may be sufficient to induce the substantial degradation of target protein.35 Indeed, it is possible to observe EC50 values substantially lower compare to Kd.36 Moreover, PROTACs, and related protein degraders, are characterized by a catalytic mode of action, that allows protein levels to decrease more significantly over time. Consequently, complete removal of the protein from the cell is achieved, resulting in a more long-lasting effect compared to classic inhibitors.
Furthermore, classic inhibitors block enzymatic functions by binding to the catalytic domain. However, non-enzymatic functions, that play roles in regulating several disease-associated cellular pathways, are non-affected by traditional inhibitors. PROTACs, and related protein degraders, eliminate all the functions of a given protein, both enzymatic and non-enzymatic, leading to improved potency and, possibly, new mechanisms of pharmacological activities.37
PROTACs are less susceptible to resistance from point mutations in the target protein, a common event when dealing with kinase modulation.30 Nevertheless, also with TPDs resistance mechanisms have been observed, such as: a) alterations in E3 ligase pathways, i.e. mutation or downregulation of E3 ligases, disrupting the formation of the ternary complex needed for protein degradation;38 b) deubiquitinase enzyme overexpression, causing insufficient levels of ubiquitinated POI;39 c) upregulation of drug efflux pumps, i.e. MDR1 pump and ABCC1/MRP1.40,41
From a medicinal chemistry's point of view PROTACs are relatively simple heterobifunctional molecules comprising two different warheads, one for POI recruitment and the other one for binding to a specific E3 ligase. The two heads are connected by a linker, that is critical for the correct formation of a productive ternary complex42,43 and for modulating solubility and cell penetration.44 There are numerous factors to take into consideration when designing a successful PROTAC, such as selecting the right POI recruiter, the correct linker as well as the suitable ligase. The discussion of these aspects is beyond the scope of this mini-review therefore, for a more detailed discussion of these aspects, the reader is referred to these excellent papers.27,28,45–47
In recent years, alternative approaches to modulate GSK-3β have been very successful, including development of GSK-3β-directed multi-target drugs.61 Alongside, some GSK-3β-directed PROTACs have been discovered showing extremely exciting anti-AD properties (Fig. 2).62–65
![]() | ||
| Fig. 2 Design of GSK-3β-directed PROTACs. GSK-3β recruiting elements are highlighted in light green, while ligase-recruiting elements are highlighted in light orange. | ||
In 2021, Jiang et al. reported the design, synthesis, and biological evaluation of a series of PROTACs based on thalidomide, as cereblon (CRBN) ligand, and the pyridinethiazole-based inhibitor 1,66 which is a highly potent and selective ATP competitive GSK-3β inhibitor.62 Among the different analogs, a promising GSK-3β degrader, PG21, was obtained following an extensive campaign of linker optimization (Fig. 2). PG21, characterized by an octamethylene linker, showed dose-dependent GSK-3β-degradation, resulting in a 44.2% reduction in the enzyme levels at 2.8 μM, relying on the ubiquitin–proteasome system (UPS) mechanism. Interestingly, other PEG-linked analogs exhibited no significant activity. PG21 exhibited good selectivity for GSK-3α/β enzymes among 18 kinases, as well as anti-inflammatory effects in mouse BV2 microglial cells treated with Lipopolysaccharide (LPS). It significantly reduced the expression of the pro-inflammatory cytokine TNF-α, which is related to several human diseases, including AD.67 In addition, PG21 prevented glutamate-induced cytotoxicity in HT-22 hippocampal neuronal cells, suggesting a neuroprotective role that preserves cellular morphology and neuronal integrity.
The same year, Wang and co-workers discovered PT-65, a potent GSK-3β degrader, which exhibited DC50 values of 28.3 nM and 34.2 nM against GSK-3α and GSK-3β in SH-SY5Y cells, respectively (Fig. 2).63 AZD2858, a potent and selective competitive ATP inhibitor,68 was selected as GSK-3β warhead while pomalidomide was chosen as CRBN ligand. PT-65 shows a high affinity for GSK-3β (KD = 12.41 nM), with only about a twofold decrease compared to AZD2858, indicating its notable degradation potency. Further evaluations were conducted to explore the mechanism of degradation of PT-65. Pre-treatments of SH-SY5Y cells with pomalidomide and MG-132, a known proteasome inhibitor, markedly attenuated PT-65-induced GSK-3 degradation, suggesting an UPS-mediated degradation. Moreover, the treatment with PT-65 in cells containing high levels of GSK-3β and p-Tau showed a reduction in GSK-3β and p-Tau levels in a dose-dependent manner, without altering the total level of Tau. The same treatment was conducted using AZD2858, which inhibited the enzymatic activity of GSK-3β, leading to a dose-dependent decrease in p-Tau. After removing the compounds through washing out, GSK-3β activity and p-Tau levels rapidly returned to normal in cells treated with AZD2858. On the contrary, GSK-3β took longer to recover in cells treated with PT-65. These findings underline the potential therapeutic advantages of PT-65 as a protein degrader in the treatment of AD, due to its longer duration of efficacy and, consequently, a potential reduced number of administrations. Furthermore, PT-65 reduced Aβ-induced tau hyperphosphorylation and neurotoxicity in SH-SY5Y cells, a common cellular model for AD, increasing cell viability in a dose-dependent manner with no observed toxicity up to 100 μM. Moreover, PT-65 can improve learning and memory impairment on AD model rats induced by administration of okadaic acid (OA). Indeed, the compound showed a reduction in the escape latency in the Morris water maze (MWM) test, indicating cognitive improvement, and a decreased p-Tau and GSK-3α/β expression in the hippocampus.
In 2023, our research group reported a small set of novel GSK-3β degraders based on PROTAC technology. This involved linking two different GSK-3β inhibitors, SB-216763 and tideglusib, which weren't previously explored in degraders, to pomalidomide, selecting different-length PEG linkers.64 The study also aimed to evaluate potential differences in ATP- and non-ATP-competitive GSK-3β recruitment. Indeed, SB-216763 is a selective reversible maleimide-based ATP competitive inhibitor of GSK-3β while tideglusib is a selective GSK-3β inhibitor that acts via a non-ATP competitive mechanism.69 While derivatives of tideglusib, such as compound 4, did not exhibit appreciable degradation of the POI, the SB-216763 derived compounds did show significant decrease in the GSK-3β level. However, the lack of activity is unlikely to be related to the allosteric binding: a more plausible explanation lies in the linker design, which plays a critical role in ternary complex formation.43 Compound 3, characterized by the SB-216763 warhead and the 3–4–3 PEG linker, emerged as the most potent degrader of the set inducing GSK-3β degradation, due to the formation of a more stable ternary complex (Fig. 2). This was demonstrated also by the docking analysis, which showed that a longer linker allowed the pomalidomide to reach the β-hairpin loop of the kinase, encouraging the binding with CRBN. The inhibitory profile of compound 3, evaluated through Kinase-Glo luminescent assay, seemed sufficient to engage GSK-3β. PROTAC 3 exhibited an IC50 of 833 nM, a value that is more than one order of magnitude higher compared to the starting inhibitor itself (SB-216763 IC50 = 34 nM). However, one of the critical differences between PROTACs and classic inhibitors is that for PROTACs a moderate binding to the POI is sufficient to trigger the degradation process. PROTAC 3 showed no neurotoxicity in SH-SY5Y cell lines up to 20 μM and induced significant degradation starting from 0.5 μM in a dose dependent manner. As in previous studies, the involvement of the UPS system in the degradation of GSK-3β was also demonstrated by using lactacystin, a specific proteosome inhibitor. PROTAC 3 significantly reduced the neurotoxicity induced by Aβ25–35 peptide and CuSO4 in SH-SY5Y cells in a dose-dependent manner. Since the ability to cross the blood–brain barrier (BBB) is notably important for AD-drugs, PROTAC 3 was evaluated through PAMPA BBB assay, exhibiting an effective permeability (Pe) of about 15.33. This classifies PROTAC 3 as CNS± permeable, approaching CNS+, i.e. it exhibits a moderate ability to cross the BBB, with a value not far from the optimal one for crossing the BBB.
It is well known that PROTACs physicochemical properties remain a significant challenge, especially in the CNS PROTAC landscape.70 In this context, Holmqvist et al. demonstrated that the use of orthogonally reactive linkers enables the identification of CNS in vivo active degraders of GSK-3β from a single screen (Fig. 3).65 The orthogonally reactive linkers method leads to a rapid and selective combination of the POI ligand and the E3 ligase binder, based on the fact that during the assembly one end of the linker reacts exclusively with the POI binder and the other end only with the E3 ligase recruiter, without any disruption. Therefore, the orthogonally reactive linker binds selectively and independently the GSK-3β ligand and the E3 ligase recruiter, avoiding cross-reactivity and/or unwanted side reactions that can occur with standard linkers. This approach enables the synthesis of multiple combinations, resulting in a large library of compounds and substantially increasing the probability of identifying molecules with good degradation activity and suitable physicochemical properties. In this context, the authors selected 12 non-excessive polar and non-long orthogonal reactive linkers presenting an alkyne group and a secondary amine: first the secondary amine undergoes an SN2 with a bromine-containing E3-ligase recruiter and, second, the alkyne reacts with an azido-containing POI binder via click chemistry (Fig. 3). As GSK-3β-recruiters, the authors selected two different binders, PT-367 and CMP-47,71 both modified by the addition of an azido group. The CRBN-recruiting fragments were selected based on minimal molecular weight and high hydrolytic stability. The synthesis was performed in a 96-well plate, starting with the SN2 reaction followed by the click reaction. A different plate was used for the two selected GSK-3β ligands, with the seven different E3 binders distributed in rows and the twelve linkers in columns. The plates were purified using a 96-well strong cation exchange (SCX) plate, facilitated by the presence of the basic center in the linkers, which encourages the separation of non-reacted species and by-products from the desired PROTACs. 103 of the 147 compounds obtained exhibited >50% of purity when detected in LC-MS. The compounds were directly screened through a direct-to biology strategy72 to evaluate the ability of crude compounds to degrade GSK-3β in GSK-3β-HiBiT knock-in HEK293 cell lines after 24 h treatment. Six compounds emerged, exhibiting a good degradation profile and were subsequently re-synthesized and purified via preparative HPLC to compare the degradation efficacy. The DC50 values left shifted from 2.5 to 20 folds from the crude to the purified compound, confirming that all DC50 values were lower than 20 nM. In particular, compound 5 was selected for further evaluation (Fig. 3). It degrades GSK-3β and GSK-3α after a 2 hour-treatment with a 10 nM concentration, and its selectivity to GSK-3 paralogs was confirmed by tandem-mass-tagging. Furthermore, they investigated the effects of GSK-3 degradation on the level of β-catenin and on collapsin response mediator protein 2 levels (CRMP2), which are hyperphosphorylated in the brain of AD patients. Compound 5, after a 24 hour-treatment in SH-SY5Y cells, induced no phosphorylation in β-catenin up to 100 nM, whereas p-CRMP2 levels were decreased at already 0.1 nM. Moreover, in mice 5 mg kg−1 i.v. of compound 5 leads to a complete GSK-3β elimination in liver and a partial remotion in mouse brain within 4 hours, attributing to 5 good potentialities and confirming the reliability of the use of orthogonally reactive linkers and a direct-to biology approach to accelerate new SNC PROTACs discovery.
In 2023, Son et al. reported a direct selective degradation of phosphorylated-p38 (p-p38), the p38 active form, by exploiting the glycine flip-intrinsic conformation of the activated p-p38.85 According to this, a series of PROTACs have been designed by selecting the previously reported p38 benzophenone inhibitor 6, which targets the glycine flip,86 and pomalidomide, which acts as an E3 ligase binder, connected via linker of various types and lengths (Fig. 4). Among them, compound 7 emerged as the most potent p-p38 degrader since it exhibited higher selectivity for the activated p-p38 compared to the non-phosphorylated counterpart, as confirmed by docking studies and degradation assays performed on various neuronal cells, such as mouse astrocytes (C8-D1A), mouse neuroblasts (N2a) and mouse hippocampal neuronal cells (HT22). Furthermore, compound 7 selectively targets p-p38, significantly reducing its level, compared to a panel of 96 kinases sharing a similar function and/or structure with p38. Additionally, the phosphorylation state of upstream MAPK kinases (MAPKKs), which is responsible for the p-38 activation, is not affected by compound 7, confirming its selectivity.
![]() | ||
| Fig. 4 Design of p38α MAPK-directed PROTAC. p38α MAPK recruiting elements are highlighted in light green, while ligase-recruiting elements are highlighted in light orange. | ||
The degradation activity of compound 7 depends on its interaction with CRBN, as p-p38 degradation was hindered when cells were treated with the compound lacking the pomalidomide moiety. This highlights the crucial role of pomalidomide in inducing the effect. Furthermore, treatment with MG-132 decreased the degradation rate of p-p38, whereas inhibition of the lysosomal pathway using chloroquine (CQ) had no effect on p-p38 levels. These findings further confirmed that the degradation was mediated through the UPS. Compound 7 proved to be able to reduce the neuroinflammation mediated by p-p38, which plays an important role in the regulation of proinflammatory responses by promoting the mRNA expression of pro-inflammatory cytokines. Treatment of BV-2 microglial cells with increasing concentrations of compound 7 significantly decreased the levels of IL-6, IL-8, IL-12 and TNF-α in a dose-dependent manner. In contrast, the p38 mRNA level remained unchanged, as proof that suppression of the neuroinflammation depends on a degradation mechanism rather than the inhibition of p38 synthesis.
Intranasal administration of compound 7 suppressed Aβ deposition-induced neuroinflammation in 5xFAD mice aged 8 to 9 months old, showing a notable reduction in p-p38 levels and in Aβ deposition in both cortex and hippocampus after 1 month treatment. Moreover, treatment with compound 7 improved MWM performance in terms of spatial memory and learning, suggesting beneficial effects on cognitive function. After treatment with compound 7, the level of p-Tau decreased in both the 5xFAD model and the rat AD model PS19, showing memory improvement. Taken together, these results demonstrated that compound 7 decreases Aβ deposition, p-Tau levels, and proinflammatory cytokine production by degrading p-p38, leading to pathological amelioration in AD rat model. Furthermore, the intranasal administration confirmed to be an efficient strategy to reach the CNS by bypassing the BBB, not easily penetrated by CNS-PROTACs, due to their relevant molecular weight.
Significant effort has already been made to target neurodegenerative disease-related proteins, such as tau, with degraders or proximity-inducing modulators.89 However, there are several problems associated with the use of PROTACs, mainly related to their unfavorable chemical–physical properties, i.e. high molecular weight, high H-bond donor and acceptor number etc. Indeed, PROTACs usually have poor cellular permeability and oral bioavailability. Moreover, these kinase-directed PROTACs are expected to exhibit higher concentrations in the periphery than in the CNS, leading to severe on- and off-target side effects. However, these issues could be reduced by exploiting CNS-specific E3 ligases, such as RNF18290 and tripartite motif-containing protein 9 (TRIM9),91 enabling tissue- and cell-type-specific target degradation. Furthermore, PROTACs optimization continues to be an empirical, and time-consuming process of “trial and error” driven by numerous iterative cycles of data collected from compound testing. Fortunately, new platforms are emerging to accelerate the process of PROTAC discovery and optimization, such as the direct-to-biology approach, which combines high-throughput chemistry with high-throughput cell-based assays.72,92
A boost to the research for new degraders of AD-related kinases may come from the success of the medicinal chemistry campaign that led to the discovery of ARV-102, an leucine-rich repeat kinase 2-targeting PROTAC exhibiting remarkable BBB penetration, safety and good in vivo degradation activity.31
We strongly believe that the examples herein reported clearly demonstrate encouraging potential for the treatment of neurodegenerative diseases, and we are optimistic that new studies for the treatment of AD will complement these efforts, broadening the therapeutic landscape.
| This journal is © The Royal Society of Chemistry 2025 |