Next Article in Journal
Stability Assessment of Furosemide Oral Suspension in Hospital Extemporaneous Preparations
Previous Article in Journal
Regulation of L-Lactate in Glutamate Excitotoxicity Under Cerebral Ischemia: Pathophysiology and Preventive Strategy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Computational Insights into the Polypharmacological Landscape of BCR-ABL Inhibitors: Emphasis on Imatinib and Nilotinib

1
Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
2
Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
3
Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2025, 18(7), 936; https://doi.org/10.3390/ph18070936
Submission received: 2 May 2025 / Revised: 8 June 2025 / Accepted: 13 June 2025 / Published: 20 June 2025

Abstract

Background: BCR-ABL inhibitors such as imatinib and nilotinib exhibit multi-kinase activity that extends beyond oncology, offering significant potential for drug repurposing. Objectives: This study aims to systematically evaluate and prioritize the repurposing potential of BCR-ABL inhibitors, particularly imatinib and nilotinib. Methods: An integrated pharmacoinformatics framework was applied to analyze seven BCR-ABL inhibitors. Structural clustering, cheminformatics analysis, and transcriptomic profiling using the Connectivity Map were employed to evaluate structural relationships, target profiles, and gene expression signatures associated with non-oncology indications. Results: Structurally, imatinib and nilotinib clustered closely, while HY-11007 exhibited distinct features. Nilotinib’s high selectivity correlated with strong transcriptional effects in neurodegeneration-related pathways (e.g., HSP90 and LYN), whereas imatinib’s broader kinase profile (PDGFR and c-KIT) was linked to fibrosis and metabolic regulation. Connectivity Map analysis identified more than 30 non-cancer indications, including known off-target uses (e.g., imatinib for pulmonary hypertension) and novel hypotheses (e.g., nilotinib for Alzheimer’s via HSPA5 modulation). A substantial portion of these predictions aligned with the existing literature, underscoring the translational relevance of the approach. Conclusions: These findings highlight the importance of integrating structure–activity relationships and transcriptomic signatures to guide rational repurposing. We propose prioritizing nilotinib for CNS disorders and imatinib for systemic fibrotic diseases, supporting their advancement into preclinical and clinical evaluation. More broadly, this framework offers a versatile platform for uncovering hidden therapeutic potential across other drug classes with complex polypharmacology.

1. Introduction

Breakpoint Cluster Region–Abelson Murine Leukemia Viral Oncogene Fusion Protein (BCR-ABL) inhibitors, such as imatinib, nilotinib, and related compounds, were originally developed for the treatment of chronic myeloid leukemia (CML) and other cancers expressing the BCR-ABL fusion oncogene [1]. More recently, these inhibitors have demonstrated promising potential in non-cancer indications, owing to their multi-kinase activity and off-target inhibition of kinases such as PDGFR, c-KIT, FLT3, and native ABL1 [2]. Moreover, their adverse effects, including cardiovascular toxicity, fluid retention, and cytopenias, further highlight their polypharmacological profiles [3]. Clinical investigations have also explored the use of nilotinib and bosutinib in neurodegenerative diseases, providing further evidence of their therapeutic versatility beyond oncology [4].
Imatinib (Gleevec®) was the first tyrosine kinase inhibitor (TKI) designed to selectively inhibit the BCR-ABL fusion protein, which results from the oncogenic Philadelphia chromosome translocation occurring in chronic myeloid leukemia (CML) and some cases of acute lymphoblastic leukemia (ALL) [5]. It has moderate selectivity towards BCR-ABL. It also targets ABL1, ARG, c-KIT (CD117), DDR1, NQO2, PDGFRα, and PDGFRβ. Nilotinib is a highly selective BCR-ABL inhibitor with minimal off-target kinase activity compared to imatinib and other BCR-ABL inhibitors targeting the ATP site, but it is less selective than asciminib (ABL001), which binds allosterically to the myristoyl pocket of ABL1 [6]. Imatinib and nilotinib bind the ATP-binding site of BCR-ABL; both bind the inactive form of the ABL protein.
Despite their primary use in oncology, BCR-ABL inhibitors have shown emerging promise in non-cancer indications, largely attributable to their inhibition of kinases implicated in fibrosis, neurodegeneration, immune modulation, and metabolic regulation [7]. Therefore, BCR-ABL inhibitors are not only historically significant for their pioneering role in targeted cancer therapy via well-characterized mechanisms, but are also pharmacologically versatile, making them unique tools for both precision oncology and the expanding field of drug repurposing and systems pharmacology. This expanding therapeutic scope is supported not only by preclinical studies but also by the clinical observations of their off-target activities. Understanding the structural determinants, transcriptional effects, and network pharmacology underlying the broader impact of BCR-ABL inhibitors is therefore critical for rational drug repurposing and precision medicine initiatives.
In this study, we present a comprehensive informatics workflow to explore the structural, biological, and therapeutic diversity of BCR-ABL inhibitors, with a particular focus on the distinct transcriptomic profiles of imatinib and nilotinib. We begin by applying cheminformatics approaches to evaluate the chemical and structural diversity across the BCR-ABL inhibitor class. Subsequently, we employ a chemogenomics strategy to characterize the transcriptional effects of these compounds and generate gene expression signatures. These signatures are then used to query the Connectivity Map (CMap) [8] database, the gene expression L1000 platform version 1.0, to identify compounds and genetic perturbations that elicit similar gene expression responses to BCR-ABL inhibitors. Finally, the matched compounds and gene perturbations are prioritized and examined to generate polypharmacology hypotheses, which are further supported through evidence mined from the biomedical literature. This integrative and cost-effective strategy provides a scalable tool for investigating the polypharmacology of other drug classes and guiding hypothesis-driven drug repurposing projects.

2. Results

An informatics workflow (Figure 1) was developed and applied to study the network pharmacology of BCR-ABL inhibitors, based on methods developed by Hajjo et al. [9,10,11,12], to formulate testable hypotheses regarding non-cancer indications and polypharmacological mechanisms.

2.1. Chemical Dissimilarity

All seven BCR-ABL inhibitors analyzed contained heterocyclic cores (e.g., pyrimidine, quinoline, and thiazole), which is essential for ATP-binding site interactions; this is a feature that is linked to cross-pharmacology with kinases like SRC and aurora kinases. Cheminformatics analysis using pairwise Tanimoto distances from binary fingerprints (Figure 2A) showed that nilotinib and imatinib were the most similar (distance = 0.48), while bosutinib, dasatinib, and tozasertib formed a moderately similar subcluster (~0.7–0.8); HY-11007 and AT-9283 were more structurally distinct (>0.85). Pairwise Euclidean distances and 2D alvaDesc descriptors (Figure 2B) captured physicochemical dissimilarity, with tozasertib–dasatinib and tozasertib–imatinib showing the greatest similarity (distances ~8.6–8.8), and nilotinib and HY-11007 showing the greatest divergence (≥11.5). This broader range (~8.6 to 12.1) indicated that Euclidean metrics detected subtler variations compared to Tanimoto distances. A total of 3874 2D molecular descriptors (Supplementary Table S1) were calculated out of 5305 available descriptors, after excluding all 3D descriptors. Of these, 1889 descriptors were used for the principal component analysis (PCA) following the elimination of descriptors with constant or near-constant values, descriptors with at least one missing value (or all missing values), and descriptors with a standard deviation less than 0.0001.
The principal component analysis (PCA) of the descriptors (Figure 2C) further confirmed chemical diversity, i.e., nilotinib and HY-11007 were well-separated along the Y-axis, while imatinib, bosutinib, dasatinib, and tozasertib clustered together. AT-9283 appeared isolated along the X-axis, reflecting unique physicochemical features. The DLS_cons color scale (red = high consensus similarity) visualized relative drug-likeness. Overall, complementary molecular representations effectively revealed structural and physicochemical diversity among the inhibitors.

2.2. Biological Dissimilarity

The seven BCR-ABL inhibitors analyzed, identified from the Connectivity Map (CMap) based on their annotated mechanisms of action (Figure 3A), exhibited distinct pharmacological profiles. Evidence from the literature ranks nilotinib as the most selective BCR-ABL inhibitor, followed by imatinib. AT-9283 and tozasertib also target aurora kinases, while bosutinib and dasatinib more potently inhibit SRC family kinases. HY-11007, in contrast, primarily targets FLT3 and JAK2, distinguishing it mechanistically. CMap data further supported these differences at the transcriptional level. As shown in Figure 3B, the inhibitors displayed diverse transcriptional activity scores (TASs) and connectivity patterns across nine cancer cell lines. Tozasertib, dasatinib, and nilotinib induced strong, consistent responses (TAS ≥ 0.5), reflected by thick black bars and dense red connectivity chords, indicating broad-spectrum activity. In contrast, imatinib and AT-9283 showed moderate, cell-line-specific responses, while HY-11007 exhibited minimal transcriptional activity, suggesting limited cellular engagement despite structural similarity to the other inhibitors.

2.3. Hypothesis Generation Polypharmacological Effects

Results from the CMap analysis (Figure 4) suggest that BCR-ABL inhibitors exert broader effects beyond ABL inhibition, consistent with their profiles as multi-target kinase inhibitors. Predicted compound and genetic connections could support the generation of strong hypotheses regarding mechanisms of action, off-target effects, and drug repurposing opportunities by leveraging transcriptomic similarities identified through high-scoring positive CMap connections.

2.3.1. Non-Cancer Indications of BCR-ABL Inhibitors

The CMap analysis revealed distinct transcriptional profiles among the seven BCR-ABL inhibitors (Figure 4A). Compounds such as dasatinib, bosutinib, tozasertib, and AT-9283 exhibited widespread positive and negative connections across multiple classes, compounds, and genes. A strong connectivity with unrelated perturbagen classes (e.g., aurora kinases, EGFR inhibitors, VEGFR inhibitors, PDGFR inhibitors, and HIV protease inhibitors), which are not directly tied to BCR-ABL signaling, indicated a broad polypharmacology and a reduced selectivity of inhibitors, consistent with their known multi-kinase activity.
Compound connections shown in Table 1 further confirmed strong similarities to EGFR inhibitors (e.g., afatinib, HG-6-64-01, and canertinib) and other multi-kinase inhibitors (e.g., tetrindole, SU-11652, and WZ-4-145). Negative compound connections, such as cholic acid, BRD-A80383043, and BAS-09104376, further suggested opposing transcriptional effects, possibly linked to metabolic or unknown pathways.
Gene-level connections showed strong similarity with ERF1, C9ORF96, UGC, and others, with a notable overlap with the PI3K/AKT and RAS signaling pathways (e.g., AKT3, KRAS, and FBP4), reinforcing the involvement of oncogenic signaling. Few negative gene connections (e.g., PIK3C3, XRCC4, CTSK, and P2RY12) pointed to possible effects on autophagy, DNA repair, and inflammation.

2.3.2. Non-Cancer Indications of Imatinib

Imatinib connected strongly to other tyrosine kinase inhibitors, but also showed connectivity to the diverse drug classes shown in Table 2, including serotonin and dopamine receptor antagonists, indicating off-target transcriptional effects. Gene connectivity revealed a partial overlap with relevant signaling components to BCR-ABL signaling effects but also included non-BCR-ABL-related genes, suggesting a less-focused transcriptional footprint than nilotinib. The non-cancer indications connected to imatinib via CMap positive-scoring connections are diverse, covering neurological, cardiovascular, metabolic, inflammatory, and infectious diseases. There is a strong neuropsychiatric signal, broader than seen with nilotinib (Table 3), but also a clear footprint in immune modulation and vascular pathology.
Table 2. Top CMap positive-scoring perturbagen connections to imatinib and their known non-cancer indications.
Table 2. Top CMap positive-scoring perturbagen connections to imatinib and their known non-cancer indications.
#PerturbagenType ScoreNon-Cancer Indications
1Mineralocorticoid AgonistClass98.22Orthostatic Hypotension (FDA-approved); Addison Disease (FDA-approved).
2PLK InhibitorClass97.63Inflammation (Preclinical) [58]; HIV Infections (Preclinical) [59].
3Dopamine Receptor Grp1Class96.93Parkinson’s Disease (Clinical) [60]; Depressive Disorder (Preclinical) [61].
4PKC ActivatorClass95.54Alzheimer’s Disease (Clinical) [62]; HIV infection (Clinical) [62].
5VEGFR InhibitorClass95.45Uveitis (FDA-approved); Atherosclerosis (Preclinical) [63].
6Opioid Receptor AgonistClass95.09Pain (FDA-approved); Cough (FDA-approved).
7Dopamine Receptor Grp2Class92.62Schizophrenia (FDA-approved); Parkinson’s Disease (Clinical) [60]; Alzheimer’s Disease (Clinical) [64].
8ROCK InhibitorClass90.26Glaucoma (FDA-approved) [65]; Hypertension, Pulmonary (Clinical) [66]; Parkinson’s Disease (Clinical) [67].
9Na-K-Cl Transporter InhibitorClass89.83Kidney Diseases; Hypertension (FDA-approved for non-selective inhibitors); Epilepsy (Clinical for selective inhibitors) [68]; Autistic Disorder (Clinical for selective inhibitors) [69].
10Phosphodiesterase InhibitorClass89.61Erectile dysfunction (FDA-approved); Pulmonary Disease, Chronic Obstructive (FDA-approved); Heart Diseases (FDA-approved); Schizophrenia (Clinical) [70].
11ImatinibCompound99.97Heart Diseases (Clinical) [3]; Asthma (Clinical) [71]; Insulin Resistance (Clinical) [72].
12LorazepamCompound99.61Anxiety Disorders (FDA-approved).
13ParecoxibCompound99.56Pain (FDA-approved); Inflammation (FDA-approved).
14KUC103904NCompound99.51Not detected.
15CefalexinCompound99.51Surgical Wound Infection (FDA-approved); Urinary Tract Infections (FDA-approved); Respiratory Tract Infections (FDA-approved); Skin Diseases, Infectious (FDA-approved); Otitis Media (FDA-approved).
16SNS-314Compound99.37Pulmonary Fibrosis (Clinical) [73]; Arthritis, Rheumatoid (Clinical) [74]; Alzheimer’s Disease (Preclinical) [13]; Psoriasis (Preclinical) [75].
17PioglitazoneCompound99.28Diabetes Mellitus; Insulin Resistance (FDA-approved).
18GSK-1904529ACompound99.12None found.
19BRD-A97035593Compound99.09None found.
20TandutinibCompound99.07Parkinson’s Disease (Preclinical) [2].
21STAT4Gene (kd)99.34Autoimmune Diseases (Clinical) [76]; Inflammation (Clinical); (Preclinical) [77]; Hypersensitivity (Preclinical) [77].
22YTHDF2Gene (kd)99.29Alzheimer’s Disease (Preclinical) [78].
23ZNF92Gene (kd)99.21Alzheimer’s Disease (Preclinical) [79]; Parkinson’s Disease (Preclinical) [79].
24C2Gene (oe)99.12Systemic Lupus Erythematosus (Clinical) [80]; Sjögren’s Syndrome (Clinical) [80].
25CLPBGene (oe)99.07Neutropenia (Clinical) [81]; Parkinson’s Disease (Preclinical) [82].
26KIF5CGene (kd)99.03Neurodegenerative Diseases (Preclinical) [83].
27TKTGene (oe)98.98Diabetes Mellitus (Preclinical) [84]; Alzheimer’s Disease (Preclinical) [85].
28ACAT1Gene (kd)98.82Hypertension (Preclinical) [86]; Dementia (Preclinical) [87].
29SLC3A2Gene (oe)98.74Wound Healing (Preclinical) [88].
30YTHDF1Gene (kd)98.74Diabetes Mellitus (Clinical) [89]; Alzheimer’s Disease (Preclinical) [90].
Non-cancer indications not identified in the literature or publicly available sources, as of 28 April 2025, are denoted as ‘None found’. Validity levels are described in the Methods section.
The frequency analysis of the non-cancer indications of strong CMap positive-scoring connections indicated that imatinib exhibits a broad systemic pharmacology, consistent with its known off-target actions affecting immune cells, vasculature, and metabolism. This analysis highlighted Alzheimer’s disease (seven times), Parkinson’s disease (six times), diabetes mellitus (four times), hypertension (four times), inflammation (three times), schizophrenia (two times), heart diseases (two times), and insulin resistance (two times). Other non-cancer indications that appeared only once are listed in Table 2. These findings are consistent with imatinib’s multi-kinase inhibition profile, particularly against PDGFR, c-KIT, and ABL kinases, and suggest broad repositioning opportunities across CNS, cardiovascular, metabolic, and autoimmune conditions.

2.3.3. Non-Cancer Indications of Nilotinib

Nilotinib showed a tight clustering of connections, specifically with aurora kinase inhibitors, heat shock protein inhibitors, and other BCR-ABL or tyrosine kinase-related perturbagens. Its compound connectivity profile is extremely specific, with perfect (score = 100) or near-perfect similarity to related kinase inhibitors. Gene-level connectivity includes genes directly involved in cell cycle regulation and kinase signaling, reinforcing its focused biological activity and minimal off-target effects.
The frequency analysis of non-cancer indications (using MeSH terms) indicated that nilotinib’s top CMap positive-scoring perturbagen connections are strongly enriched for neurological, autoimmune, fibrotic, and inflammatory diseases (Table 3). This analysis highlighted Alzheimer’s disease (six times), autoimmune diseases (three times), diabetes mellitus (three times), Parkinson’s disease (three times), and pulmonary fibrosis (three times). Other notable indications included amyotrophic lateral sclerosis, rheumatoid arthritis, COVID-19, ulcerative colitis, inflammation, leukoencephalopathy, nervous system diseases, neurodegenerative diseases, and psoriasis, reflecting a diverse enrichment across neurodegenerative, autoimmune, inflammatory, and infectious disease categories. These findings regarding nilotinib’s polypharmacology are consistent with previous evidence, which highlighted neurodegeneration and immunomodulation as key non-cancer repositioning opportunities [91]. However, this study suggests more repurposing opportunities and underscores the importance of multi-pathway inhibitors in identifying therapeutic strategies for complex diseases.
Table 3. Top CMap positive-scoring perturbagen connections to nilotinib and their known non-cancer indications.
Table 3. Top CMap positive-scoring perturbagen connections to nilotinib and their known non-cancer indications.
#PerturbagenTypeScoreNon-Cancer Indications
1Aurora Kinase inhibitor Grp1Class99.90Alzheimer’s Disease (Preclinical) [13]; Liver Cirrhosis, Experimental (Preclinical) [92]; Pulmonary Fibrosis (Preclinical) [14].
2JAK inhibitorClass99.28Alopecia Areata (FDA-approved); Colitis, Ulcerative (FDA-approved); Crohn’s Disease (FDA-approved); Vitiligo (FDA-approved); Arthritis, Rheumatoid (Clinical) [93]; Autoimmune Diseases (Clinical) [94]; Arthritis (Clinical) [95]; Psoriasis (Clinical) [95]; Dermatitis, Atopic (Clinical) [96]; Diabetes Mellitus (Clinical) [97].
3Vesicular transportClass99.28None found.
4EGFR inhibitorClass99.09Psoriasis (Preclinical) [98]; Pulmonary Fibrosis (Preclinical) [99]; Atherosclerosis (Preclinical) [100]; Neovascularization, Pathologic (Preclinical) [101].
5BRAF RAF1 inhibitorClass98.91Pulmonary Fibrosis (Preclinical) [28]; Arthritis, Rheumatoid (Preclinical) [27].
6EIF proteinsClass98.73Leukoencephalopathy (Preclinical) [102]; Diabetes Mellitus (In silico) [103].
7NFKB activationClass98.57Inflammation (Clinical) [104]; Neurodegenerative Diseases (Preclinical) [105].
8BCL2 and related protein inhibitorClass98.33Alzheimer’s Disease (Preclinical) [106]; Lupus Erythematosus, Cutaneous (Preclinical) [107];
9GSK3 inhibitorClass98.23Psychiatric Disorders (Clinical) [108]; Alzheimer’s Disease (Preclinical) [109]; Diabetes Mellitus (Clinical) [110]; Inflammation (Preclinical) [111].
10RAR agonist Grp2Class98.21Acne Vulgaris (Clinical) [112].
11NilotinibCompound99.99Parkinson’s Disease (Clinical) [7]; Alzheimer’s Disease (Clinical) [113]; Amyotrophic Lateral Sclerosis (Preclinical) [114].
12AT-9283Compound99.79Hypertension, Pulmonary (Preclinical) [115]; COVID-19 (In silico) [116].
13AlisertibCompound99.68None found.
14ZM-447439Compound99.61None found.
15Avrainvillamide-analog-2Compound99.58Antibacterial Activity (Preclinical) [117].
16TozasertibCompound99.40Optic Nerve Injuries (Preclinical) [118].
17CrizotinibCompound99.38None found.
18ErastinCompound99.30None found.
19KI-8751Compound99.19Cystitis (Preclinical) [119].
20MK-5108Compound99.12Kidney Fibrosis (Preclinical) [120].
21HSPA5Gene (kd)99.84Fatty Liver, Nonalcoholic (Clinical) [121]; Alzheimer’s Disease (Preclinical) [122]; Parkinson’s Disease (Preclinical) [123]; Myositis (Preclinical) [124]; COVID-19 (Preclinical) [125].
22COPAGene (kd)99.84Autoimmune Diseases (Clinical) [126].
23GMDSGene (oe)99.81Glaucoma, Open-Angle (Preclinical) [127].
24SFPQGene (oe)99.77HIV Infection (Clinical) [128]; Nervous System Diseases (Preclinical) [129]; Congenital Structural Myopathies (Preclinical) [130]; Amyotrophic Lateral Sclerosis (Preclinical) [131].
25HSP90B1Gene (kd)99.73Tuberculosis (Preclinical) [132]; Polycystic Ovary Syndrome (Preclinical) [133].
26HRSP12Gene (oe)99.68Osteoarthritis (Preclinical) [134]; Diabetic Nephropathies (In silico) [135].
27EIF2B2Gene (kd)99.66Leukoencephalopathy (Preclinical) [136]; Nervous System Diseases (Preclinical) [137].
28NIT1Gene (kd)99.63None found.
29NFE2L2Gene (oe)99.63Parkinson’s Disease (Preclinical) [138]; Heart Failure (Preclinical) [139].
30LYNGene (oe)99.63Autoimmune Diseases (Preclinical) [140]; Neurodegenerative Diseases (Preclinical) [140]; Alzheimer’s Disease (Preclinical) [141].
If no non-cancer indications were identified in the published literature or open-source databases, as of 28 April 2025, the designation “None found” was assigned. Validity levels are described in the Methods section.

2.4. Supporting Evidence

Supporting evidence from network biology analyses and/or the biomedical literature is a valuable layer of validation for computational hypotheses. The reliability of such an approach depends on the source quality, context, and convergence with other evidence. Thus, manual curation and critical evaluations are essential.

2.4.1. BCR-ABL PPI Network and Annotated Enriched Pathways

BCR-ABL is a fusion oncogene that is characteristic of CML, whereas BCR and ABL1 are separate genes. Most pathway databases treat BCR-ABL as a functional complex. As shown in Figure 5, BCR-ABL inhibitors not only modulate leukemic signaling but also key hubs such as STAT5, GRB2, and CBL. Enrichment in pathways like ErbB, insulin signaling, and focal adhesion aligns with CMap predictions, suggesting potential for repurposing these inhibitors in neurodegenerative diseases, fibrosis, and metabolic disorders. These findings underscore the broader therapeutic relevance of BCR-ABL beyond CML.

2.4.2. Literature-Based Validation

Most BCR-ABL inhibitors, including imatinib, nilotinib, dasatinib, bosutinib, tozasertib, and AT-9283, are FDA-approved for the treatment of hematological malignancies such as leukemia—myelogenous, chronic, BCR-ABL-positive (CML); acute lymphocytic leukemia (ALL); mastocytosis; gastrointestinal stromal tumors; hypereosinophilic syndrome; and other related conditions. In addition to their established oncological uses, these inhibitors have also been associated with non-cancer indications at various stages of validation, including clinical, preclinical, and hypothesis-driven studies as reported in Table 4. These emerging applications span across areas such as neurodegeneration, inflammation, fibrosis, and metabolic dysfunction. Thus, these literature-based findings confirm that BCR-ABL inhibitors exhibit strong non-cancer repositioning potential toward neurodegenerative, autoimmune, fibrotic, metabolic, and infectious diseases. Therefore, this serves as additional proof that some CMap-predicted indications align closely with known clinical and preclinical observations, supporting the robustness of connectivity mapping for drug repurposing hypotheses. Other unreported indications predicted from the CMap could serve as robust novel hypotheses.
Table 4. Cancer and non-cancer indications for BCR-ABL inhibitors and drug targets.
Table 4. Cancer and non-cancer indications for BCR-ABL inhibitors and drug targets.
#PerturbagenCancer Indications and ValidityNon-Cancer Effects and Validity
1AT-9283Leukemia, Lymphoid (Clinical) [142]; Multiple Myeloma (Clinical) [143]; Neoplasms (Clinical) [144]; Lymphoma, B-Cell (Preclinical) [145].Myeloproliferative Disorders (Preclinical) [146].
2BosutinibLeukemia, Myelogenous, Chronic, BCR-ABL Positive (FDA-approved).Lewy Body Disease (Clinical) [147].
3DasatinibLeukemia, Myelogenous, Chronic, BCR-ABL Positive (FDA-approved).Alzheimer’s Disease (Clinical) [148]; COVID-19 (Clinical) [149]; Hepatitis B, Chronic (Clinical) [150]; Pulmonary Fibrosis (Clinical) [151]; Obesity (Preclinical) [152].
4HY-1107Liver Neoplasms (Preclinical) None found.
5ImatinibLeukemia, Lymphoid (FDA-approved); Mastocytosis (FDA-approved); Leukemia, Myelogenous, Chronic, BCR-ABL Positive (FDA-approved); Dermatofibrosarcoma Protuberans (FDA-approved); Hypereosinophilic Syndrome (FDA-approved); Leukemia, Eosinophilic, Chronic (FDA-approved); Gastrointestinal Stromal Tumors (FDA-approved); Myeloproliferative Disorders (FDA-approved). Anemia, Sickle Cell (Clinical) [153]; Diabetes Mellitus, Type 1 (Clinical) [154]; Pulmonary Fibrosis (Clinical) [155]; Stroke (Clinical) [156]; Liver Cirrhosis (Clinical) [157].
6NilotinibLeukemia, Myelogenous, Chronic, BCR-ABL Positive (FDA-approved).Alzheimer’s Disease (Clinical) [158]; Parkinson’s Disease (Clinical) [7].
7TozasertibGlioma (Clinical) [159]; Melanoma (Preclinical) [160].Hypersensitivity (Preclinical) [161]; Neuralgia (Preclinical) [162].
8BCR-ABLLeukemia, Myelogenous, Chronic, BCR-ABL Positive (Clinical) [163].None found.
9BCRNone found.Autoimmune Diseases (Clinical) [164].
10ABLNone found.Developmental Disabilities (Preclinical) [165]; Parkinson’s Disease (Preclinical) [166].
If no indications were identified in the published literature or open-source databases, as of 28 April 2025, the designation “None found” was assigned. Validity levels are described in the Methods section.
The strong positive CMap associations of imatinib and nilotinib with non-oncological indications suggest high-confidence repurposing opportunities warranting clinical investigation. Imatinib’s robust connectivity with PDGFR and c-KIT indicates its potential efficacy in treating pulmonary fibrosis. PDGFR-α/β signaling is known to drive fibroblast proliferation, migration, and extracellular matrix production, processes which are central to fibrotic progression; thus, its inhibition can attenuate these fibrotic responses [167]. Similarly, c-KIT facilitates the recruitment of bone marrow-derived progenitor cells that differentiate into myofibroblasts, contributing to fibrosis; inhibiting c-KIT may reduce this pathogenic cell population [168].
Nilotinib’s strong positive connectivity with HSP90, HSPA5, and LYN suggests repurposing potential for Alzheimer’s disease. HSP90, HSPA5, and LYN are implicated in key Alzheimer’s disease pathologies, including neuroinflammation and disrupted protein homeostasis. Evidence indicates that nilotinib may exert neuroprotective effects by promoting the autophagic clearance of misfolded proteins, reducing endoplasmic reticulum (ER) stress, and modulating the unfolded protein response (UPR) pathways [169,170]. Nilotinib’s inhibition of HSP90 could reduce tau phosphorylation and amyloid-β accumulation, further supporting its therapeutic potential in Alzheimer’s disease. It is known that HSP90 activation drives the production of pathological tau aggregates [171]. Furthermore, it has been reported that the administration of Hsp90 inhibitors could prevent Aβ-induced neurotoxicity by increasing the levels of HSP70 and Hsp90 [172].

2.5. Mechanistic Insight

The analysis of strong positive gene connections with CMap scores ≥ 90.00 for both imatinib and nilotinib (Supplementary Tables S2 and S3), using protein–protein interaction networks and pathway enrichment analysis, revealed distinct biological signatures for each compound (Figure 6). A complete list of enriched pathways is provided in Supplementary Tables S4 and S5.
Imatinib’s associated network is comparatively sparse and modular, with a lower connectivity among gene products. This pattern suggests a broader and less convergent transcriptional response, potentially involving multiple biological domains without a single dominant signaling axis. The top KEGG-enriched pathways linked to imatinib include EGFR signaling, glioma, neurotrophin signaling, and various metabolic processes.
In contrast, the PPI network associated with nilotinib is highly interconnected, indicating a focused and coherent biological response. Its strong enrichment in KEGG immune-related pathways, including TNF, NFκB, Toll-like receptor, IL17, and NOD-like receptor signaling, highlights alignment with core immunoregulatory mechanisms.

3. Discussion

This study presents a comprehensive informatics workflow for investigating the network pharmacology of BCR-ABL inhibitors, leveraging cheminformatics, transcriptomics, and literature mining processes to uncover their polypharmacological potential beyond oncology. The results highlight structural and biological diversity among these inhibitors, their broad transcriptional effects, and their potential repurposing for non-cancer indications.
A cheminformatics analysis revealed significant chemical diversity among the seven BCR-ABL inhibitors, with distinct structural clusters identified by Tanimoto fingerprint and Euclidean distance metrics. Nilotinib and imatinib showed the highest similarity, while HY-11007 and AT-9283 were structurally distinct, as confirmed by PCA. Integrating fingerprint- and descriptor-based metrics uncovered orthogonal dimensions of similarity, supporting the hypothesis that structural diversity underlies the polypharmacologic and off-target profiles of these inhibitors. Compounds with high DLS_cons values clustered within favorable drug-like chemical spaces, whereas structurally distant compounds, though potentially potent, may require formulation or ADMET optimization. These findings highlight the value of integrated cheminformatics approaches in modern drug design, network pharmacology, and precision medicine.
Biologically, the BCR-ABL inhibitors exhibited varying selectivity and polypharmacology. Nilotinib was the most selective, while dasatinib and bosutinib showed broader kinase inhibition, including SRC family targets. Despite structural similarity, HY-11007 displayed minimal transcriptional activity, indicating limited cellular engagement. CMap analysis corroborated these patterns, with dasatinib and nilotinib inducing strong transcriptional responses, whereas HY-11007 showed weak activity. These findings highlight the importance of integrative approaches that consider both structural and functional profiles when repurposing kinase inhibitors.
The CMap analysis revealed extensive polypharmacology among BCR-ABL inhibitors, with strong connections to unrelated drug classes (e.g., EGFR inhibitors and VEGFR inhibitors) and genes involved in diverse pathways (e.g., PI3K/AKT and RAS signaling). These findings align with their known multi-kinase activities and suggest potential repurposing opportunities for non-cancer diseases. Key non-cancer indications for BCR-ABL inhibitors include neurodegenerative diseases, with Alzheimer’s and Parkinson’s diseases frequently being associated, particularly for nilotinib and dasatinib, which have shown preclinical and clinical neuroprotective effects. Fibrotic disorders such as pulmonary and liver fibrosis also emerged, likely reflecting the inhibition of PDGFR and other pro-fibrotic kinases. Additionally, links to autoimmune and inflammatory diseases, including rheumatoid arthritis and psoriasis, were supported by connections to JAK inhibitors and immune-modulatory genes like STAT4. Selective JAK inhibitors such as baricitinib have been primarily developed and approved for rheumatoid arthritis, COVID-19 (emergency use) [173], and atopic dermatitis. Metabolic disorders, notably diabetes mellitus and insulin resistance, were also associated, possibly through the modulation of insulin signaling pathways.
Notably, both imatinib and nilotinib are BCR-ABL inhibitors; their non-cancer profiles diverged markedly. Imatinib exhibited broad systemic effects, with strong associations to cardiovascular (hypertension), metabolic (diabetes), and neuropsychiatric (schizophrenia and depression) conditions, consistent with its multi-kinase inhibition profile, targeting PDGFR and c-KIT. In contrast, nilotinib showed more focused activity in neurodegeneration (Alzheimer’s and Parkinson’s) and autoimmune diseases, reflecting its higher selectivity and unique off-target interactions, such as HSP90 inhibition. These differences underscore how structural features and target selectivity shape repurposing potential, with imatinib offering versatility but greater off-target risks, and nilotinib presenting a more favorable profile for CNS-focused applications.
These predictions serve as repurposing hypotheses for BCR-ABL inhibitors. Predictions were supported by evidence from the literature, as reported in Table 1, Table 2 and Table 3, where many CMap-identified indications matched known preclinical or clinical findings. For example, imatinib’s anti-fibrotic effects in pulmonary fibrosis and nilotinib’s neuroprotective role in Parkinson’s disease are well-documented (references are cited in Table 4). This convergence of computational and experimental data strengthens the validity of the hypotheses generated. Furthermore, the functional protein–protein interaction (PPI) network analysis (Figure 5) revealed that BCR-ABL inhibitors modulate key hubs beyond oncogenic signaling (e.g., STAT5 and GRB2). Enriched pathways included ErbB signaling, insulin signaling, and focal adhesion, which are implicated in neurodegeneration, fibrosis, and metabolic disorders. This system-level perspective further reinforces the CMap predictions and provides mechanistic insights into how these inhibitors might exert therapeutic effects in non-cancer contexts.
A deeper analysis of CMap gene connections for imatinib and nilotinib, using protein–protein interaction networks and pathway enrichment, highlighted distinct mechanistic profiles. Imatinib’s enrichment in EGFR signaling reflects its broader polypharmacological activity and less-selective targeting of BCR ABL. Given the emerging role of EGFR as a central mediator of profibrotic programs across organs, often acting upstream of TGF beta1-driven fibroblast activation [174], imatinib may hold potential as an antifibrotic agent warranting further investigation. In contrast, nilotinib showed strong enrichment in immune-related pathways such as TNF, NFκB, Toll like receptor, IL17, and NOD-like receptor signaling. These pathways are closely linked to neuroinflammatory mechanisms in central nervous system disorders, including Alzheimer’s and Parkinson’s disease [174], suggesting that nilotinib may modulate microglial activation and reduce disease-associated proteinopathies, supporting its repurposing potential in Alzheimer’s and Parkinson’s diseases.

3.1. Translational and Safety Implications

This hypothesis-generating study investigates new therapeutic opportunities for BCR ABL inhibitors beyond oncology. By integrating chemical, transcriptomic, and pathway level data, we identified mechanistic overlaps with key processes in fibrosis, neurodegeneration, and immune-mediated diseases. These findings provide a framework for repurposing approved drugs for conditions with limited treatment options. While not immediately applicable to clinical practice, this work lays the groundwork for future trials by offering testable hypotheses and mechanistic insights to support preclinical validation and targeted therapy design. Our approach may also reduce uncertainty, cost, and timelines in early-stage drug development.
We also acknowledge that kinase inhibitors often present complex safety profiles due to their polypharmacological nature and off-target effects. For example, nilotinib has been associated with an increased risk of cardiovascular events, including arterial occlusion, myocardial ischemia, and stroke, while imatinib is frequently linked to fluid retention, leading to periorbital and generalized edema [175,176]. Although a full in silico toxicogenomic assessment was beyond the scope of this study, we recognize the value of incorporating predictive safety tools into future workflows. This will allow for a more comprehensive evaluation of the risk–benefit profiles of repurposing candidates and further enhance their translational readiness.

3.2. Limitations

Despite the strengths of our integrative workflow, several limitations should be noted. The CMap platform uses immortalized cancer cell lines, which may not reflect the tissue-specific context of diseases like fibrosis or neurodegeneration. Its focus on transcriptomic data excludes proteomic and phenotypic layers that are essential for full mechanistic insight. Variability in cell lines, dosing, and timing may also affect reproducibility, and reliance on inferred expression from a limited set of landmark genes could introduce errors in pathway analysis.

3.3. Future Perspectives

Realizing the translational potential of BCR ABL inhibitors beyond oncology will require coordinated, multidisciplinary efforts. Future research should focus on validating the most promising non-oncologic indications through preclinical studies and targeted clinical trials. High-throughput multi-omics approaches, including transcriptomics, proteomics, and metabolomics, will be essential for mapping the systemic effects of these compounds across disease models, particularly in fibrotic, neurodegenerative, and immune-mediated conditions. Integrating machine learning with systems pharmacology may accelerate the discovery of repurposing opportunities and improve the prediction of adverse events. Additionally, leveraging pharmacogenomic and real-world data will support patient stratification and guide the development of personalized therapeutic strategies.

4. Methods

4.1. Chemical Biology Informatics Workflow

We applied an informatics workflow, based on the methods developed by Hajjo et al. [9,10,11,12]. The bioinformatics workflow method was used to study the network biology of BCR-ABL inhibitors. This workflow involved the following three steps: (1) identifying gene signatures representative of the chemical compound’s biological action; (2) identifying genetic perturbations that result in transcriptomics profiles resembling those of BCR-ABL inhibitors; and (3) a pathway enrichment and literature mining process to elucidate the biological activities of similar gene and compound connections, respectively. Furthermore, cheminformatics methods were employed to assess and compare chemical similarities/dissimilarities between BCR-ABL inhibitors.
In this study, Connectivity Map (CMap) was used to identify transcriptomic signatures and prioritize repurposing hypotheses for BCR ABL inhibitors through the following steps:
  • Querying CMap: Gene expression signatures were retrieved using the search terms “mechanism = BCR ABL inhibitors”, “compound = imatinib”, and “compound = nilotinib”.
  • Generating query signatures: The transcriptomic profiles of compounds identified from the initial search were used as input query signatures.
  • Identifying similar perturbations: These signatures were used to search CMap for compounds and genetic perturbations exhibiting similar expression profiles.
  • Prioritization of hits: Repurposing hypotheses were prioritized based on the top 10 gene and compound connections, all enriched pharmacological classes, and supporting preclinical or clinical evidence.
  • Filtering by connectivity scores: The results were filtered using CMap connectivity scores, as previously described [8], to retain only relevant and strong associations.
  • Pathway enrichment: High-confidence gene connections (CMap scores ≥ 90.00) were used for pathway enrichment analysis to gain mechanistic insights.

4.2. Bioinformatics Methods

4.2.1. The Connectivity Map (CMap)

The CMap computational tool and a chemogenomics database [8] constitute a cornerstone of the chemocentric informatics workflow developed by Hajjo et al. [9,10,11,12]. This tool was used to formulate testable hypotheses by reading compounds, genes, and compound classes that share a strong similarity with BCR-ABL inhibitors on the transcriptomics level. The CMap database lists 1.3 million profiles of transcriptional responses of human cells to chemical and genetic perturbations. Currently, there are 27,927 perturbagens (19,911 small molecules and 7494 genetic perturbagens) generating 476,251 expression signatures in nine human cell lines—PC3, VCAP, A375, A459, HA1E, HCC515, HT29, MCF7, and HEPG2. This database uses the L1000 platform version 1.0 [8], a high-throughput gene expression assay that measures the mRNA transcript abundance of 978 “landmark” genes in human cells.

4.2.2. Protein–Protein Interactions

A systematic search for BCR-ABL nearest neighbor (NN) genes/proteins was conducted in Cytoscape version 3.10.3, using the STRING App version 2.1.0 [177]. All retrieved protein–protein interactions (PPIs), including both physical and functional associations, were collected using a medium confidence threshold of 0.4. In STRING, a confidence score of 0.4 reflects a moderate level of predicted interaction reliability, balancing sensitivity and specificity by including experimentally supported and computationally inferred interactions. The resulting data were then used to generate PPI networks using Cytoscape’s network visualization and analysis tools, followed by enrichment analysis-leveraging modules from Gene Ontology (Biological Process, Molecular Function, and Cellular Component), KEGG, Reactome, WikiPathways, protein domain annotations, DISEASES, COMPARTMENTS, and TISSUES.
Furthermore, PPI networks for high-confidence gene connections (CMap scores ≥ 90.00) of imatinib and nilotinib, generated using Cytoscape’s STRING App version 2.1.0, were used for pathway enrichment analysis to gain mechanistic insights.

4.2.3. Enrichment Analysis

Pathway enrichment analyses for PPI networks were performed using the STRING functional enrichment tool in Cytoscape [177] (version 3.10.3). The significance of each enrichment result was assessed by calculating hypergeometric p-values. Next, all ontology terms were ranked according to their calculated false discovery rates (FDRs). Pathways with FDRs below a threshold of 0.05 were considered statistically significant and were selected for further biological investigation.

4.3. Cheminformatics Methods

4.3.1. Molecular Descriptors

Two-dimensional (2D) molecular descriptors from alvaDesc version 1.0.18 [178] and circular fingerprints (Morgan fingerprints) were generated for BCR-ABL inhibitors. The 2D chemical structures of seven inhibitors were first retrieved from PubChem and exported in standardized Structure Data File (SDF) format. Molecular standardization was performed using RDKit version 2024.03.1 [179], in the Python environment version 3.12.2, to ensure consistency in structural representation. This process included the removal of salts, charge neutralization, tautomer canonicalization, aromatic ring perception (aromatization), and the addition of explicit hydrogen atoms.

4.3.2. Similarity and Distance Matrix Calculation

Pairwise molecular similarities were computed using the following two approaches: (1) Tanimoto coefficients with circular fingerprints (Morgan fingerprints) generated using RDKit, and (2) Euclidean distances with 2D alvaDesc molecular descriptors. The Tanimoto distance was calculated as 1–Tanimoto Similarity, yielding a symmetric distance matrix assembled using pandas version 2.2.2. The distance matrix was visualized using a heatmap generated with Seaborn version 0.13.2 (sns.heatmap) and Matplotlib version 3.8.4. All generated plots were saved in TIFF format.

4.4. Standardization of Condition Terms Using MeSH Vocabulary

Condition terms in all tables were standardized using Medical Subject Headings (MeSH)-controlled vocabulary to ensure terminological consistency. All condition variants (e.g., user-defined, literature-derived, or dataset-specific) were converted to standard descriptors (i.e., MeSH IDs) via automated mapping using the MeSH Resource Description Framework (MeSH RDF) [180].

4.5. Therapeutic Evidence

To systematically assess the confidence and novelty of repurposing candidates, we categorized all perturbagens (including small molecules and genetic perturbations) into four hierarchically ordered evidence levels based on translational validation, as follows:
  • FDA-approved: Interventions that are already approved for non-cancer indications. These reflect previously validated clinical uses and are not novel findings of this study but were included for benchmarking and validation purposes.
  • Clinical: Candidates with reported efficacy in human studies or clinical trials but without formal regulatory approval. These are supported by clinical evidence yet still represent off-label or investigational use.
  • Preclinical: Agents with supportive data from animal models or in vitro experiments but lacking human clinical validation.
  • In silico/Hypothesis: Computational predictions generated in this study that have not yet been tested experimentally.
This tiered framework enabled a structured prioritization of repurposing hypotheses, balancing novelty with confidence. It was consistently applied across all reported associations and is visually encoded in the tables and figures to clearly distinguish between novel predictions and previously validated indications, thereby enhancing interpretability and translational relevance.

5. Conclusions

This study presents an integrative informatics framework that effectively maps the polypharmacology of BCR-ABL inhibitors, highlighting their potential for repurposing in neurodegenerative, fibrotic, autoimmune, and metabolic diseases. The convergence of cheminformatics, transcriptomics, and evidence from the literature demonstrates the power of computational methods in drug repositioning. Notably, imatinib and nilotinib emerged as promising candidates for diverse non-cancer applications, with their distinct profiles offering complementary therapeutic opportunities. In this approach, CMap enabled the identification of biological similarities with imatinib and nilotinib based on transcriptomics effects. In the last step, literature mining identified the biological activities of compound hits, while enrichment analysis identified enriched biological pathways for the identified gene hits, which served as robust supporting evidence for the polypharmacology hypotheses derived from CMap. These findings support the rationale for targeted experimental validation and clinical investigation to translate computational hypotheses into real-world therapies. Moreover, this integrative approach provides a broadly applicable model for studying the polypharmacological effects of other classes of inhibitors.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ph18070936/s1. Supplementary Table S1: List of calculated 2D molecular descriptors generated using alvaDesc. Supplementary Table S2: CMap-derived positive gene expression connections for imatinib (CMap scores ≥ 90.00). Supplementary Table S3: CMap-derived positive gene expression connections for nilotinib (CMap scores ≥ 90.00). Supplementary Table S4: Enriched pathway maps based on the PPI network of imatinib’s CMap-positive gene connections (CMap scores ≥ 90.00). Supplementary Table S5: Enriched pathway maps based on the PPI network of nilotinib’s CMap-positive gene connections (CMap scores ≥ 90.00).

Author Contributions

Conceptualization, R.H.; methodology, R.H.; software, R.H.; validation, R.H., D.A.S., R.A., A.A. and S.K.B.; formal analysis, R.H.; investigation, R.H., D.A.S., R.A., A.A. and S.K.B.; resources, R.H. and D.A.S.; data curation, R.H., D.A.S., R.A., A.A. and S.K.B.; writing—original draft preparation, R.H.; writing—review and editing, R.H. and S.K.B.; visualization, R.H.; supervision, R.H.; project administration, R.H.; funding acquisition, R.H. and D.A.S. All authors have read and agreed to the published version of the manuscript.

Funding

Funding received from the Deanship of Scientific Research at Al-Zaytoonah University of Jordan (Grant numbers: 2023-2022/17/50 and 2025-2024/06/29).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The raw cheminformatics descriptors, transcriptomics profiles from CMap analysis, and processed network pharmacology datasets generated in this study are available on GitHub at [https://github.com/rhajjo/BCR-ABL] (accessed on 15 June 2025). These data should enable the full reproducibility of the reported findings.

Acknowledgments

R.H. and D.A.S. acknowledge the generous funding support from the Deanship of Scientific Research at Al-Zaytoonah University of Jordan.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ABLAbelson Murine Leukemia Viral Oncogene
ALKAnaplastic Lymphoma Kinase
BCR-ABLBreakpoint Cluster Region–Abelson Murine Leukemia Viral Oncogene Fusion Protein
BCRBreakpoint Cluster Region
CMLChronic Myeloid Leukemia
DCMDilated Cardiomyopathy
FDRFalse Discovery Rate
MeSHMedical Subject Headings
PPIProtein–Protein Interaction
PCAPrincipal Component Analysis
TASTranscriptional Activity Score
TKITyrosine kinase inhibitor

References

  1. Roskoski, R. Targeting BCR-Abl in the Treatment of Philadelphia-Chromosome Positive Chronic Myelogenous Leukemia. Pharmacol. Res. 2022, 178, 106156. [Google Scholar] [CrossRef] [PubMed]
  2. Mansour, H.M.; El-Khatib, A.S. Repositioning of Receptor Tyrosine Kinase Inhibitors: From Cancer to Neurodegenerative and Psychiatric Disorders. In Receptor Tyrosine Kinases in Neurodegenerative and Psychiatric Disorders; Academic Press: Cambridge, MA, USA, 2023; pp. 353–401. [Google Scholar] [CrossRef]
  3. Dong, B.; Chen, D.F.; Bu, X.H.; Qin, H.B.; Xi, Y. Effect of Imatinib on DOCA-Induced Myocardial Fibrosis in Rats through P38 MAPK Signaling Pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 2028–2036. [Google Scholar] [CrossRef] [PubMed]
  4. Tocci, D.; Fogel, M.; Gupta, V.; Kim, P.; Latimer, J.; Adlimoghaddam, A.; Robison, L.S.; Albensi, B.C. Beyond Expectations: Investigating Nilotinib’s Potential in Attenuating Neurodegeneration in Alzheimer’s Disease. Alzheimers Res. Ther. 2025, 17, 60. [Google Scholar] [CrossRef] [PubMed]
  5. Druker, B.J.; Talpaz, M.; Resta, D.J.; Peng, B.; Buchdunger, E.; Ford, J.M.; Lydon, N.B.; Kantarjian, H.; Capdeville, R.; Ohno-Jones, S.; et al. Efficacy and Safety of a Specific Inhibitor of the BCR-ABL Tyrosine Kinase in Chronic Myeloid Leukemia. N. Engl. J. Med. 2001, 344, 1031–1037. [Google Scholar] [CrossRef]
  6. Choi, E.J. Asciminib: The First-in-Class Allosteric Inhibitor of BCR::ABL1 Kinase. Blood Res. 2023, 58, S29. [Google Scholar] [CrossRef]
  7. Xie, X.; Yuan, P.; Kou, L.; Chen, X.; Li, J.; Li, Y. Nilotinib in Parkinson’s Disease: A Systematic Review and Meta-Analysis. Front. Aging Neurosci. 2022, 14, 996217. [Google Scholar] [CrossRef]
  8. Subramanian, A.; Narayan, R.; Corsello, S.M.; Peck, D.D.; Natoli, T.E.; Lu, X.; Gould, J.; Davis, J.F.; Tubelli, A.A.; Asiedu, J.K.; et al. A Next Generation Connectivity Map: L1000 Platform and the First 1,000,000 Profiles. Cell 2017, 171, 1437–1452.e17. [Google Scholar] [CrossRef]
  9. Hajjo, R.; Tropsha, A. A Systems Biology Workflow for Drug and Vaccine Repurposing: Identifying Small-Molecule BCG Mimics to Reduce or Prevent COVID-19 Mortality. Pharm. Res. 2020, 37, 212. [Google Scholar] [CrossRef]
  10. Hajjo, R.; Setola, V.; Roth, B.L.; Tropsha, A. Chemocentric Informatics Approach to Drug Discovery: Identification and Experimental Validation of Selective Estrogen Receptor Modulators as Ligands of 5-Hydroxytryptamine-6 Receptors and as Potential Cognition Enhancers. J. Med. Chem. 2012, 55, 5704–5719. [Google Scholar] [CrossRef]
  11. Hajjo, R.; Sabbah, D.A.; Bardaweel, S.K. Chemocentric Informatics Analysis: Dexamethasone Versus Combination Therapy for COVID-19. ACS Omega 2020, 5, 29765–29779. [Google Scholar] [CrossRef]
  12. Hajjo, R.; Momani, E.; Sabbah, D.A.; Baker, N.; Tropsha, A. Identifying a Causal Link between Prolactin Signaling Pathways and COVID-19 Vaccine-Induced Menstrual Changes. NPJ Vaccines 2023, 8, 129. [Google Scholar] [CrossRef] [PubMed]
  13. Afghah, Z.; Khan, N.; Datta, G.; Halcrow, P.W.; Geiger, J.D.; Chen, X. Involvement of Endolysosomes and Aurora Kinase A in the Regulation of Amyloid β Protein Levels in Neurons. Int. J. Mol. Sci. 2024, 25, 6200. [Google Scholar] [CrossRef] [PubMed]
  14. Yang, Y.; Santos, D.M.; Pantano, L.; Knipe, R.; Abe, E.; Logue, A.; Pronzati, G.; Black, K.E.; Spinney, J.J.; Giacona, F.; et al. Screening for Inhibitors of YAP Nuclear Localization Identifies Aurora Kinase A as a Modulator of Lung Fibrosis. Am. J. Respir. Cell Mol. Biol. 2022, 67, 36–49. [Google Scholar] [CrossRef] [PubMed]
  15. Katsha, A.; Soutto, M.; Sehdev, V.; Peng, D.; Washington, M.K.; Piazuelo, M.B.; Tantawy, M.N.; Manning, H.C.; Lu, P.; Shyr, Y.; et al. Aurora Kinase A Promotes Inflammation and Tumorigenesis in Mice and Human Gastric Neoplasia. Gastroenterology 2013, 145, 1312–1322.e8. [Google Scholar] [CrossRef]
  16. Shi, L.; Du, X.; Wang, B.; Zhang, G. CircAKR1B10 Interacts with EIF4A3 to Stabilize AURKA and Promotes IL-22-Induced Proliferation, Migration and Invasion in Keratinocytes. Arch. Dermatol. Res. 2024, 316, 561. [Google Scholar] [CrossRef]
  17. Czókolyová, M.; Hamar, A.; Pusztai, A.; Tajti, G.; Végh, E.; Pethő, Z.; Bodnár, N.; Horváth, Á.; Soós, B.; Szamosi, S.; et al. Effects of One-Year Tofacitinib Therapy on Lipids and Adipokines in Association with Vascular Pathophysiology in Rheumatoid Arthritis. Biomolecules 2022, 12, 1483. [Google Scholar] [CrossRef]
  18. Jung, S.C.; Kang, D.; Ko, E.A. Roles of PDGF/PDGFR Signaling in Various Organs. Korean J. Physiol. Pharmacol. 2024, 29, 139. [Google Scholar] [CrossRef]
  19. He, C.; Medley, S.C.; Hu, T.; Hinsdale, M.E.; Lupu, F.; Virmani, R.; Olson, L.E. PDGFRβ Signaling Regulates Local Inflammation and Synergizes with Hypercholesterolemia to Promote Atherosclerosis. Nat. Commun. 2015, 6, 7770. [Google Scholar] [CrossRef]
  20. Trempus, C.S.; Papas, B.N.; Sifre, M.I.; Bortner, C.D.; Scappini, E.; Tucker, C.J.; Xu, X.; Johnson, K.L.; Deterding, L.J.; Williams, J.G.; et al. Functional Pdgfra Fibroblast Heterogeneity in Normal and Fibrotic Mouse Lung. JCI Insight 2023, 8, e164380. [Google Scholar] [CrossRef]
  21. Schramm, F.; Schaefer, L.; Wygrecka, M. EGFR Signaling in Lung Fibrosis. Cells 2022, 11, 986. [Google Scholar] [CrossRef]
  22. Madan, V.; Ortiz-López, L.I.; Sakunchotpanit, G.; Chen, R.; Nayudu, K.; Nambudiri, V.E. Psoriasis in the Era of Targeted Cancer Therapeutics: A Systematic Review on De Novo and Pre-Existing Psoriasis in Oncologic Patients Treated with Emerging Anti-Neoplastic Agents. Dermatol. Ther. 2024, 14, 1755. [Google Scholar] [CrossRef] [PubMed]
  23. Jiang, D.; Xu, T.; Zhong, L.; Liang, Q.; Hu, Y.; Xiao, W.; Shi, J. Research Progress of VEGFR Small Molecule Inhibitors in Ocular Neovascular Diseases. Eur. J. Med. Chem. 2023, 257, 115535. [Google Scholar] [CrossRef] [PubMed]
  24. Jiang, F.; Wang, M.Q.; Zhang, M.Y.; Gu, S.L.; Xie, Y.W.; Huang, Y.; Zhou, M.Y.; Li, F.L.; Yang, Y.C.; Zhang, P.P.; et al. CPD-002, a Novel VEGFR2 Inhibitor, Relieves Rheumatoid Arthritis by Reducing Angiogenesis through the Suppression of the VEGFR2/PI3K/AKT Signaling Pathway. Int. Immunopharmacol. 2024, 131, 111850. [Google Scholar] [CrossRef] [PubMed]
  25. Fan, Q.; Wu, G.; Chen, M.; Luo, G.; Wu, Z.; Huo, H.; Li, H.; Zheng, L.; Luo, M. Cediranib Ameliorates Portal Hypertensive Syndrome via Inhibition of VEGFR-2 Signaling in Cirrhotic Rats. Eur. J. Pharmacol. 2024, 964, 176278. [Google Scholar] [CrossRef]
  26. Chen, Y.; Tai, Z.; Zhu, C.; Yu, Q.; Zhu, Q.; Chen, Z. Vascular Endothelial Growth Factor A VEGFA Inhibition: An Effective Treatment Strategy for Psoriasis. Int. J. Mol. Sci. 2024, 25, 59. [Google Scholar] [CrossRef]
  27. Su, X.; Yuan, B.; Tao, X.; Guo, W.; Mao, X.; Wu, A.; Wang, Q.; Liu, C.; Zhang, Y.; Kong, X.; et al. Anti-Angiogenic Effect of YuXueBi Tablet in Experimental Rheumatoid Arthritis by Suppressing LOX/Ras/Raf-1 Signaling. J. Ethnopharmacol. 2022, 298, 115611. [Google Scholar] [CrossRef]
  28. Zhang, M.; Wang, W.; Liu, K.; Jia, C.; Hou, Y.; Bai, G. Astragaloside IV Protects against Lung Injury and Pulmonary Fibrosis in COPD by Targeting GTP-GDP Domain of RAS and Downregulating the RAS/RAF/FoxO Signaling Pathway. Phytomedicine 2023, 120, 155066. [Google Scholar] [CrossRef]
  29. Chen, X.; Nie, X.; Lin, X.; Wang, Y.; Zhang, L.; Chen, Z.; Lin, M. Regulation of Keratinocyte Barrier Function and Inflammatory Response by the EGFR-STAT3 Pathway: Potential Therapeutic Implications of Osimertinib and Afatinib. Cytokine 2025, 185, 156802. [Google Scholar] [CrossRef]
  30. Clerk, A.; Meijles, D.N.; Hardyman, M.A.; Fuller, S.J.; Chothani, S.P.; Cull, J.J.; Cooper, S.T.E.; Alharbi, H.O.; Vanezis, K.; Felkin, L.E.; et al. Cardiomyocyte BRAF and Type 1 RAF Inhibitors Promote Cardiomyocyte and Cardiac Hypertrophy in Mice in Vivo. Biochemical Journal 2022, 479, 401. [Google Scholar] [CrossRef]
  31. Bano, S.; Copeland, M.A.; Stoops, J.W.; Orr, A.; Jain, S.; Paranjpe, S.; Mooli, R.G.R.; Ramakrishnan, S.K.; Locker, J.; Mars, W.M.; et al. Hepatocyte-Specific Epidermal Growth Factor Receptor Deletion Promotes Fibrosis but Has No Effect on Steatosis in Fast-Food Diet Model of Metabolic Dysfunction-Associated Steatotic Liver Disease. Cell. Mol. Gastroenterol. Hepatol. 2024, 18, 101380. [Google Scholar] [CrossRef]
  32. Korshunov, V.A.; Murashev, A.N.; Ivashev, M.N.; Dugin, S.F.; Medvedev, O.S. [Comparative Study of Hemodynamic Effects of Antidepressants (Tetrindole and Desipramine) during Immobilization Stress in Hypertensive Rats]. Eksperimental’naia i Klinicheskaia Farmakologiia 2000, 63, 18–20. [Google Scholar] [PubMed]
  33. Chen, Y.L.; Tsaur, M.L.; Wang, S.W.; Wang, T.Y.; Hung, Y.C.; Lin, C.S.; Chang, Y.F.; Wang, Y.C.; Shiue, S.J.; Cheng, J.K. Chronic Intrathecal Infusion of Mibefradil, Ethosuximide and Nickel Attenuates Nerve Ligation-Induced Pain in Rats. Br. J. Anaesth. 2015, 115, 105–111. [Google Scholar] [CrossRef]
  34. Park, J.; Choi, H.; Kim, Y.D.; Kim, S.H.; Kim, Y.; Gwon, Y.; Lee, D.Y.; Park, S.H.; Heo, W.D.; Jung, Y.K. Aberrant Role of ALK in Tau Proteinopathy through Autophagosomal Dysregulation. Mol. Psychiatry 2021, 26, 5542–5556. [Google Scholar] [CrossRef] [PubMed]
  35. Wang, L.; Wang, Y.; Chen, A.; Jalali, A.; Liu, S.; Guo, Y.; Na, S.; Nakshatri, H.; Li, B.Y.; Yokota, H. Effects of a Checkpoint Kinase Inhibitor, AZD7762, on Tumor Suppression and Bone Remodeling. Int. J. Oncol. 2018, 53, 1001. [Google Scholar] [CrossRef] [PubMed]
  36. Ivanov, D.; Drobintseva, A.; Rodichkina, V. Inflammaging: Expanding the Molecular Palette of Cellular Immunophenotype. 2024; 2024040644, Preprints. [Google Scholar]
  37. Mahady, L.J.; He, B.; Malek-Ahmadi, M.; Mufson, E.J. Telomeric Alterations in the Default Mode Network during the Progression of Alzheimer’s Disease: Selective Vulnerability of the Precuneus. Neuropathol. Appl. Neurobiol. 2021, 47, 428–440. [Google Scholar] [CrossRef]
  38. Harmak, H.; Redouane, S.; Charoute, H.; Aniq Filali, O.; Barakat, A.; Rouba, H. In Silico Exploration and Molecular Dynamics of Deleterious SNPs on the Human TERF1 Protein Triggering Male Infertility. J. Biomol. Struct. Dyn. 2023, 41, 14665–14688. [Google Scholar] [CrossRef]
  39. Ain, N.U.; Saith, A.; Ruan, A.; Yang, R.; Burton, A.; Mistry, P.K. Eliglustat and Cardiac Comorbidities in Gaucher Disease: A Pharmacogenomic Approach to Safety and Efficacy. Front. Med. 2025, 12, 1535099. [Google Scholar] [CrossRef]
  40. Zang, C.; Liu, Y.; Chen, H. The Sphingolipids Metabolism Mechanism and Associated Molecular Biomarker Investigation in Keloid. Comb. Chem. High Throughput Screen. 2022, 26, 2003–2012. [Google Scholar] [CrossRef]
  41. Liu, J.; Li, W.; Jiao, R.; Liu, Z.; Zhang, T.; Chai, D.; Meng, L.; Yang, Z.; Liu, Y.; Wu, H.; et al. Miglustat Ameliorates Isoproterenol-Induced Cardiac Fibrosis via Targeting UGCG. Mol. Med. 2025, 31, 55. [Google Scholar] [CrossRef]
  42. Chen, S.; Wang, Y.; Kong, L.; Ji, Y.; Cui, J.; Shen, W. Role of UDP-Glucose Ceramide Glucosyltransferase in Venous Malformation. Front. Cell Dev. Biol. 2023, 11, 1178045. [Google Scholar] [CrossRef] [PubMed]
  43. Brazdis, R.M.; von Zimmermann, C.; Lenz, B.; Kornhuber, J.; Mühle, C. Peripheral Upregulation of Parkinson’s Disease-Associated Genes Encoding α-Synuclein, β-Glucocerebrosidase, and Ceramide Glucosyltransferase in Major Depression. Int. J. Mol. Sci. 2024, 25, 3219. [Google Scholar] [CrossRef] [PubMed]
  44. Alcantara, D.; Timms, A.E.; Gripp, K.; Baker, L.; Park, K.; Collins, S.; Cheng, C.; Stewart, F.; Mehta, S.G.; Saggar, A.; et al. Mutations of AKT3 Are Associated with a Wide Spectrum of Developmental Disorders Including Extreme Megalencephaly. Brain 2017, 140, 2610. [Google Scholar] [CrossRef] [PubMed]
  45. Guerau-de-Arellano, M.; Piedra-Quintero, Z.L.; Tsichlis, P.N. Akt Isoforms in the Immune System. Front. Immunol. 2022, 13, 990874. [Google Scholar] [CrossRef]
  46. Howell, K.R.; Floyd, K.; Law, A.J. PKBγ/AKT3 Loss-of-Function Causes Learning and Memory Deficits and Deregulation of AKT/MTORC2 Signaling: Relevance for Schizophrenia. PLoS ONE 2017, 12, e0175993. [Google Scholar] [CrossRef]
  47. Wang, X.; Zhang, Z.; Peng, P.; Peng, J. The Phenotypic and Genetic Spectrum of AKT3-Related Neurodevelopmental Condition. Sci. Rep. 2025, 15, 7484. [Google Scholar] [CrossRef]
  48. Gu, S.; Dai, H.; Zhao, X.; Gui, C.; Gui, J. AKT3 Deficiency in M2 Macrophages Impairs Cutaneous Wound Healing by Disrupting Tissue Remodeling. Aging 2020, 12, 6928–6946. [Google Scholar] [CrossRef]
  49. Okada, K.; Fukai, A.; Mori, D.; Hosaka, Y.; Yano, F.; Chung, U.I.; Kawaguchi, H.; Tanaka, S.; Ikeda, T.; Saito, T. Identification of SCAN Domain Zinc-Finger Gene ZNF449 as a Novel Factor of Chondrogenesis. PLoS ONE 2014, 9, e115169. [Google Scholar] [CrossRef]
  50. Newbury, L.J.; Wang, J.H.; Hung, G.; Hendry, B.M.; Sharpe, C.C. Inhibition of Kirsten-Ras Reduces Fibrosis and Protects against Renal Dysfunction in a Mouse Model of Chronic Folic Acid Nephropathy. Sci. Rep. 2019, 9, 14010. [Google Scholar] [CrossRef]
  51. Laryushina, Y.; Parakhina, V.; Turmukhambetova, A.; Turgunova, L.; Ibraeva, L.; Amirkhanova, D.; Nildibayeva, F. The Relationship between the Level Fabp4, Risks of Type 2 Diabetes Mellitus, and Cardiovascular Events. Open Access Maced. J. Med. Sci. 2020, 8, 762–768. [Google Scholar] [CrossRef]
  52. Wu, Y.W.; Chang, T.T.; Chang, C.C.; Chen, J.W. Fatty-Acid-Binding Protein 4 as a Novel Contributor to Mononuclear Cell Activation and Endothelial Cell Dysfunction in Atherosclerosis. Int. J. Mol. Sci. 2020, 21, 9245. [Google Scholar] [CrossRef] [PubMed]
  53. Taguchi, K.; Chen, L.; Usawachintachit, M.; Hamamoto, S.; Kang, M.; Sugino, T.; Unno, R.; Tzou, D.T.; Sherer, B.A.; Okada, A.; et al. Fatty Acid–Binding Protein 4 Downregulation Drives Calcification in the Development of Kidney Stone Disease. Kidney Int. 2020, 97, 1042–1056. [Google Scholar] [CrossRef] [PubMed]
  54. Başarır Sivri, F.N.; Çiftçi, S. A New Insight into Fatty Acid Binding Protein 4 Mechanisms and Therapeutic Implications in Obesity-Associated Diseases: A Mini Review. Mol. Nutr. Food Res. 2024, 68, 2300840. [Google Scholar] [CrossRef] [PubMed]
  55. Guo, Y.; Sun, C.-K.; Tang, L.; Tan, M.-S. Microglia PTK2B/Pyk2 in the Pathogenesis of Alzheimer’s Disease. Curr. Alzheimer Res. 2023, 20, 692–704. [Google Scholar] [CrossRef]
  56. Ionescu, M.I. Adenylate Kinase: A Ubiquitous Enzyme Correlated with Medical Conditions. Protein J. 2019, 38, 120–133. [Google Scholar] [CrossRef]
  57. Thawanaphong, S.; Nair, A.; Volfson, E.; Nair, P.; Mukherjee, M. IL-18 Biology in Severe Asthma. Front. Med. 2024, 11, 1486780. [Google Scholar] [CrossRef]
  58. Baldrighi, M.; Doreth, C.; Li, Y.; Zhao, X.; Warner, E.; Chenoweth, H.; Kishore, K.; Umrania, Y.; Minde, D.-P.; Thome, S.; et al. PLK1 Inhibition Dampens NLRP3 Inflammasome–Elicited Response in Inflammatory Disease Models. J. Clin. Investig. 2023, 133, e162129. [Google Scholar] [CrossRef]
  59. Zhou, D.; Hayashi, T.; Jean, M.; Kong, W.; Fiches, G.; Biswas, A.; Liu, S.; Yosief, H.O.; Zhang, X.; Bradner, J.; et al. Inhibition of Polo-like Kinase 1 (PLK1) Facilitates the Elimination of HIV-1 Viral Reservoirs in CD4+ T Cells Ex Vivo. Sci. Adv. 2020, 6, eaba1941. [Google Scholar] [CrossRef]
  60. Latif, S.; Jahangeer, M.; Maknoon Razia, D.; Ashiq, M.; Ghaffar, A.; Akram, M.; El Allam, A.; Bouyahya, A.; Garipova, L.; Ali Shariati, M.; et al. Dopamine in Parkinson’s Disease. Clin. Chim. Acta 2021, 522, 114–126. [Google Scholar] [CrossRef]
  61. Desormeaux, C.; Demars, F.; Davenas, E.; Jay, T.M.; Lavergne, F. Selective Activation of D1 Dopamine Receptors Exerts Antidepressant-like Activity in Rats. J. Psychopharmacol. 2020, 34, 1443–1448. [Google Scholar] [CrossRef]
  62. Kawano, T.; Inokuchi, J.; Eto, M.; Murata, M.; Kang, J.H. Activators and Inhibitors of Protein Kinase C (PKC): Their Applications in Clinical Trials. Pharmaceutics 2021, 13, 1748. [Google Scholar] [CrossRef] [PubMed]
  63. Dabravolski, S.A.; Khotina, V.A.; Omelchenko, A.V.; Kalmykov, V.A.; Orekhov, A.N. The Role of the VEGF Family in Atherosclerosis Development and Its Potential as Treatment Targets. Int. J. Mol. Sci. 2022, 23, 931. [Google Scholar] [CrossRef] [PubMed]
  64. Liu, X.; Cheng, Z.Y.; Li, Y.F.; Liu, C.; Wang, C.; Gong, X.J.; He, L. Dopamine D2 Receptor Agonist Bromocriptine Ameliorates Aβ1-42-Induced Memory Deficits and Neuroinflammation in Mice. Eur. J. Pharmacol. 2023, 938, 175443. [Google Scholar] [CrossRef] [PubMed]
  65. Lin, J.B.; Harris, J.M.; Baldwin, G.; Goss, D.; Margeta, M.A. Ocular Effects of Rho Kinase (ROCK) Inhibition: A Systematic Review. Eye 2024, 38, 3418–3428. [Google Scholar] [CrossRef]
  66. Montagnoli, T.L.; da Silva, J.S.; Sudo, S.Z.; Santos, A.D.; Gomide, G.F.; de Sá, M.P.L.; Zapata-Sudo, G. ROCK Inhibition as Potential Target for Treatment of Pulmonary Hypertension. Cells 2021, 10, 1648. [Google Scholar] [CrossRef]
  67. Quadir, H.; Hakobyan, K.; Gaddam, M.; Ojinnaka, U.; Ahmed, Z.; Kannan, A.; Mostafa, J.A. Role of Rho-Associated Protein Kinase Inhibition As Therapeutic Strategy for Parkinson’s Disease: Dopaminergic Survival and Enhanced Mitophagy. Cureus 2021, 13, e16973. [Google Scholar] [CrossRef]
  68. Hampel, P.; Römermann, K.; Gailus, B.; Johne, M.; Gericke, B.; Kaczmarek, E.; Löscher, W. Effects of the NKCC1 Inhibitors Bumetanide, Azosemide, and Torasemide Alone or in Combination with Phenobarbital on Seizure Threshold in Epileptic and Nonepileptic Mice. Neuropharmacology 2021, 185, 108449. [Google Scholar] [CrossRef]
  69. Savardi, A.; Patricelli Malizia, A.; De Vivo, M.; Cancedda, L.; Borgogno, M. Preclinical Development of the Na-K-2Cl Co-Transporter-1 (NKCC1) Inhibitor ARN23746 for the Treatment of Neurodevelopmental Disorders. ACS Pharmacol. Transl. Sci. 2023, 6, 1–11. [Google Scholar] [CrossRef]
  70. Gilleen, J.; Nottage, J.; Yakub, F.; Kerins, S.; Valdearenas, L.; Uz, T.; Lahu, G.; Tsai, M.; Ogrinc, F.; Williams, S.C.; et al. The Effects of Roflumilast, a Phosphodiesterase Type-4 Inhibitor, on EEG Biomarkers in Schizophrenia: A Randomised Controlled Trial. J. Psychopharmacol. 2021, 35, 15–22. [Google Scholar] [CrossRef]
  71. Cahill, K.N.; Katz, H.R.; Cui, J.; Lai, J.; Kazani, S.; Crosby-Thompson, A.; Garofalo, D.; Castro, M.; Jarjour, N.; DiMango, E.; et al. KIT Inhibition by Imatinib in Patients with Severe Refractory Asthma. N. Engl. J. Med. 2017, 376, 1911. [Google Scholar] [CrossRef]
  72. Pichavaram, P.; Shawky, N.M.; Hartney, T.J.; Jun, J.Y.; Segar, L. Imatinib Improves Insulin Resistance and Inhibits Injury-Induced Neointimal Hyperplasia in High Fat Diet-Fed Mice. Eur. J. Pharmacol. 2020, 890, 173666. [Google Scholar] [CrossRef] [PubMed]
  73. Kasam, R.K.; Ghandikota, S.; Soundararajan, D.; Reddy, G.B.; Huang, S.K.; Jegga, A.G.; Madala, S.K. Inhibition of Aurora Kinase B Attenuates Fibroblast Activation and Pulmonary Fibrosis. EMBO Mol. Med. 2020, 12, e12131. [Google Scholar] [CrossRef] [PubMed]
  74. Glant, T.T.; Besenyei, T.; Kádár, A.; Kurkó, J.; Tryniszewska, B.; Gál, J.; Soós, G.; Szekanecz, Z.; Hoffmann, G.; Block, J.A.; et al. Differentially Expressed Epigenome Modifiers, Including Aurora Kinases A and B, in Immune Cells in Rheumatoid Arthritis in Humans and Mouse Models. Arthritis Rheum. 2013, 65, 1725–1735. [Google Scholar] [CrossRef]
  75. Tang, H.; Tang, X.; Guo, Z.; Cheng, H.; Zheng, X.; Chen, G.; Huang, H.; Wang, W.; Gao, J.; Sheng, Y.; et al. AURKA Facilitates the Psoriasis-Related Inflammation by Impeding Autophagy-Mediated AIM2 Inflammasome Suppression. Immunol. Lett. 2021, 240, 98–105. [Google Scholar] [CrossRef] [PubMed]
  76. Baghdassarian, H.; Blackstone, S.A.; Clay, O.S.; Philips, R.; Matthiasardottir, B.; Nehrebecky, M.; Hua, V.K.; McVicar, R.; Liu, Y.; Tucker, S.M.; et al. Variant STAT4 and Response to Ruxolitinib in an Autoinflammatory Syndrome. N. Engl. J. Med. 2023, 388, 2241–2252. [Google Scholar] [CrossRef]
  77. Tolomeo, M.; Cascio, A. STAT4 and STAT6, Their Role in Cellular and Humoral Immunity and in Diverse Human Diseases. Int. Rev. Immunol. 2024, 43, 394–418. [Google Scholar] [CrossRef]
  78. Tasaki, S.; Avey, D.R.; Kearns, N.A.; Iatrou, A.; Yu, C.; De Tissera, S.; Vyas, H.; Xu, J.; Flood, D.J.; Rothamel, K.; et al. The YTHDF Proteins Shape the Brain Gene Signatures of Alzheimer’s Disease. bioRxiv 2024. [Google Scholar] [CrossRef]
  79. Long, H.Z.; Cheng, Y.; Zhou, Z.W.; Luo, H.Y.; Wen, D.D.; Gao, L.C. PI3K/AKT Signal Pathway: A Target of Natural Products in the Prevention and Treatment of Alzheimer’s Disease and Parkinson’s Disease. Front. Pharmacol. 2021, 12, 648636. [Google Scholar] [CrossRef]
  80. Lundtoft, C.; Sjöwall, C.; Rantapää-Dahlqvist, S.; Bengtsson, A.A.; Jönsen, A.; Pucholt, P.; Wu, Y.L.; Lundström, E.; Eloranta, M.L.; Gunnarsson, I.; et al. Strong Association of Combined Genetic Deficiencies in the Classical Complement Pathway With Risk of Systemic Lupus Erythematosus and Primary Sjögren’s Syndrome. Arthritis Rheumatol. 2022, 74, 1842–1850. [Google Scholar] [CrossRef]
  81. Warren, J.T.; Cupo, R.R.; Wattanasirakul, P.; Spencer, D.H.; Locke, A.E.; Makaryan, V.; Bolyard, A.A.; Kelley, M.L.; Kingston, N.L.; Shorter, J.; et al. Heterozygous Variants of CLPB Are a Cause of Severe Congenital Neutropenia. Blood 2022, 139, 779. [Google Scholar] [CrossRef]
  82. Cupo, R.R.; Shorter, J. Skd3 (Human Clpb) Is a Potent Mitochondrial Protein Disaggregase That Is Inactivated by 3-Methylglutaconic Aciduria-Linked Mutations. Elife 2020, 9, e55279. [Google Scholar] [CrossRef] [PubMed]
  83. Berth, S.H.; Lloyd, T.E. Disruption of Axonal Transport in Neurodegeneration. J. Clin. Investig. 2023, 133, e168554. [Google Scholar] [CrossRef] [PubMed]
  84. Ge, T.; Yang, J.; Zhou, S.; Wang, Y.; Li, Y.; Tong, X. The Role of the Pentose Phosphate Pathway in Diabetes and Cancer. Front. Endocrinol. 2020, 11, 365. [Google Scholar] [CrossRef] [PubMed]
  85. Gibson, G.E.; Feldman, H.H.; Zhang, S.; Flowers, S.A.; Luchsinger, J.A. Pharmacological Thiamine Levels as a Therapeutic Approach in Alzheimer’s Disease. Front. Med. 2022, 9, 1033272. [Google Scholar] [CrossRef]
  86. Zhou, T.; Yang, H.; Wang, H.; Luo, N.; Xia, Y.; Jiang, X. Association between ACAT1 Rs1044925 and Increased Hypertension Risk in Tongdao Dong. Medicine 2022, 101, e32196. [Google Scholar] [CrossRef]
  87. Alavez-Rubio, J.S.; Martínez-Rodríguez, N.; Escobedo-de-la-Peña, J.; Garrido-Acosta, O.; Juárez-Cedillo, T. Relationship Between Genetic Variants of ACAT1 and APOE with the Susceptibility to Dementia (SADEM Study). Mol. Neurobiol. 2021, 58, 905–912. [Google Scholar] [CrossRef]
  88. Zhang, X.; Lin, Y.; Shi, L.; Zhai, A.; Wu, C.; Zhu, Q.Y. Disulfidptosis-Related Gene SLC3A2: A Novel Prognostic Biomarker in Nasopharyngeal Carcinoma and Head and Neck Squamous Cell Carcinoma. Front. Oncol. 2025, 15, 1451034. [Google Scholar] [CrossRef]
  89. Li, X.; Yang, Y.; Chen, Z. Downregulation of the M6A Reader Protein YTHDC1 Leads to Islet β-Cell Failure and Diabetes. Metabolism 2023, 138, 155339. [Google Scholar] [CrossRef]
  90. Technology—YTH Domain-Containing Family Protein 1 (YTHDF1): A Novel Therapeutic Target for Alzheimer’s Disease. Available online: https://usf.technologypublisher.com/technology/56448 (accessed on 21 April 2025).
  91. Tripathi, V.B.; Baskaran, P.; Shaw, C.E.; Guthrie, S. Tar DNA-Binding Protein-43 (TDP-43) Regulates Axon Growth in Vitro and in Vivo. Neurobiol. Dis. 2014, 65, 25–34. [Google Scholar] [CrossRef]
  92. Dai, G.; Lin, J.; Jiang, Y.; Liu, X.; Chen, P.; Zhang, Y.; Song, Z.; Zhuang, X.; Cong, J.; Li, Y.; et al. Aurora Kinase A Promotes Hepatic Stellate Cell Activation and Liver Fibrosis through the Wnt/β-Catenin Pathway. Front. Oncol. 2024, 14, 1517226. [Google Scholar] [CrossRef]
  93. Important Safety Information for Janus Kinase (JAK) Inhibitors | Therapeutic Goods Administration (TGA). Available online: https://www.tga.gov.au/news/safety-updates/important-safety-information-janus-kinase-jak-inhibitors (accessed on 20 April 2025).
  94. Benucci, M.; Bernardini, P.; Coccia, C.; De Luca, R.; Levani, J.; Economou, A.; Damiani, A.; Russo, E.; Amedei, A.; Guiducci, S.; et al. JAK Inhibitors and Autoimmune Rheumatic Diseases. Autoimmun. Rev. 2023, 22, 103276. [Google Scholar] [CrossRef] [PubMed]
  95. Sarabia, S.; Ranjith, B.; Koppikar, S.; Wijeratne, D.T. Efficacy and Safety of JAK Inhibitors in the Treatment of Psoriasis and Psoriatic Arthritis: A Systematic Review and Meta-Analysis. BMC Rheumatol. 2022, 6, 71. [Google Scholar] [CrossRef] [PubMed]
  96. Chovatiya, R.; Paller, A.S. JAK Inhibitors in the Treatment of Atopic Dermatitis. J. Allergy Clin. Immunol. 2021, 148, 927–940. [Google Scholar] [CrossRef] [PubMed]
  97. Zhou, M.; Shen, Q.; Li, B. JAK Inhibitors: A New Choice for Diabetes Mellitus? Diabetol. Metab. Syndr. 2025, 17, 33. [Google Scholar] [CrossRef]
  98. Furue, K.; Ito, T.; Tanaka, Y.; Hashimoto-Hachiya, A.; Takemura, M.; Murata, M.; Kido-Nakahara, M.; Tsuji, G.; Nakahara, T.; Furue, M. The EGFR-ERK/JNK-CCL20 Pathway in Scratched Keratinocytes May Underpin Koebnerization in Psoriasis Patients. Int. J. Mol. Sci. 2020, 21, 434. [Google Scholar] [CrossRef]
  99. Sangamesh, V.C.; Alagundagi, D.B.; Jayaswamy, P.K.; Kuriakose, N.; Shetty, P. Targeting AnxA2-EGFR Signaling: Hydroxychloroquine as a Therapeutic Strategy for Bleomycin-Induced Pulmonary Fibrosis. Naunyn Schmiedebergs Arch. Pharmacol. 2024, 398, 2015–2026. [Google Scholar] [CrossRef]
  100. Abulizi, A.; Simayi, J.; Nuermaimaiti, M.; Han, M.; Hailati, S.; Talihati, Z.; Maihemuti, N.; Nuer, M.; Khan, N.; Abudurousuli, K.; et al. Quince Extract Resists Atherosclerosis in Rats by Down-Regulating the EGFR/PI3K/Akt/GSK-3β Pathway. Biomed. Pharmacother. 2023, 160, 114330. [Google Scholar] [CrossRef]
  101. Qian, J.; Yu, J.; Zhu, X.; Liang, S. MiR-335 Promotes Corneal Neovascularization by Targeting EGFR. BMC Ophthalmol. 2022, 22, 267. [Google Scholar] [CrossRef]
  102. van der Knaap, M.S.; Bugiani, M.; Abbink, T.E.M. Vanishing White Matter. Handb. Clin. Neurol. 2024, 204, 77–94. [Google Scholar] [CrossRef]
  103. Soliman, G.A.; Schooling, C.M. Causal Association between MTOR-Dependent EIF-4E and EIF-4A Circulating Protein Levels and Type 2 Diabetes: A Mendelian Randomization Study. Sci. Rep. 2020, 10, 15737. [Google Scholar] [CrossRef]
  104. Zhang, T.; Ma, C.; Zhang, Z.; Zhang, H.; Hu, H. NF-ΚB Signaling in Inflammation and Cancer. MedComm 2021, 2, 618–653. [Google Scholar] [CrossRef] [PubMed]
  105. Kaltschmidt, B.; Helweg, L.P.; Greiner, J.F.W.; Kaltschmidt, C. NF-ΚB in Neurodegenerative Diseases: Recent Evidence from Human Genetics. Front. Mol. Neurosci. 2022, 15, 954541. [Google Scholar] [CrossRef] [PubMed]
  106. Nuovo, G.J.; Rice, M.; Zanesi, N.; Sawant, D.; Crilly, C.; Tili, E. The Prevention of Fatal Tauopathy in a Mouse Model of Alzheimer Disease by Blocking BCL2. Appl. Immunohistochem. Mol. Morphol. 2025, 33, 142–151. [Google Scholar] [CrossRef] [PubMed]
  107. Jiang, J.; Zhu, H.; Yang, M.; Yang, B.; Wu, H.; Lu, Q. Topical Administration of a BCL-2 Inhibitor Alleviates Cutaneous Lupus Erythematosus. Int. Immunopharmacol. 2024, 142, 113132. [Google Scholar] [CrossRef]
  108. Snitow, M.E.; Bhansali, R.S.; Klein, P.S. Lithium and Therapeutic Targeting of GSK-3. Cells 2021, 10, 255. [Google Scholar] [CrossRef]
  109. Zhao, J.; Wei, M.; Guo, M.; Wang, M.; Niu, H.; Xu, T.; Zhou, Y. GSK3: A Potential Target and Pending Issues for Treatment of Alzheimer’s Disease. CNS Neurosci. Ther. 2024, 30, e14818. [Google Scholar] [CrossRef]
  110. Bala, A.; Roy, S.; Das, D.; Marturi, V.; Mondal, C.; Patra, S.; Haldar, P.K.; Samajdar, G. Role of Glycogen Synthase Kinase-3 in the Etiology of Type 2 Diabetes Mellitus: A Review. Curr. Diabetes Rev. 2021, 18, 1–7. [Google Scholar] [CrossRef]
  111. Kandar, C.C.; Sen, D.; Maity, A. Anti-Inflammatory Potential of GSK-3 Inhibitors. Curr. Drug Targets 2021, 22, 1464–1476. [Google Scholar] [CrossRef]
  112. Kassir, M.; Karagaiah, P.; Sonthalia, S.; Katsambas, A.; Galadari, H.; Gupta, M.; Lotti, T.; Wollina, U.; Abdelmaksoud, A.; Grabbe, S.; et al. Selective RAR Agonists for Acne Vulgaris: A Narrative Review. J. Cosmet. Dermatol. 2020, 19, 1278–1283. [Google Scholar] [CrossRef]
  113. Nobili, A.; D’Amelio, M.; Viscomi, M.T. Nilotinib: From Animal-Based Studies to Clinical Investigation in Alzheimer’s Disease Patients. Neural Regen. Res. 2023, 18, 803–804. [Google Scholar] [CrossRef]
  114. Myszczyńska, M.A. Uncovering Novel Drug Therapies and Targets for Amyotrophic Lateral Sclerosis (ALS) Using Artificial Intelligence (AI). Ph.D. Thesis, University of Sheffield, Sheffield, UK, 2021. [Google Scholar]
  115. Dong, H.; Li, X.; Cai, M.; Zhang, C.; Mao, W.; Wang, Y.; Xu, Q.; Chen, M.; Wang, L.; Huang, X. Integrated Bioinformatic Analysis Reveals the Underlying Molecular Mechanism of and Potential Drugs for Pulmonary Arterial Hypertension. Aging 2021, 13, 14234. [Google Scholar] [CrossRef] [PubMed]
  116. Kandwal, S.; Fayne, D. Repurposing Drugs for Treatment of SARS-CoV-2 Infection: Computational Design Insights into Mechanisms of Action. J. Biomol. Struct. Dyn. 2022, 40, 1316–1330. [Google Scholar] [CrossRef] [PubMed]
  117. Myers, A.G.; Herzon, S.B.; Wulff, J.E.; Siegrist, R.; Svenda, J.; Zajac, M.A. Synthesis of Avrainvillamide, Stephacidin b, and Analogues Thereof. US Patent US20110166170A1, 7 July 2011. [Google Scholar]
  118. Yin, C.; Huang, G.F.; Sun, X.C.; Guo, Z.; Zhang, J.H. Tozasertib Attenuates Neuronal Apoptosis via DLK/JIP3/MA2K7/JNK Pathway in Early Brain Injury after SAH in Rats. Neuropharmacology 2016, 108, 316–323. [Google Scholar] [CrossRef] [PubMed]
  119. Beca, K. Mediators of Bladder Dysfunction in a Rat Model of Mediators of Bladder Dysfunction in a Rat Model of Cyclophosphamide-Induced Cystitis Cyclophosphamide-Induced Cystitis; The University of Vermont and State Agricultural College: Burlington, VT, USA, 2021. [Google Scholar]
  120. Jiang, M.; Bai, M.; Xu, S.; Wang, T.; Lei, J.; Xu, M.; Huang, S.; Jia, Z.; Zhang, A. Blocking AURKA with MK-5108 Attenuates Renal Fibrosis in Chronic Kidney Disease. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2021, 1867, 166227. [Google Scholar] [CrossRef]
  121. Rehati, A.; Abuduaini, B.; Liang, Z.; Chen, D.; He, F. Identification of Heat Shock Protein Family A Member 5 (HSPA5) Targets Involved in Nonalcoholic Fatty Liver Disease. Genes Immun. 2023, 24, 124–129. [Google Scholar] [CrossRef]
  122. Tian, L.; Li, H.; Xiong, W.; Li, X.; Duan, S.; Yang, C.; Shi, C. Proteomic Alteration in Catalpol Treatment of Alzheimer’s Disease by Regulating HSPA5/ GPX4. Eur. J. Pharmacol. 2025, 987, 177075. [Google Scholar] [CrossRef]
  123. Enogieru, A.B.; Omoruyi, S.I.; Hiss, D.C.; Ekpo, O.E. GRP78/BIP/HSPA5 as a Therapeutic Target in Models of Parkinson’s Disease: A Mini Review. Adv. Pharmacol. Pharm. Sci. 2019, 2019, 2706783. [Google Scholar] [CrossRef]
  124. Xiao, F.; Tan, J.Z.; Xu, X.Y.; Wang, X.F. Increased Levels of HSPA5 in the Serum of Patients with Inflammatory Myopathies—Preliminary Findings. Clin. Rheumatol. 2015, 34, 715–720. [Google Scholar] [CrossRef]
  125. Li, T.; Fu, J.; Cheng, J.; Elfiky, A.A.; Wei, C.; Fu, J. New Progresses on Cell Surface Protein HSPA5/BiP/GRP78 in Cancers and COVID-19. Front. Immunol. 2023, 14, 1166680. [Google Scholar] [CrossRef]
  126. Nguyen, H.T.N.; Salman, R.; Vogel, T.P.; Silva-Carmona, M.; DeGuzman, M.; Guillerman, R.P. Imaging Findings of COPA Syndrome. Pediatr. Radiol. 2023, 53, 844–853. [Google Scholar] [CrossRef]
  127. Cui, X.J.; Zhao, A.G.; Wang, X.L. Correlations of AFAP1, GMDS and PTGFR Gene Polymorphisms with Intra-Ocular Pressure Response to Latanoprost in Patients with Primary Open-Angle Glaucoma. J. Clin. Pharm. Ther. 2017, 42, 87–92. [Google Scholar] [CrossRef] [PubMed]
  128. Kikhai, T.; Agapkina, Y.; Silkina, M.; Prikazchikova, T.; Gottikh, M. The Cellular SFPQ Protein as a Positive Factor in the HIV-1 Integration. Biochimie 2024, 222, 9–17. [Google Scholar] [CrossRef] [PubMed]
  129. Takeuchi, A.; Iida, K.; Tsubota, T.; Hosokawa, M.; Denawa, M.; Brown, J.B.; Ninomiya, K.; Ito, M.; Kimura, H.; Abe, T.; et al. Loss of Sfpq Causes Long-Gene Transcriptopathy in the Brain. Cell Rep. 2018, 23, 1326–1341. [Google Scholar] [CrossRef] [PubMed]
  130. Hosokawa, M.; Takeuchi, A.; Tanihata, J.; Iida, K.; Takeda, S.; Hagiwara, M. Loss of RNA-Binding Protein Sfpq Causes Long-Gene Transcriptopathy in Skeletal Muscle and Severe Muscle Mass Reduction with Metabolic Myopathy. iScience 2019, 13, 229–242. [Google Scholar] [CrossRef]
  131. Neeves, J. Examining the Role of SFPQ and Intron Retention in Physiology and Amyotrophic Lateral Sclerosis. Doctoral Thesis, UCL (University College London), London, UK, 2022. [Google Scholar]
  132. Graustein, A.D.; Misch, E.A.; Musvosvi, M.; Shey, M.; Shah, J.A.; Seshadri, C.; Aguoju, A.; Bowman, K.; Mulenga, H.; Veldsman, A.; et al. Toll-like Receptor Chaperone HSP90B1 and the Immune Response to Mycobacteria. PLoS ONE 2018, 13, e0208940. [Google Scholar] [CrossRef]
  133. Li, L.; Mo, H.; Zhang, J.; Zhou, Y.; Peng, X.; Luo, X. The Role of Heat Shock Protein 90B1 in Patients with Polycystic Ovary Syndrome. PLoS ONE 2016, 11, e0152837. [Google Scholar] [CrossRef]
  134. Liu, Y.; Yang, Y.; Lin, Y.; Wei, B.; Hu, X.; Xu, L.; Zhang, W.; Lu, J. N6-Methyladenosine-Modified CircRNA RERE Modulates Osteoarthritis by Regulating β-Catenin Ubiquitination and Degradation. Cell Prolif. 2023, 56, e13297. [Google Scholar] [CrossRef]
  135. Guo, L.; Liu, F.; Li, H.; Yuan, Y.; Lu, F. Cuproptosis Gene Characterizes the Immune Microenvironment of Diabetic Nephropathy. Transpl. Immunol. 2025, 89, 102175. [Google Scholar] [CrossRef]
  136. Escobar-Pacheco, M.; Luna-Álvarez, M.; Dávila-Ortiz De Montellano, D.; Yescas-Gómez, P.; Ramírez-García, M.Á. Ovarioleukodystrophy Due to EIF2B Genes: Systematic Review and Case Report. Cureus 2024, 16, e64497. [Google Scholar] [CrossRef]
  137. Moon, S.L.; Sonenberg, N.; Parker, R. Neuronal Regulation of EIF2α Function in Health and Neurological Disorders. Trends Mol. Med. 2018, 24, 575–589. [Google Scholar] [CrossRef]
  138. Gui, Y.X.; Zhang, L.S.; Lv, W.; Zhang, W.M.; Zhao, J.J.; Hu, X.Y. NFE2L2 Variations Reduce Antioxidant Response in Patients with Parkinson Disease. Oncotarget 2016, 7, 10756. [Google Scholar] [CrossRef] [PubMed]
  139. Wen, J.J.; Porter, C.; Garg, N.J. Inhibition of NFE2L2-Antioxidant Response Element Pathway by Mitochondrial Reactive Oxygen Species Contributes to Development of Cardiomyopathy and Left Ventricular Dysfunction in Chagas Disease. Antioxid. Redox Signal. 2017, 27, 550–566. [Google Scholar] [CrossRef] [PubMed]
  140. Weerawarna, P.M.; Richardson, T.I. Lyn Kinase Structure, Regulation, and Involvement in Neurodegenerative Diseases: A Mini Review. Kinases Phosphatases 2023, 1, 23–38. [Google Scholar] [CrossRef]
  141. Islam, R.; Choudhary, H.H.; Zhang, F.; Mehta, H.; Yoshida, J.; Thomas, A.J.; Hanafy, K. Microglial TLR4-Lyn Kinase Is a Critical Regulator of Neuroinflammation, Aβ Phagocytosis, Neuronal Damage, and Cell Survival in Alzheimer’s Disease. Sci. Rep. 2025, 15, 11368. [Google Scholar] [CrossRef]
  142. Vormoor, B.; Veal, G.J.; Griffin, M.J.; Boddy, A.V.; Irving, J.; Minto, L.; Case, M.; Banerji, U.; Swales, K.E.; Tall, J.R.; et al. A Phase I/II Trial of AT9283, a Selective Inhibitor of Aurora Kinase in Children with Relapsed or Refractory Acute Leukemia: Challenges to Run Early Phase Clinical Trials for Children with Leukemia. Pediatr. Blood Cancer 2017, 64, e26351. [Google Scholar] [CrossRef]
  143. Hay, A.E.; Murugesan, A.; DiPasquale, A.M.; Kouroukis, T.; Sandhu, I.; Kukreti, V.; Bahlis, N.J.; Lategan, J.; Reece, D.E.; Lyons, J.F.; et al. A Phase II Study of AT9283, an Aurora Kinase Inhibitor, in Patients with Relapased or Refractory Multiple Myloma; NCIC Clinical Trials Group IND.191. Blood 2014, 124, 5734. [Google Scholar] [CrossRef]
  144. Moreno, L.; Marshall, L.V.; Pearson, A.D.J.; Morland, B.; Elliott, M.; Campbell-Hewson, Q.; Makin, G.; Halford, S.E.R.; Acton, G.; Ross, P.; et al. A Phase I Trial of AT9283 (a Selective Inhibitor of Aurora Kinases) in Children and Adolescents with Solid Tumors: A Cancer Research UK Study. Clin. Cancer Res. 2015, 21, 267–273. [Google Scholar] [CrossRef]
  145. Jiang, K.; Bai, L.; Wang, C.; Xiao, X.; Cheng, Z.; Peng, H.; Liu, S. The Aurora Kinase Inhibitor AT9283 Inhibits Burkitt Lymphoma Growth by Regulating Warburg Effect. PeerJ 2023, 11, e16581. [Google Scholar] [CrossRef]
  146. Dawson, M.A.; Curry, J.E.; Barber, K.; Beer, P.A.; Graham, B.; Lyons, J.F.; Richardson, C.J.; Scott, M.A.; Smyth, T.; Squires, M.S.; et al. AT9283, a Potent Inhibitor of the Aurora Kinases and Jak2, Has Therapeutic Potential in Myeloproliferative Disorders. Br. J. Haematol. 2010, 150, 46–57. [Google Scholar] [CrossRef]
  147. Pagan, F.L.; Torres-Yaghi, Y.; Hebron, M.L.; Wilmarth, B.; Turner, R.S.; Matar, S.; Ferrante, D.; Ahn, J.; Moussa, C. Safety, Target Engagement, and Biomarker Effects of Bosutinib in Dementia with Lewy Bodies. Alzheimers Dement. 2022, 8, e12296. [Google Scholar] [CrossRef]
  148. Millar, C.L.; Iloputaife, I.; Baldyga, K.; Norling, A.M.; Boulougoura, A.; Vichos, T.; Tchkonia, T.; Deisinger, A.; Pirtskhalava, T.; Kirkland, J.L.; et al. A Pilot Study of Senolytics to Improve Cognition and Mobility in Older Adults at Risk for Alzheimer’s Disease. EBioMedicine 2025, 113, 105612. [Google Scholar] [CrossRef] [PubMed]
  149. Hayashi, T.; Konishi, I. Cancer Therapy with Decreased SARS-CoV-2 Infection Rates in Cancer Patients. Br. J. Cancer 2022, 126, 521–522. [Google Scholar] [CrossRef] [PubMed]
  150. Yu, T.; Li, W.; Yu, T. Management of Chronic Myelogenous Leukemia with COVID-19 and Hepatitis B. Front. Oncol. 2023, 13, 1217023. [Google Scholar] [CrossRef] [PubMed]
  151. Justice, J.N.; Nambiar, A.M.; Tchkonia, T.; LeBrasseur, N.K.; Pascual, R.; Hashmi, S.K.; Prata, L.; Masternak, M.M.; Kritchevsky, S.B.; Musi, N.; et al. Senolytics in Idiopathic Pulmonary Fibrosis: Results from a First-in-Human, Open-Label, Pilot Study. EBioMedicine 2019, 40, 554–563. [Google Scholar] [CrossRef]
  152. Islam, M.T.; Tuday, E.; Allen, S.; Kim, J.; Trott, D.W.; Holland, W.L.; Donato, A.J.; Lesniewski, L.A. Senolytic Drugs, Dasatinib and Quercetin, Attenuate Adipose Tissue Inflammation, and Ameliorate Metabolic Function in Old Age. Aging Cell 2023, 22, e13767. [Google Scholar] [CrossRef]
  153. Karimi, M.; Bahadoram, M.; Mafakher, L.; Rastegar, M. Impact of Imatinib on Reducing the Painful Crisis in Patients with Sickle Cell Disease. Hematol. Transfus. Cell Ther. 2024, 46, 387–392. [Google Scholar] [CrossRef]
  154. Gitelman, S.E.; Bundy, B.N.; Ferrannini, E.; Lim, N.; Blanchfield, J.L.; DiMeglio, L.A.; Felner, E.I.; Gaglia, J.L.; Gottlieb, P.A.; Long, S.A.; et al. Imatinib Therapy for Patients with Recent-Onset Type 1 Diabetes: A Multicentre, Randomised, Double-Blind, Placebo-Controlled, Phase 2 Trial. Lancet Diabetes Endocrinol. 2021, 9, 502–514. [Google Scholar] [CrossRef]
  155. Wu, X.; Li, W.; Luo, Z.; Chen, Y. A Comprehensive Comparison of the Safety and Efficacy of Drugs in the Treatment of Idiopathic Pulmonary Fibrosis: A Network Meta-Analysis Based on Randomized Controlled Trials. BMC Pulm. Med. 2024, 24, 58. [Google Scholar] [CrossRef]
  156. Wahlgren, N.; Thorén, M.; Höjeberg, B.; Käll, T.-B.; Laska, A.-C.; Sjöstrand, C.; Höijer, J.; Almqvist, H.; Holmin, S.; Lilja, A.; et al. Randomized Assessment of Imatinib in Patients with Acute Ischaemic Stroke Treated with Intravenous Thrombolysis. J. Intern. Med. 2017, 281, 273–283. [Google Scholar] [CrossRef]
  157. Study Details | Effect of Imatinib in Advance Liver Fibrosis Patients | ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/study/NCT05224128 (accessed on 1 June 2025).
  158. Turner, R.S.; Hebron, M.L.; Lawler, A.; Mundel, E.E.; Yusuf, N.; Starr, J.N.; Anjum, M.; Pagan, F.; Torres-Yaghi, Y.; Shi, W.; et al. Nilotinib Effects on Safety, Tolerability, and Biomarkers in Alzheimer’s Disease. Ann. Neurol. 2020, 88, 183–194. [Google Scholar] [CrossRef]
  159. Jane, E.P.; Premkumar, D.R.; Rajasundaram, D.; Thambireddy, S.; Reslink, M.C.; Agnihotri, S.; Pollack, I.F. Reversing Tozasertib Resistance in Glioma through Inhibition of Pyruvate Dehydrogenase Kinases. Mol. Oncol. 2021, 16, 219. [Google Scholar] [CrossRef] [PubMed]
  160. Wang, Q.; Liu, W.; Zhou, H.; Lai, W.; Hu, C.; Dai, Y.; Li, G.; Zhang, R.; Zhao, Y. Tozasertib Activates Anti-Tumor Immunity through Decreasing Regulatory T Cells in Melanoma. Neoplasia 2024, 48, 100966. [Google Scholar] [CrossRef] [PubMed]
  161. Zhang, L.; Ji, K.; Sun, Y.; Hou, Y.; Chen, J. Aurora Kinase Inhibitor Tozasertib Suppresses Mast Cell Activation in Vitro and in Vivo. Br. J. Pharmacol. 2020, 177, 2848–2859. [Google Scholar] [CrossRef] [PubMed]
  162. Uniyal, A.; Shantanu, P.A.; Vaidya, S.; Belinskaia, D.A.; Shestakova, N.N.; Kumar, R.; Singh, S.; Tiwari, V. Tozasertib Attenuates Neuropathic Pain by Interfering with Aurora Kinase and KIF11 Mediated Nociception. ACS Chem. Neurosci. 2021, 12, 1948–1960. [Google Scholar] [CrossRef]
  163. Liu, J.; Zhang, Y.; Huang, H.; Lei, X.; Tang, G.; Cao, X.; Peng, J. Recent Advances in Bcr-Abl Tyrosine Kinase Inhibitors for Overriding T315I Mutation. Chem. Biol. Drug Des. 2021, 97, 649–664. [Google Scholar] [CrossRef]
  164. Wen, Y.; Jing, Y.; Yang, L.; Kang, D.; Jiang, P.; Li, N.; Cheng, J.; Li, J.; Li, X.; Peng, Z.; et al. The Regulators of BCR Signaling during B Cell Activation. Blood Sci. 2019, 1, 119–129. [Google Scholar] [CrossRef]
  165. Blakes, A.J.M.; Gaul, E.; Lam, W.; Shannon, N.; Knapp, K.M.; Bicknell, L.S.; Jackson, M.R.; Wade, E.M.; Robertson, S.; White, S.M.; et al. Pathogenic Variants Causing ABL1 Malformation Syndrome Cluster in a Myristoyl-Binding Pocket and Increase Tyrosine Kinase Activity. Eur. J. Hum. Genet. 2021, 29, 593–603. [Google Scholar] [CrossRef]
  166. Gutiérrez, D.; Chandía-Cristi, A.; Yáñez, M.; Zanlungo, S.; Álvarez, A. C-Abl Kinase at the Crossroads of Healthy Synaptic Remodeling and Synaptic Dysfunction in Neurodegenerative Diseases. Neural Regen. Res. 2023, 18, 237–243. [Google Scholar] [CrossRef]
  167. Rajkumar, V.S.; Shiwen, X.; Bostrom, M.; Leoni, P.; Muddle, J.; Ivarsson, M.; Gerdin, B.; Denton, C.P.; Bou-Gharios, G.; Black, C.M.; et al. Platelet-Derived Growth Factor-β Receptor Activation Is Essential for Fibroblast and Pericyte Recruitment during Cutaneous Wound Healing. Am. J. Pathol. 2006, 169, 2254–2265. [Google Scholar] [CrossRef]
  168. Tsai, M.; Valent, P.; Galli, S.J. KIT as a Master Regulator of the Mast Cell Lineage. J. Allergy Clin. Immunol. 2022, 149, 1845–1854. [Google Scholar] [CrossRef]
  169. Blair, L.J.; Sabbagh, J.J.; Dickey, C.A. Targeting Hsp90 and Its Co-Chaperones to Treat Alzheimer’s Disease. Expert Opin. Ther. Targets 2014, 18, 1219. [Google Scholar] [CrossRef] [PubMed]
  170. Guo, F.; Liu, X.; Cai, H.; Le, W. Autophagy in Neurodegenerative Diseases: Pathogenesis and Therapy. Brain Pathol. 2017, 28, 3. [Google Scholar] [CrossRef] [PubMed]
  171. Shelton, L.B.; Baker, J.D.; Zheng, D.; Sullivan, L.E.; Solanki, P.K.; Webster, J.M.; Sun, Z.; Sabbagh, J.J.; Nordhues, B.A.; Koren, J.; et al. Hsp90 Activator Aha1 Drives Production of Pathological Tau Aggregates. Proc. Natl. Acad. Sci. USA 2017, 114, 9707–9712. [Google Scholar] [CrossRef] [PubMed]
  172. Ou, J.R.; Tan, M.S.; Xie, A.M.; Yu, J.T.; Tan, L. Heat Shock Protein 90 in Alzheimer’s Disease. Biomed. Res. Int. 2014, 2014, 796869. [Google Scholar] [CrossRef]
  173. Zeng, Z.; Liao, C.; Yu, L. Molecules for COVID-19 Treatment. Chin. Chem. Lett. 2024, 35, 109349. [Google Scholar] [CrossRef]
  174. Xu, W.; Huang, Y.; Zhou, R. NLRP3 Inflammasome in Neuroinflammation and Central Nervous System Diseases. Cell. Mol. Immunol. 2025, 22, 341–355. [Google Scholar] [CrossRef]
  175. Li, S.; He, J.; Zhang, X.; Cai, Y.; Liu, J.; Nie, X.; Shi, L. Cardiovascular Adverse Events in Chronic Myeloid Leukemia Patients Treated with Nilotinib or Imatinib: A Systematic Review, Meta-Analysis and Integrative Bioinformatics Analysis. Front. Cardiovasc. Med. 2022, 9, 966182. [Google Scholar] [CrossRef]
  176. Kim, K.W.; Shinagare, A.B.; Krajewski, K.M.; Pyo, J.; Tirumani, S.H.; Jagannathan, J.P.; Ramaiya, N.H. Fluid Retention Associated with Imatinib Treatment in Patients with Gastrointestinal Stromal Tumor: Quantitative Radiologic Assessment and Implications for Management. Korean J. Radiol. 2015, 16, 304. [Google Scholar] [CrossRef]
  177. Doncheva, N.T.; Morris, J.H.; Gorodkin, J.; Jensen, L.J. Cytoscape StringApp: Network Analysis and Visualization of Proteomics Data. J. Proteome Res. 2019, 18, 623–632. [Google Scholar] [CrossRef]
  178. Alvascience. AlvaDesc (Software for Molecular Descriptors Calculation) Version 1.0.18. 2020. Available online: https://www.alvascience.com (accessed on 7 April 2025).
  179. RDKit. Available online: https://www.rdkit.org/ (accessed on 2 May 2025).
  180. Medical Subject Headings RDF. Available online: https://id.nlm.nih.gov/mesh/ (accessed on 1 May 2025).
Figure 1. Integrative informatics workflow to study the polypharmacology of BCR-ABL inhibitors.
Figure 1. Integrative informatics workflow to study the polypharmacology of BCR-ABL inhibitors.
Pharmaceuticals 18 00936 g001
Figure 2. Cheminformatics analysis of BCR-ABL inhibitors. (A) Heatmap for the distance matrix using Tanimoto coefficients and Morgan fingerprints. (B) Heatmap for the distance matrix using Euclidean distances and 2D alvaDesc molecular descriptors. (C) Principal component analysis of seven BCR-ABL inhibitors using 2D alvaDesc molecular descriptors and Euclidean distances. The color gradient reflects DLS_cons—the consensus drug-likeness score—providing an additional interpretive layer grounded in the pharmacokinetic and physicochemical profile of the molecules. The plot reveals the principal component scores for PC1 (50.79%) and PC2 (19.12%), which together explain ~70% of the total variance, suggesting good dimensional reduction and data representation fidelity.
Figure 2. Cheminformatics analysis of BCR-ABL inhibitors. (A) Heatmap for the distance matrix using Tanimoto coefficients and Morgan fingerprints. (B) Heatmap for the distance matrix using Euclidean distances and 2D alvaDesc molecular descriptors. (C) Principal component analysis of seven BCR-ABL inhibitors using 2D alvaDesc molecular descriptors and Euclidean distances. The color gradient reflects DLS_cons—the consensus drug-likeness score—providing an additional interpretive layer grounded in the pharmacokinetic and physicochemical profile of the molecules. The plot reveals the principal component scores for PC1 (50.79%) and PC2 (19.12%), which together explain ~70% of the total variance, suggesting good dimensional reduction and data representation fidelity.
Pharmaceuticals 18 00936 g002aPharmaceuticals 18 00936 g002b
Figure 3. Structural and transcriptional signatures underlying the polypharmacology of BCR-ABL inhibitors. (A) Chemical structures, target profiles, and mechanisms of action of BCR-ABL inhibitors. (B) Cell-line-specific transcriptional responses to BCR-ABL inhibitors. Thick black bars signify transcriptional activity scores (TASs) greater than or equal to 0.5; thinner black bars indicate scores less than 0.5. Red lines (chords) denote similar positive connectivity scores between cell lines, which range from 80 to 100 (pale to intense color according to the score). Chords are shown only when TASs are ≥0.5, indicating strong or biologically relevant transcriptional activity. The absence of red chords either means that the compound’s TAS is very low, or that no data are available. Source: CMap database.
Figure 3. Structural and transcriptional signatures underlying the polypharmacology of BCR-ABL inhibitors. (A) Chemical structures, target profiles, and mechanisms of action of BCR-ABL inhibitors. (B) Cell-line-specific transcriptional responses to BCR-ABL inhibitors. Thick black bars signify transcriptional activity scores (TASs) greater than or equal to 0.5; thinner black bars indicate scores less than 0.5. Red lines (chords) denote similar positive connectivity scores between cell lines, which range from 80 to 100 (pale to intense color according to the score). Chords are shown only when TASs are ≥0.5, indicating strong or biologically relevant transcriptional activity. The absence of red chords either means that the compound’s TAS is very low, or that no data are available. Source: CMap database.
Pharmaceuticals 18 00936 g003aPharmaceuticals 18 00936 g003b
Figure 4. CMap connections with BCR-ABL inhibitors across perturbagen class, compound, and genes. (A) Connections with seven BCR-ABL inhibitors. (B) Connections with imatinib. (C) Connections with nilotinib.
Figure 4. CMap connections with BCR-ABL inhibitors across perturbagen class, compound, and genes. (A) Connections with seven BCR-ABL inhibitors. (B) Connections with imatinib. (C) Connections with nilotinib.
Pharmaceuticals 18 00936 g004aPharmaceuticals 18 00936 g004b
Figure 5. BCR-ABL protein–protein interaction (PPI) network and pathway enrichment. The network was generated using BCR-ABL as the input. Nodes represent proteins directly interacting with BCR-ABL, and edges indicate known or predicted protein–protein associations. Node segments are color-coded based on the top 10 enriched KEGG pathways identified through over-representation analysis in Cytoscape. White colors indicate that the corresponding pathways are not enriched in that node. A totally grey node indicate that this node was not enriched in any of the top pathways shown in this figure.
Figure 5. BCR-ABL protein–protein interaction (PPI) network and pathway enrichment. The network was generated using BCR-ABL as the input. Nodes represent proteins directly interacting with BCR-ABL, and edges indicate known or predicted protein–protein associations. Node segments are color-coded based on the top 10 enriched KEGG pathways identified through over-representation analysis in Cytoscape. White colors indicate that the corresponding pathways are not enriched in that node. A totally grey node indicate that this node was not enriched in any of the top pathways shown in this figure.
Pharmaceuticals 18 00936 g005
Figure 6. Comparative PPI networks and enriched pathways of imatinib’s and nilotinib’s positive CMAP connections, revealing divergent polypharmacological profiles. Networks were created using all positive gene connections with CMap scores ≥ 90.00.
Figure 6. Comparative PPI networks and enriched pathways of imatinib’s and nilotinib’s positive CMAP connections, revealing divergent polypharmacological profiles. Networks were created using all positive gene connections with CMap scores ≥ 90.00.
Pharmaceuticals 18 00936 g006
Table 1. Top CMap positive-scoring perturbagen connections to BCR-ABL inhibitors and their known non-cancer indications.
Table 1. Top CMap positive-scoring perturbagen connections to BCR-ABL inhibitors and their known non-cancer indications.
#PerturbagenTypeScoreNon-Cancer Indications and Validity
1Aurora kinase inhibitorClass99.46Alzheimer’s Disease (Preclinical) [13]; Pulmonary Fibrosis (Preclinical) [14]; Inflammatory Bowel Diseases (Preclinical but weak) [15]; Arthritis, Psoriasis (Preclinical) [16]; Arthritis, Rheumatoid (In silico) [17].
2PDGFR\KIT inhibitorClass99.34Alzheimer’s Disease (Preclinical) [18]; Atherosclerosis (Preclinical) [19]; Gastrointestinal Diseases (Preclinical) [18]; Liver Cirrhosis, Experimental (Preclinical) [18] Pulmonary Fibrosis (Preclinical) [20]; Wound Healing (Preclinical) [18].
3EGFR inhibitorClass99.02Pulmonary Fibrosis (Preclinical) [21]; Psoriasis (Preclinical) [22].
4VEGFR inhibitor Class98.73Eye Diseases (Clinical/Approved) [23]; Arthritis, Rheumatoid (Preclinical) [24]; Hypertension, Portal [25]; Psoriasis (Preclinical) [26].
5RAF inhibitorClass98.67Arthritis, Rheumatoid (Preclinical) [27]; Lung Injury and Pulmonary Fibrosis (Preclinical) [28].
6Afatinib Compound99.44Skin Diseases (Preclinical) [29].
7HG-6-64-01Compound99.40Heart failure (Preclinical) [30].
8CarnertinibCompound99.26Liver Cirrhosis, Experimental (Preclinical) [31].
9Tetrindole Compound99.19Depression (Preclinical/Early clinical) [32].
10WZ-4-145Compound99.19Unknown.
11SU-11652Compound99.12Unknown. Multi-targeted TKI.
12MibefradilCompound98.98Pain, Neuropathic (Preclinical) [33].
13NVP-TAE684Compound99.98Neuroprotection; Alzheimer Disease (Preclinical) [34].
14CediranibCompound98.94Liver Cirrhosis, Experimental (Preclinical) [25].
15AZD-7762Compound98.93Osteoporosis (Preclinical) [35].
16TERF1Gene (kd)98.77Aging (Preclinical) [36]; Alzheimer’s Disease (Preclinical) [37]; Infertility, Male (Preclinical) [38].
17C9ORF96Gene (kd)98.62Unknown.
18UGCGGene (oe)98.10Gaucher Disease (Clinical) [39]; Keloid (Preclinical) [40]; Myocardial Fibrosis (Preclinical) [41]; Vascular Malformations (Preclinical) [42]; Parkinson’s Disease (Preclinical) [43], Depressive Disorder (Preclinical) [43].
19AKT3Gene (kd)97.47Brain Malformations (Clinical) [44]; Autoimmunity (Preclinical) [45]; Cognitive Dysfunction (Preclinical) [46]; Hemimegaloencephaly (Preclinical) [47]; Wound Healing (Preclinical) [48].
20ZNF449Gene (kd)97.60Chondrogenesis/Cartilage (Preclinical) [49].
21KRASGene (kd)97.50Kidney Fibrosis (Preclinical) [50].
22FABP4Gene (oe)97.32Diabetes Mellitus (Clinical) [51]; Atherosclerosis (Preclinical) [52]; Kidney Stones (Preclinical) [53]; Obesity (Preclinical) [54].
23PTK2Gene (kd)97.22Alzheimer’s Disease (Preclinical) [55].
24AK3Gene (kd)97.14Alzheimer’s Disease (In silico) [56].
25MRP1L18Gene (oe) 97.12Asthma (Preclinical) [57].
The table lists the top-scoring compounds, drug classes, and gene perturbations based on transcriptomic similarity scores. Entries include their known cancer and non-cancer indications, highlighting potential polypharmacology and repurposing opportunities. Literature-based evidence is provided for non-cancer indications. Non-cancer indications not identified in the literature or publicly available sources, as of 28 April 2025, are denoted as ‘None found’. Validity levels are described in the Methods section.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hajjo, R.; Sabbah, D.A.; Alhaded, R.; Alquabe’h, A.; Bardaweel, S.K. Computational Insights into the Polypharmacological Landscape of BCR-ABL Inhibitors: Emphasis on Imatinib and Nilotinib. Pharmaceuticals 2025, 18, 936. https://doi.org/10.3390/ph18070936

AMA Style

Hajjo R, Sabbah DA, Alhaded R, Alquabe’h A, Bardaweel SK. Computational Insights into the Polypharmacological Landscape of BCR-ABL Inhibitors: Emphasis on Imatinib and Nilotinib. Pharmaceuticals. 2025; 18(7):936. https://doi.org/10.3390/ph18070936

Chicago/Turabian Style

Hajjo, Rima, Dima A. Sabbah, Raghad Alhaded, Aye Alquabe’h, and Sanaa K. Bardaweel. 2025. "Computational Insights into the Polypharmacological Landscape of BCR-ABL Inhibitors: Emphasis on Imatinib and Nilotinib" Pharmaceuticals 18, no. 7: 936. https://doi.org/10.3390/ph18070936

APA Style

Hajjo, R., Sabbah, D. A., Alhaded, R., Alquabe’h, A., & Bardaweel, S. K. (2025). Computational Insights into the Polypharmacological Landscape of BCR-ABL Inhibitors: Emphasis on Imatinib and Nilotinib. Pharmaceuticals, 18(7), 936. https://doi.org/10.3390/ph18070936

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop