Next Article in Journal
Structural Activity Relationship Analysis of New Diphenyl PFI-3 Analogues Targeting for the Treatment of Glioblastoma
Previous Article in Journal
Advancing Neuropharmacology and Neurodegenerative Disease Therapy: Bridging Gaps and Paving New Pathways
Previous Article in Special Issue
Kynurenines and Inflammation: A Remarkable Axis for Multiple Sclerosis Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Quinoline Quest: Kynurenic Acid Strategies for Next-Generation Therapeutics via Rational Drug Design

by
Masaru Tanaka
1,*,†,
István Szatmári
2 and
László Vécsei
1,3,*,†
1
Danube Neuroscience Research Laboratory, HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
2
Institute of Pharmaceutical Chemistry and HUN-REN–SZTE Stereochemistry Research Group, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
3
Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2025, 18(5), 607; https://doi.org/10.3390/ph18050607
Submission received: 31 March 2025 / Revised: 17 April 2025 / Accepted: 19 April 2025 / Published: 22 April 2025
(This article belongs to the Special Issue Kynurenine Pathway: A Novel Therapeutic Opportunity)

Abstract

:
Background: Quinoline-derived metabolites exhibit notable chemical complexity. What causes minor structural alterations to induce significant changes in disease outcomes? Historically, eclipsed by more straightforward scaffolds, these chemicals serve as a dynamic hub in tryptophan metabolism, linking immunomodulation, excitotoxicity, and cancer. However, many of these compounds struggle to cross the blood–brain barrier, and we still do not fully understand how certain structural changes affect their bioavailability or off-target effects. Thus, contemporary research highlights halogenation, esterification, and computational modeling to enhance structure–activity relationships. Summary: This narrative review emphasizes the integration of rational drug design, multi-target ligands, and prodrug methods in enhancing quinoline scaffolds. We explore each molecule’s therapeutic promise, refine each scaffold’s design, and develop each derivative to maximize clinical utility. Translating these laboratory findings into clinical practice, however, remains a formidable challenge. Conclusions: Through the synthesis of findings regarding NMDA receptor antagonism, improved oral bioavailability, and reduced metabolic instability, we demonstrate how single-site changes might modulate excitotoxicity and immunological signaling. Advancing quinoline-based medicines will yield significant advancements in neurology, psychiatry, and oncology. This enlarged framework fosters collaborative discovery, engages various audiences, and advances the field towards next-generation disease-modifying therapies. Robust preclinical validation, patient classification, and comprehensive toxicity evaluations are crucial stages for achieving these extensive endeavors and fostering future therapeutic discoveries globally.

Graphical Abstract

1. Introduction

Tryptophan (Trp) undergoes a fascinating series of biochemical transformations through kynurenine (KYN) metabolism that leads to the production of various neuroactive and immunomodulatory compounds [1]. At the heart of this metabolic arm lies a cascade of enzymes that generates an array of quinoline-based molecules, each with distinct and sometimes opposing effects on cellular and molecular processes (Figure 1) [2]. Among the most scrutinized products are kynurenic acid (KYNA) and quinolinic acid (QUIN), which play critical roles in modulating neurotransmission, safeguarding neurons, and orchestrating immune responses [3]. Under normal conditions, these molecules help maintain the delicate balance that protects and regulates neural function [4]. However, an overabundance or misregulation of these compounds can fuel diverse pathologies, spanning neurodegenerative conditions, psychiatric disorders, and even certain cancers [5]. Remarkably, these metabolites appear across many species and have preserved their functional importance for millions of years [6]. Their enduring presence suggests a fundamental role in maintaining physiological homeostasis, yet they can also become potent drivers of disease when their levels shift [7]. This duality underscores the importance of understanding both the biochemical nuances and the broader biological contexts in which these quinoline-based substances operate [3,8].
The quinoline skeleton lies at the heart of a wide variety of biologically active molecules, encompassing endogenous metabolites, such as KYNA and QUIN, as well as numerous synthetic derivatives found in pharmaceuticals and research tools [9]. Defined by a fused ring structure that merges a benzene ring with a pyridine ring, quinoline offers countless opportunities for chemical substitutions [10]. Small modifications—such as adding carboxyl or hydroxyl groups—can significantly change a molecule’s behavior. These changes affect solubility, stability, and even affinity for specific protein targets [11]. Halogenation, for instance, may boost lipophilicity and enhance central nervous system penetration, while the presence or absence of an acidic group can shape receptor-binding preferences [12]. In many cases, these structural nuances determine whether a particular quinoline derivative acts as a neuroprotective agent or exacerbates excitotoxic processes through receptors, like N-methyl-D-aspartate (NMDA) or G protein-coupled receptor 35 (GPR35) [13]. Endogenous metabolites derived from the Trp degradation route typically exhibit distinct features that help them coordinate intricate cellular signaling, whereas synthetic derivatives, exemplified by quinolone antibiotics, have been optimized for pathogen clearance and bioavailability [14]. Despite their diverse origins, each compound features the versatile quinoline core, underscoring the need for structural precision in creating targeted therapies [15].
A hallmark of these Trp-derived quinoline compounds is their profound influence on the delicate balance between neuroprotection and neurotoxicity [16]. Their functions are finely tuned by their relative concentrations, contextual factors, and micro-environment, shaping their cumulative effects [17]. Beyond their influence in the central nervous system, these metabolites also bridge metabolic and immune pathways, as illustrated by enzymes like indoleamine 2,3-dioxygenases (IDOs) and tryptophan 2,3-dioxygenase (TDO) [18]. When this biochemical system malfunctions, it can drive inflammation and alter disease pathways in autoimmunity and cancer [19]. Emerging evidence further suggests crosstalk with the aryl hydrocarbon receptor, hinting at broader physiological roles that extend beyond neuronal circuits [20]. Such multifaceted involvement underscores the need to appreciate how these molecules orchestrate, or disrupt, redox equilibrium, synaptic communication, and immune surveillance in tandem [21]. By influencing both protective and harmful pathways, these compounds form a paradox where even small imbalances can lead to disease [22,23]. Investigating this interplay is vital for deciphering the biology behind complex disorders, as well as for pinpointing novel therapeutic entry points grounded in the chemistry of quinoline-based metabolites.
In recent years, therapeutic strategies targeting Trp metabolism have drawn increased attention, supported by encouraging preclinical findings and a rise in clinical trials [24,25]. For instance, inhibitors of IDOs showed early promise in oncology by dampening immunosuppressive pathways and restoring anti-tumor responses [26]. However, some late-stage trials yielded disappointingly modest outcomes, illustrating the complexity of translating metabolic interventions into measurable clinical benefits [27]. Another approach uses KYNA analogs—such as 7-CKA—to help alleviate schizophrenia-like symptoms by modulating glutamate receptors [28]. Although these discoveries underscore the promise of quinoline-based therapeutics, many challenges still lie ahead [29]. Achieving adequate penetration across the blood–brain barrier (BBB) can be particularly daunting, as can mitigating off-target effects that may compromise patient safety [30]. Beyond endogenously derived agents, certain quinoline derivatives traditionally used for other indications, such as quinine and related molecules, are now being explored for their neuroprotective or anti-inflammatory properties [31]. Repurposing these compounds also accelerates clinical testing by leveraging existing safety and pharmacokinetic data [32]. Yet, optimizing dosage, delivery methods, and specificity remains a priority [33]. Moving forward, interdisciplinary teams can combine synthetic chemistry, pharmacology, and clinical insights to refine these agents and unlock their untapped therapeutic potential [34].
Despite expanding research on quinoline-containing molecules, critical questions remain unresolved [35]. One pivotal gap lies in our limited understanding of how specific structural modifications translate into particular biological effects, often termed structure–activity relationships [36]. Such insights are crucial for guiding rational drug design and for predicting how a new derivative might behave in living systems. Another hurdle is the fragmented nature of pharmacokinetic and safety data. Often, these findings come from small, isolated studies rather than comprehensive analyses [37]. This piecemeal approach complicates efforts to compare efficacy and toxicity across different quinoline scaffolds [37]. Furthermore, there is a noticeable lack of head-to-head evaluations between endogenous Trp-derived metabolites and synthetic or exogenous analogs, preventing a clear assessment of relative benefits and risks. Building on these concerns, the objectives of this review are threefold. The first is to systematically evaluate the wealth of endogenous and synthetic quinoline-based compounds for their therapeutic potential in various disease contexts. The second is to synthesize current knowledge about structure–activity relationships with the explicit goal of identifying a candidate inspired by KYNA that might serve as a leading drug template. Lastly, we aim to propose innovative strategies to address translational challenges, including targeted prodrug design and approaches that can engage multiple biological targets simultaneously.

2. Unveiling Endogenous Quinolines in the Kynurenine (KYN) Pathway: A Gateway to Kynurenic Acid (KYNA)-Driven Neuroprotection and Rational Drug Design

The metabolism of Trp produces a striking array of bioactive compounds [38]. KYNA, QUIN, nicotinic acid (NA), picolinic acid, xanthurenic acid (XA), and quinaldic acid each exert profound effects on human physiology [39]. Recent findings indicate that these metabolites serve as delicate modulators capable of shifting the balance between neuroprotection and neurotoxicity [40,41]. Recent studies indicate that KYNA’s protective profile extends to antioxidant mechanisms, beyond its well-characterized role in blocking excitotoxic glutamate receptors [42]. By directly scavenging reactive oxygen species and enhancing enzymatic antioxidant systems—such as superoxide dismutase and glutathione peroxidase—KYNA can mitigate oxidative stress in both neuronal and peripheral tissues [43]. These antioxidant properties are especially relevant in neurodegenerative disorders where imbalances in redox homeostasis fuel disease progression [44]. Integrating these newer findings with KYNA’s established immunomodulatory and neuroprotective roles further underscores its multifaceted potential as a therapeutic agent [45]. KYNA, often described as a safeguard against excitotoxicity, dampens glutamatergic signaling [46]. In contrast, QUIN can escalate excitatory activity to dangerous extremes, contributing to neuronal injury under pathological conditions [47]. NA supports redox homeostasis, while picolinic acid and XA coordinate metal ions, subtly influencing cellular stability [48]. Quinaldic acid, a relatively understudied molecule in this network, has roles that are only now beginning to emerge [49]. These metabolites do not function in isolation. They respond to the surrounding microenvironment and reshape it in ways that may ultimately define the trajectory of health and disease [50] (Table 1).

2.1. Structural and Functional Synergy

The quinoline core appears in a range of Trp-derived metabolites, revealing an intriguing contrast in their biological effects [2]. KYNA and QUIN emerge from the same metabolic pathway, yet they are traditionally considered to operate on opposite ends of the spectrum [75]. One protects neurons by dampening excessive excitatory signaling; the other drives excitotoxic damage often linked to inflammation [96]. This shared structural backbone, paired with such starkly different outcomes, suggests a delicate evolutionary balance shaped by minor molecular adjustments [2]. Consider how slight modifications to substituent groups on the quinoline ring can shift a molecule’s interaction with receptors, ion channels, or transporters [97]. Even fluctuations in pH or enzyme activity are enough to sway the system toward protection or harm [98]. There is more: metal ions and cofactors add another layer of control, either amplifying or suppressing activity [99]. The challenge now lies in identifying how to steer this balance [100]. Could targeted changes to the quinoline scaffold enhance KYNA’s protective roles or curb the destructive potential of QUIN [101]? Understanding these subtle structural cues opens the door to designing molecules with more favorable effects on neural health.
Small structural modifications within quinoline-based metabolites can significantly influence their biological activity and stability [102]. Altering the position of a hydroxyl group, as observed in XA, affects the hydrogen-bonding capacity and lipophilicity, which may shift receptor affinity and metabolic processing [103]. These effects are particularly relevant when examining interactions with glutamate receptors, such as NMDA, where even minor variations can determine whether a metabolite contributes to neuroprotection or excitotoxicity [104]. Similar patterns emerge with GPR35, a receptor that responds differently based on the specific substituents attached to the quinoline scaffold [105]. Variations in these groups regulate receptor engagement and downstream signaling pathways, shaping distinct physiological outcomes [98]. Alongside receptor interactions, metabolic stability is also sensitive to changes in ionization states and enzyme susceptibility, both of which are closely tied to the positioning of carboxyl, hydroxyl, or amino groups [106]. Mapping these structural features across endogenous metabolites provides insights into how they influence pathological processes [107,108]. Examining the relationship between the substitution patterns, receptor selectivity, and metabolic half-life may contribute to the design of compounds that adjust the activity of quinoline derivatives in targeted therapeutic contexts.

2.2. Key Gaps in Translation

Despite their diverse biological activities, translating endogenous quinoline-based compounds into therapeutic interventions faces significant hurdles [109]. A chief concern is poor bioavailability, particularly evident in KYNA [110]. Its polar carboxylate group hinders passive diffusion across biological membranes and limits the ability to reach target sites in sufficient concentrations [111]. On the other hand, picolinic acid, while exhibiting promising immunomodulatory and metal-chelating activities, undergoes rapid metabolism, leading to a short half-life and unstable plasma levels [112]. These kinetic and physicochemical drawbacks are further complicated by the complexity of enzymatic pathways involved, where small fluctuations in metabolic rates can greatly alter the overall balance of protective versus pathogenic metabolites [113]. Such pharmacokinetic challenges underscore the difficulty of achieving consistent dosing in clinical settings [114]. As highlighted in recent efforts to refine KYN system monitoring, relying solely on static concentrations or simplistic ratios may be insufficient. A more nuanced, integrative biomarker framework is needed to capture the dynamic regulation of tryptophan metabolism and guide precision medicine applications in neuropsychiatric and neurodegenerative disorders [115]. Moreover, existing animal models do not always recapitulate human physiology, complicating the interpretation of efficacy and toxicity data [62]. Without efficient strategies to enhance bioavailability—such as prodrug approaches, targeted delivery systems, or structural modifications to increase lipophilicity—these compounds may struggle to meet clinical requirements [116,117]. As research deepens our understanding of how structural and functional attributes influence pharmacokinetics, efforts to overcome these limitations can be directed more precisely, potentially unlocking the therapeutic promise that many of these metabolites have long suggested.
Translating endogenous quinoline compounds into real-world treatments often involves navigating paradoxical biological roles, as exemplified by NA [118,119]. Widely recognized for its capacity to modulate lipid metabolism and improve cardiovascular profiles, NA also harbors less-appreciated functions in inflammatory pathways [120]. This duality creates both opportunity and challenge: on one hand, its impact on lipid-lowering has made it a mainstay in certain therapeutic regimens; on the other hand, emerging studies suggest that NA can engage molecular cascades involved in neuroinflammation, potentially influencing conditions like neurodegenerative diseases [121,122]. Such conflicting actions underscore a larger theme in quinoline-related research, where a single molecule may oscillate between protective and pathogenic effects depending on the context and concentration [123]. The incomplete mapping of these dual functionalities hampers our ability to refine dosing strategies or identify the specific patient populations who might benefit the most [98]. Additionally, the diverse array of downstream metabolites and receptors further complicates the picture, making it difficult to anticipate long-term outcomes [124]. As a result, bridging the gap from bench to bedside requires a thorough understanding of how NA’s manifold roles intersect. Only by disentangling these intricate mechanisms can researchers devise targeted interventions that harness the therapeutic benefits of NA while minimizing unanticipated consequences.
A major obstacle in harnessing the therapeutic potential of endogenous quinoline compounds lies in their limited tissue penetration and rapid clearance [56]. KYNA, for example, is known to possess neuroprotective properties but struggles to traverse biological barriers effectively, largely due to its polar carboxyl group [110]. One promising strategy is to employ prodrug approaches, such as 4-chlorokynurenine and SZR-104, whereby this polar moiety is temporarily masked through esterification or similar chemical modifications [56,125]. By doing so, researchers can enhance lipophilicity, potentially improving permeability across the gut or BBB. Once inside target tissues, endogenous enzymes would cleave the ester bond, liberating active KYNA precisely where it is needed [126]. Another avenue involves synthesizing hybrid molecules, combining the core quinoline structure with lipid-soluble components [127]. This approach aims not only to improve membrane permeability but also to confer additional functional advantages, such as greater stability or targeted delivery [128]. For instance, merging KYNA with a fatty acid chain could optimize drug distribution in neuronal membranes, where excitotoxic processes frequently unfold [69]. While these strategies are conceptually appealing, they require systematic evaluations to ensure that altered pharmacokinetics do not come at the cost of diminished efficacy or unexpected side effects. Nevertheless, these innovations are promising for turning endogenous metabolites into practical therapies that address significant clinical needs.
One way to translate preclinical findings into clinical success is through dual-targeting compounds. These agents can influence key Trp-metabolizing enzymes while also modulating neuroinflammatory pathways [129]. By tackling both processes at once, these agents have the potential to dampen the production of harmful quinoline-based metabolites, such as QUIN, while also curbing inflammatory cascades that can exacerbate neuronal damage [130]. For example, inhibiting enzymes like IDOs or TDO while suppressing pro-inflammatory cytokines may reduce excitotoxic damage in neurodegenerative or chronic inflammatory conditions [131,132,133]. In diseases like multiple sclerosis, recent work emphasizes the critical importance of accurately monitoring redox dynamics as both a diagnostic tool and a therapeutic target, revealing that an oxidative imbalance directly influences disease progression and treatment responsiveness [134]. The rationale is to address multiple disease mechanisms in parallel, thereby enhancing therapeutic efficacy and possibly reducing the likelihood of resistance or relapse [135]. However, designing these dual-acting agents demands a nuanced understanding of each target’s regulatory networks, as altering enzyme activity may unintentionally shift the balance toward alternative metabolic routes or trigger compensatory immune responses [136]. Moreover, the BBB remains a formidable challenge, necessitating delivery strategies that ensure sufficient drug concentrations within the central nervous system [137].
While enhancing BBB penetration is essential, true therapeutic success often hinges on directing molecules to the specific CNS regions and cell populations most affected by disease. Recent work on ligand-decorated nanoparticles and receptor-specific peptides illustrates how researchers can guide drugs toward, for example, the hippocampus in Alzheimer’s disease or the substantia nigra in Parkinson’s, rather than distributing agents uniformly throughout the brain. Equally important is ensuring that active compounds localize to the relevant intracellular compartments—such as mitochondria or lysosomes—where key neuroprotective or anti-inflammatory mechanisms operate. By combining BBB-focused strategies with region- and cell-specific design, future quinoline-based therapies may significantly improve both efficacy and safety, reducing off-target burdens and optimizing outcomes in complex neurological conditions. Despite these hurdles, dual-targeting compounds offer a forward-thinking strategy, uniting key biochemical nodes into a single intervention that could revolutionize how we harness endogenous quinoline metabolites for clinical benefits.

3. Expanding the Quinoline Landscape: Derivatives Beyond the Kynurenine (KYN) Metabolic Pathway

Isonicotinic acid (INA) shares a structurally similar pyridine ring with KYNA’s quinoline core, creating promising opportunities for scaffold hybridization. These include investigating underexplored agents, like dipicolinic acid (DPA), in neurodegenerative applications [138]. However, these compounds may present limitations, including off-target bacterial DNA gyrase inhibition (e.g., nalidixic acid) or toxicity concerns (cinchoninic acid derivatives) [139,140]. Structural homology between isonicotinic acid (INA)’s pyridine ring and KYNA’s quinoline core highlights opportunities for scaffold hybridization and the exploration of underexamined agents, like dipicolinic acid (DPA), in neurodegeneration (Table 2). Further, systematic SAR studies on broad-spectrum drugs, exemplified by quinidine, could isolate neuroactive fragments for targeted therapeutic applications and ultimately safer use [141].
Despite the different historical trajectories of classical quinoline agents (like antimalarials and antibiotics) and those directly linked to the KYN pathway, they share structural underpinnings and overlapping challenges. Lessons from modifying older quinoline frameworks to improve safety, enhance stability, or extend clinical utility can be leveraged to optimize newer KYNA-inspired compounds. For instance, understanding how subtle ring substitutions in antibacterial quinolones influence enzyme targeting may inform strategies to fine-tune receptor specificity and pharmacokinetics in KYNA analogues. By connecting these two research streams, we establish a cohesive framework that underscores the universal importance of precise scaffold manipulation in drug discovery.

3.1. Repurposing Potential

XA, although not a central intermediate in the KYN pathway, illustrates how precise structural changes can influence receptor interactions and metabolic stability [210]. Introducing a hydroxyl group at specific sites on the quinoline ring modifies hydrogen bonding capacity, which may shift affinity toward NMDA receptors or GPR35 [105]. Such alterations affect not only receptor binding but also downstream processes, including neurotransmission and immune signaling [211]. Adjustments in stereochemistry or the addition of other substituents can further redirect molecular preferences, raising the possibility of selectively targeting distinct physiological pathways [212]. Yet, enhancing receptor specificity often comes with trade-offs [213]. For instance, while certain hydroxylations strengthen binding, they may also increase vulnerability to enzymatic degradation [214]. Strategies, such as incorporating protective groups or additional ring modifications, can help extend the plasma half-life and improve pharmacokinetic profiles [215]. Beyond the structural design, patient stratification based on genetic variations in enzymes, like IDOs and kynurenine 3-monooxygenase (KMO), offers a route to refine therapeutic applications [216]. Identifying metabolic subgroups may improve treatment outcomes and reduce adverse effects, particularly in neurodegenerative and neuroinflammatory conditions, where individual responses vary widely [217].
Quinoline derivatives beyond the KYN pathway present structural opportunities but are often limited by safety concerns that complicate therapeutic development [109]. Nalidixic acid, widely recognized for its antibacterial activity through the inhibition of DNA gyrase, raises concerns when considered for neuroprotective or immunomodulatory purposes [218]. Interference with DNA metabolism, while effective against bacterial targets, may introduce risks to human cells, particularly in long-term or systemic applications [219]. Similar challenges appear with cinchoninic acid derivatives, which share structural features with KYNs yet exhibit toxicity that restricts their clinical use [220]. In several cases, these effects appear to be linked to the formation of reactive metabolites or unintended interactions with metabolic enzymes [221]. Even minor modifications to the quinoline scaffold can redirect metabolic pathways, sometimes amplifying toxic outcomes [222]. Addressing these complexities requires a detailed understanding of structure–toxicity relationships and strategies that preserve pharmacological activity while minimizing off-target effects [223]. Approaches, such as selective functional group modifications and alternative synthetic routes, are under investigation [224]. Advancing these efforts depends on collaboration across medicinal chemistry, toxicology, and clinical research to reduce liabilities and support the development of safer quinoline-based compounds [225].

3.2. Bridging to Kynurenic Acid (KYNA)-Based Targets

Structural similarities between INA and KYNA have prompted interest in hybrid molecules that combine elements of both frameworks [226]. Each features a nitrogen-containing heterocycle capable of influencing receptor interactions and electron distribution [227]. By integrating portions of the quinoline core from KYNA with the pyridine ring of INA, researchers aim to enhance selectivity at targets, such as NMDA receptors and GPR35. This approach allows for adjustments in the ring orientation and substituent placement, offering opportunities to fine-tune receptor engagement while addressing challenges related to metabolic stability [228]. The isonicotinic scaffold, in particular, provides flexibility for introducing functional groups that may improve solubility or permeability [229]. While these modifications hold promise for minimizing off-target effects, the relationship between structural changes and pharmacological outcomes remains complex [230]. Balancing synthetic accessibility with therapeutic potential requires a careful evaluation of how hybrid designs alter binding properties and metabolic pathways. Drawing from the existing knowledge of INA derivatives, ongoing studies will assess whether these structural combinations can deliver improved performance while limiting adverse effects, especially in applications involving neuroprotective and immunomodulatory pathways.
DPA, a pyridine-2,6-dicarboxylic acid, remains relatively underexplored despite its capacity to bind metal ions commonly implicated in neurodegenerative diseases [231]. Through interactions with iron, zinc, and copper, DPA may contribute to regulating the metal balance, a process closely linked to oxidative stress and protein aggregation [232]. Preliminary findings suggest potential roles in modulating inflammatory responses and excitatory signaling, raising questions about its relevance to therapeutic strategies aimed at neuroprotection [233]. Structurally, the resemblance between DPA’s pyridine core and the quinoline ring of KYNA presents opportunities to combine metal-chelation with receptor targeting [234]. This dual functionality may prove valuable, particularly in disorders where oxidative damage and glutamatergic dysregulation intersect [235]. Parkinson’s disease and Alzheimer’s disease are two examples where such mechanisms converge, though much remains unknown regarding DPA’s pharmacological behavior [236]. Determining its metabolic stability, toxicity profile, and specific enzyme interactions will be essential for guiding further development [237]. By applying knowledge from KYNA analog research, it may be possible to design compounds that extend the benefits of metal-binding while refining receptor selectivity, supporting efforts to address complex neurodegenerative conditions [238].

3.3. Innovative Directions

Identifying neuroactive components within broad-spectrum quinoline derivatives offers a pathway to develop treatments with fewer adverse effects [12]. Quinidine, primarily recognized for its role in modulating cardiac ion channels, provides a useful example [225]. Through structure–activity relationship analyses, specific molecular features can be mapped to isolate those contributing to neurological benefits, such as limiting excitotoxicity, while reducing the risk of arrhythmias [239,240]. Computational docking and high-throughput screening techniques assist in pinpointing functional groups that influence ion channels within the central nervous system, rather than the heart [241]. These insights guide the design of analogues tailored to interact with neuronal targets, including channels and receptors implicated in neurodegenerative disorders [242]. Achieving this balance between the therapeutic effect and safety often relies on incremental structural adjustments—altering side chains, introducing new substituents, or modifying ring systems can significantly affect the binding behavior and metabolic stability [243]. While quinidine’s properties are well characterized, questions remain about how best to adapt its framework for neurological applications [244]. Ongoing work with related compounds may help define strategies to refine multi-purpose quinoline scaffolds and explore their potential in treating conditions, such as epilepsy, chronic pain, and neurodegeneration.

4. Exogenous Horizons: Synthetic Quinoline Scaffolds in Rational Drug Design

Driven by rational design approaches, synthetic modifications to quinoline-based scaffolds have enhanced both target engagement and pharmacokinetic profiles [245]. For example, chlorination in 7-CKA and sulfonamide substitution in Gavestinel improved NMDA receptor blockade and bioavailability, respectively [246,247,248,249]. Despite encouraging preclinical results, clinical outcomes have sometimes fallen short. For example, Gavestinel ultimately failed in stroke trials [250]. Encouragingly, newer therapies, like laquinimod, show immunomodulatory benefits in multiple sclerosis [251]. These findings highlight the difficulty of balancing selectivity and polypharmacology. L-701,324, for instance, exhibits strong NMDA receptor antagonism yet also causes unwanted glutamatergic side effects (Table 3) [252]. Future strategies emphasize BBB-penetrant analogs and multi-target ligands exemplified by tasquinimod, including HDAC (histone deacetylase) 4 inhibition and aryl hydrocarbon receptor modulation.

4.1. Rational Design Successes and Failure

Refining quinoline scaffolds through targeted synthetic modifications has proven effective in improving receptor selectivity and pharmacokinetic properties [293]. In the case of 7-CKA, introducing a chlorine atom at the 7-position strengthens antagonism at NMDA receptors, surpassing the activity of KYNA [253]. This substitution not only stabilizes the molecule’s conformation at the binding site but also alters its metabolic profile, with potential implications for its clearance and half-life [294]. Gavestinel presents a related example, where the incorporation of sulfonamide groups enhances solubility and may support passage across the BBB, features that are critical for central nervous system activity [295]. While such modifications can improve receptor engagement and bioavailability, they also introduce challenges [296]. Altering chemical structures to enhance efficacy may inadvertently affect safety, particularly as compounds advance toward clinical evaluations [297]. Balancing these factors requires careful attention to the relationship between functional groups and biological outcomes [298]. Through studies of compounds, like 7-CKA and Gavestinel, medicinal chemistry continues to identify structural features that support NMDA receptor modulation, offering a foundation for the development of new treatments targeting neurological disorders.
A clear pattern emerges when considering how discrete structural modifications influence the pharmacological profiles of quinoline-based derivatives. Introducing halogens (e.g., Cl, F) at critical ring positions can improve receptor blockade—especially at NMDA’s glycine site—while often enhancing lipophilicity for better BBB penetration, as seen with 7-CKA. Conversely, sulfonamide substitutions (such as those in Gavestinel) significantly boost solubility yet require careful balancing to avoid off-target interactions. Fluorinated derivatives likewise tend to display enhanced metabolic stability, extending the half-life without substantially raising the toxicity risk. By systematically mapping how each substituent alters binding affinity, membrane permeability, and metabolic clearance, we gain a cohesive SAR framework. Such an approach clarifies why certain motifs are selected to reduce excitotoxicity in neurodegenerative models, enhance anti-inflammatory effects, or leverage immunomodulatory mechanisms for oncology applications. Centralizing these SAR principles not only consolidates the scattered findings but also serves as a blueprint for future chemical refinements and targeted therapeutic design (Table 4).
The challenges of translating quinoline-derived compounds into clinical success are illustrated by the case of Gavestinel. Developed to limit excitotoxic injury following stroke through NMDA receptor antagonism, Gavestinel advanced from preclinical studies with strong supporting data [299]. Yet, in clinical trials, it failed to produce meaningful improvements in patient outcomes, raising questions about the limitations of commonly used experimental models [300]. Stroke in humans presents variability in its onset, severity, and timing that is difficult to replicate in animal studies [301]. Many preclinical trials rely on designs lacking adequate randomization, blinding, or statistical power, factors known to overestimate efficacy in stroke research [302]. Differences in treatment windows also complicate translation [303]. While preclinical models often apply interventions early, real-world scenarios involve delays, with irreversible damage frequently present before therapy begins [303]. Addressing these discrepancies requires trial designs that account for the complexity of human stroke and consider combining neuroprotective strategies with reperfusion approaches [304]. Without such adjustments, promising compounds remain at risk of falling short in clinical settings, regardless of structural refinement or the pharmacological rationale.
The failure of IDO inhibitors in oncology trials has raised important considerations for the development of quinoline-based therapies [305]. Epacadostat, an IDO inhibitor evaluated for melanoma, did not meet its clinical endpoints, partly due to limited patient stratification and the absence of reliable biomarkers to predict treatment responses [306]. Similar issues have been observed with Gavestinel in stroke studies, where variability in disease onset and delayed intervention may have obscured the potential benefits [307]. These examples point to the value of using biomarkers—such as measures of KYN pathway activity or inflammatory cytokines—to guide patient selection and improve trial outcomes [308]. Combining KYNA analogs with immune checkpoint inhibitors or metabolic regulators may also help counteract compensatory mechanisms that reduce therapeutic effectiveness [309]. Trial designs that include real-time pharmacokinetic assessments, like cerebrospinal fluid analysis to verify central nervous system exposure, could support better decision-making during clinical evaluation [310]. Failures, such as epacadostat’s, also reflect the limitations of animal models, which often fall short of replicating the complexity of human pathophysiology [311,312]. Integrating techniques, like metabolomics and single-cell RNA sequencing, into preclinical research may help identify specific patient subgroups who are more likely to benefit from targeted quinoline derivatives.
Laquinimod, a quinoline-3-carboxamide derivative, offers a contrasting example of how molecular design can align with the disease context to support therapeutic development [313]. In multiple sclerosis, where treatment focuses on managing chronic inflammation leading to neurodegeneration rather than responding to acute injury, laquinimod’s ability to reduce pro-inflammatory cytokines and support neuronal preservation has led to more favorable outcomes compared to agents like Gavestinel [314]. Unlike stroke, where delays in intervention often limit the impact of neuroprotective strategies, the progressive nature of multiple sclerosis provides a broader window in which immunomodulation may alter disease progression [315]. These differences raise an important question: how can molecular targets be matched more effectively to the underlying biology of a condition? Laquinimod’s selective focus on immune pathways contrasts sharply with the NMDA receptor antagonism of Gavestinel, highlighting the value of mechanisms that reflect the complexity of the disease environment. While setbacks in clinical translation are common, cases like laquinimod suggest that carefully tailored approaches, applied to appropriate patient groups, may improve the likelihood of success and offer meaningful advances in treating neurodegenerative disorders [316,317].
The varied structural modifications outlined (e.g., halogenation in 7-CKA, sulfonamide substitution in Gavestinel, or the esterification approaches in KYNA analogues) reveal how each tweak can be purposefully directed toward distinct pathologies. For neurodegenerative disorders, strengthening NMDA receptor antagonism while preserving BBB penetrability is pivotal to safeguarding neurons against excitotoxicity. In oncology, motifs that improve solubility and selectivity could be paired with known immune checkpoint inhibitors, leveraging the immunomodulatory potential of quinoline scaffolds. Meanwhile, the anti-inflammatory efficacy hinges on substituents that curb cytokine release without undermining systemic safety. By systematically mapping these structural “levers” to disease-specific objectives, researchers can shape more refined lead compounds and rapidly iterate toward clinically viable designs.

4.2. Mechanistic Trade-Offs

L-701,324 has drawn attention for its strong ability to block NMDA receptors and suppress excitatory neurotransmission [318]. In conditions, such as epilepsy and neuropathic pain, this level of inhibition may offer therapeutic benefits [319]. Yet, the same mechanism that provides relief can also carry significant risks [320]. Cognitive disturbances, sedation, and symptoms resembling psychosis have been linked to excessive glutamatergic suppression, raising difficult questions about how far receptor blockade should go [321]. Should drug development focus on narrowing selectivity to avoid unintended effects, or might a broader range of activity offer advantages in disorders shaped by multiple interacting pathways? While some support the latter, NMDA receptor antagonists present clear limitations [322]. Even small deviations in targeting can disrupt neural functions, underscoring the importance of precise molecular engagement [323]. The case of L-701,324 highlights this tension between therapeutic potential and safety [324]. Adjusting the receptor affinity without tipping the balance too far remains a central challenge [322]. As research moves forward, designing compounds that maintain effective modulation while minimizing disruption will be key to expanding the use of NMDA antagonists in clinical settings.
A growing strategy involves developing BBB-penetrant derivatives that build on the neuroprotective potential of KYNA while addressing its limited access to the central nervous system [56]. One example includes modifying KYNA with tert-butyl ester groups, which may improve its transport across the BBB before enzymatic processes release active KYNA within neural tissue [325]. Alongside these efforts, multi-target approaches are attracting interest, particularly for conditions shaped by overlapping biological pathways [326]. Tasquinimod offers a case in point, combining HDAC4 inhibition with aryl hydrocarbon receptor modulation to influence both immune responses and cellular growth [327]. Such dual-action strategies aim to address the complexity of disorders like cancer and autoimmune diseases, where inflammation, proliferation, and tissue damage converge [328]. Whether the goal is to strengthen the delivery of KYNA analogs or to design compounds that coordinate multiple mechanisms, the challenge lies in maintaining therapeutic benefits while controlling off-target effects [329]. Progress will depend on refining these scaffolds through systematic screening, predictive modeling, and careful pharmacokinetic evaluations [330]. Together with biomarker-informed clinical studies, these efforts may guide the development of quinoline-based therapies for neurological, oncological, and inflammatory conditions.
Quinoline derivatives are gaining attention for their ability to influence epigenetic mechanisms, particularly through the modulation of histone deacetylation and DNA methylation [331]. Tasquinimod, targeting HDAC4, has demonstrated potential in addressing both neurodegenerative conditions and cancer, while newer compounds are being designed to expand activity across additional HDAC and DNMT targets [332]. These effects may help to restore tumor-suppressor functions or regulate inflammatory pathways [333]. Genetic variation within the KYN pathway, including polymorphisms in IDOs and KMO, adds another layer of complexity by shifting the balance between protective and harmful metabolites [334]. Combining genotyping with metabolic profiling may guide treatment strategies [335]. Moving forward, refining these molecules will require careful structure–activity studies and genotype-based clinical trials.
Recent findings also underscore the potential of lipid-based nanoparticle systems to improve the delivery of quinoline derivatives across the BBB [336]. For instance, encapsulating these molecules in lipid vesicles can mitigate their rapid clearance, thereby increasing CNS bioavailability [337]. This strategy not only allows for sustained drug release but also reduces off-target toxicity—advances that could reinvigorate clinical interest in repurposing or refining existing quinoline scaffolds [338]. In parallel, polymeric nanoparticles and biomimetic vesicles, such as exosome-based carriers, are emerging as next-generation vehicles for transporting hydrophilic or ionizable quinoline species [339]. By tailoring the particle size, surface charge, and release kinetics, researchers can customize how these agents distribute and act in vulnerable CNS regions. Nanotechnology offers promising avenues to overcome the delivery challenges of polar quinoline derivatives, like KYNA. By encapsulating these molecules in exosomes or polymeric nanoparticles, it is possible to enhance their stability, improve BBB penetration, and achieve targeted release at the site of action. This advanced delivery strategy not only increases the local drug concentration but also minimizes systemic exposure, paving the way for more effective neuroprotective therapies. Incorporating such advanced delivery mechanisms will be crucial for translating preclinical ‘hits’ into viable, patient-friendly therapeutics.

5. Next-Generation Kynurenic Acid (KYNA) Analogues: The SZR Series

Structure–activity relationship (SAR) studies yield SZR derivatives with improved pharmacological profiles [340]. SZR-72 shows enhanced neuroprotection, effective blood–brain barrier (BBB) penetration, and behavioral modulation [341,342,343]. SZR-104 displays high BBB permeability and neuroprotective effects in sepsis models [54,344]. SZRG-21 demonstrates the effect on motor activity and emotional behavior [343]. SZR-109 stands out for its strong BBB penetration, suppression of TNF-α, upregulation of TSG-6, and anti-convulsant activity [56,345,346]. While SZR-73 enhances mitochondrial functions and reduces systemic inflammation in sepsis, it does not restore microcirculation. This limitation curtails its overall therapeutic potential [229]. SZR-105, while highly BBB-permeable and anti-inflammatory, requires further validation for its long-term safety [56,345]. Prioritizing SZR-81 may be premature due to poor BBB penetration, although its antidepressant-like effects and neuroprotective potential are promising (Table 5) [347]. Lead optimization may benefit from synergistic strategies. For instance, combining these analogs with IDO inhibitors could more effectively modulate the KYN pathway and boost therapeutic efficacy.

5.1. SAR-Driven Innovations

In-depth SAR investigations reveal how subtle modifications to the KYNA core can profoundly affect receptor affinity and metabolic traits [349]. SZR-72, for instance, features a small methyl group that contributes to its enhanced neuroprotective effects, blood–brain barrier penetration, and behavioral modulation—suggesting that subtle steric modifications may play a significant role in optimizing CNS-targeted activity [343]. Meanwhile, SZR-104’s C3 addition of the polar ring system demonstrates how introducing electronegative groups can enhance BBB permeability and support neuroprotection, especially in systemic inflammatory conditions [343]. By tweaking these chemical features—adding a methyl here, attaching a ring system there—researchers can systematically fine-tune key pharmacological properties, like the potency and half-life [350]. Such directed alterations reflect a deeper understanding of how receptor–ligand interactions depend on precise spatial and electronic configurations. In some cases, these modifications not only boost the BBB permeability but also reduce off-target binding, mitigating potential side effects [39]. Additionally, selecting the right functional group can influence polarity and solubility, traits that shape oral bioavailability [351]. Together, these iterative design strategies exemplify the power of SAR-driven innovations to optimize molecules for both efficacy and safety [352]. Beyond SZR-72 and SZR-104, further explorations could unleash even more advanced analogues that are fine-tuned to specific therapeutic goals. As we learn more, SAR mapping remains a cornerstone of rational drug discovery for KYNA derivatives.
Among the ever-growing list of KYNA analogues, SZRG-21 and SZR-109 stand out for their distinct therapeutic profiles in vivo [343,346]. Meanwhile, SZR-72, SZR-104, and SZRG-21 influence motor-associated curiosity and emotional behavior, implying broader neuromodulatory effects [343]. On the other hand, SZR-109 exhibits potent neuroprotective properties, notably suppressing TNF-α production and upregulating TSG-6 [345]. Its ability to effectively penetrate the blood–brain barrier and its anti-convulsant effects—preventing seizure-like activity—further highlight its therapeutic potential in central nervous system disorders [56,346]. The contrasting profiles of these analogues underscore how strategic SAR modifications can yield compounds tailored to specific pathophysiologies. While SZRG-21 appears suited to modulate behavioral aspects, SZR-109 is structurally optimized for neuroprotection [343,346]. These findings emphasize the flexibility of the KYNA scaffold, which allows for fine-tuned substituents to address diverse pathological targets. Further studies on their binding affinities and pharmacokinetics may unlock new therapeutic applications and refine their clinical relevance. Ultimately, SZRG-21 and SZR-109 exemplify how SAR-guided innovation can push the boundaries of KYNA-based therapeutics.

5.2. Translational Barriers

Despite ongoing refinements to KYNA analogues, multiple translational barriers must be addressed before these compounds can achieve widespread clinical utility. Prodrug strategies or nanoparticle delivery systems could potentially mitigate such clearance problems, yet these approaches introduce their own complexities in formulation and manufacturing [353,354]. On another front, SZR-105 demonstrates considerable promise but poses its own safety concern: off-target kinase inhibition [56,355]. Even minor structural tweaks can inadvertently shift binding toward other enzymes or receptors, jeopardizing patient safety [343,356].
Recent advances in nanoparticle encapsulation, lipid-based vesicles, and exosome carriers have demonstrated notable improvements in delivering quinoline derivatives across the blood–brain barrier. For example, encapsulating polar KYNA analogs in liposomes can maintain effective drug concentrations in CNS tissues, while minimizing systemic exposure and associated side effects. Complementary to these delivery strategies, computational modeling—particularly machine-learning-driven structure-activity predictions—can highlight potential off-target receptors or enzymes early in the design process. This approach helps to refine scaffolds before in vivo testing, thereby cutting down on costly late-stage failures and patient safety risks. Integrating both cutting-edge formulation methods and in silico screening thus represents a powerful path to overcoming longstanding challenges in quinoline-based neurotherapeutics.
Balancing pharmacological potency with a clean off-target profile is therefore a central challenge in modern drug development [357,358]. Medicinal chemists must undertake detailed SAR investigations, systematically exploring substituents and functional groups to minimize toxicity while preserving therapeutic efficacy [359]. Such iterative optimization demands close collaboration among chemists, pharmacologists, and toxicologists, each focusing on a different aspect of a molecule’s life cycle [360]. By carefully addressing problems, like rapid clearance and unintended kinase inhibition, researchers can pave the way for safer, longer-lasting KYNA-based treatments that ultimately improve patient outcomes.

5.3. Path to Lead Optimization

Achieving an optimal lead among KYNA analogues calls for balancing efficacy, selectivity, and favorable pharmacokinetics [361]. Balanced profiles are critical in translational research, where the nuances of absorption, distribution, metabolism, and excretion (ADME) intersect with complex biological pathways [362]. High bioavailability ensures that the compound can be administered conveniently, improving patient compliance and potentially reducing the dosing frequency [363]. Meanwhile, precise receptor selectivity helps to mitigate adverse effects by limiting unwanted off-target binding [364]. These attributes underscore the importance of methodical SAR evaluations, wherein subtle modifications to the parent scaffold can bolster both pharmacodynamics and pharmacokinetics [365]. Complementary techniques, like computational modeling and in vitro assays, further expedite the refinement process, shedding light on metabolic pathways and guiding structural tweaks [366]. By identifying leads in the SZR series, researchers can streamline development cycles and reduce the risk of late-stage failures in clinical trials [367]. Ultimately, focusing on candidates that marry oral bioavailability with selective receptor engagement represents a viable strategy to usher KYNA-based therapies toward their full therapeutic potential.
Adopting combinatorial approaches that integrate multiple therapeutic strategies can heighten the efficacy of KYNA-based treatments [368]. One promising avenue involves combining SZR analogs, designed to selectively modulate GPR35 or NMDA receptors, with established IDO inhibitors that curtail the overall flux through the KYN pathway [369]. Recent findings underscore the intestinal microbiome’s pivotal role in generating, modifying, and regulating KYN pathway metabolites. Microbial enzymes can convert dietary tryptophan into an array of quinoline derivatives that enter systemic circulation, affecting immune homeostasis and neural functions. This dynamic also presents new opportunities for the oral delivery of quinoline-based drugs. Formulations designed to remain stable in the gastrointestinal tract—or even to be activated by specific microbial enzymes—may boost the local or systemic availability of therapeutic agents. Understanding the interplay between the gut microbiota and the KYN route thus represents a crucial frontier for optimizing both the efficacy and safety of next-generation quinoline therapies.
By partially blocking the production of downstream metabolites, IDO inhibitors reduce the burden of neurotoxic or immunomodulatory intermediates, thus creating a biochemical context where KYNA analogs may exert stronger protective or immunoregulatory effects [370]. This two-pronged tactic could help maintain stable levels of beneficial metabolites, like KYNA, while simultaneously minimizing harmful byproducts, leading to more robust interventions in neurodegenerative or inflammatory conditions [371]. Preclinical data suggest that the synergy may stem from the interplay between decreased KYN production and amplified receptor engagement by the analogs [372]. However, identifying optimal drug combinations will require meticulous dose–response studies and toxicological evaluations [373]. In particular, attention to pharmacokinetic profiles, such as how these agents interact or compete for metabolic pathways, is essential to avoid unintended consequences [374]. Ultimately, harnessing a combinatorial approach, like SZR analogs plus IDO inhibitors, illustrates the potential of integrated interventions targeting multiple points along the KYN pathway [375]. Such strategies may yield tangible clinical outcomes that surpass those achievable with single-agent therapies alone.

6. Discussion

The KYN metabolic system serves as a critical nexus in metabolism, intricately linking Trp catabolism to the production of neuroactive and immunomodulatory quinoline-based compounds [376]. Central to this pathway are endogenous metabolites, such as KYNA and QUIN, which exert intrinsically distinct and often opposing effects both individually and in relation to each other, modulating the equilibrium between neuroprotection and excitotoxicity, immune responses, and the redox balance [8,377]. Structural nuances within their quinoline scaffolds, including carboxylation, halogenation, and hydroxylation, profoundly influence their receptor specificity (e.g., NMDA, GPR35) and metabolic stability, underscoring the interplay between chemistry and physiology [124]. Despite their therapeutic promise, translational hurdles, such as poor bioavailability, rapid clearance, and paradoxical biological effects, impede clinical progress [378]. This review systematically evaluates endogenous and synthetic quinoline derivatives, emphasizing their structural diversity, mechanistic trade-offs, and therapeutic potential across neurodegenerative, inflammatory, and oncological contexts. By synthesizing structure–activity relationships and translational challenges, we aim to identify a lead compound inspired by KYNA, prioritizing innovations, like prodrug strategies and multi-target engagement, to bridge the gap between preclinical promise and clinical efficacy [379,380]. Our findings aim to catalyze rational drug design for quinoline-based therapies in complex diseases.
Building on the preceding sections, we highlight how research on the chemical identity, structural nuances, and biological roles of quinoline-based metabolites points to significant therapeutic opportunities [225]. Across various endogenous and synthetic derivatives, the document emphasizes the delicate interplay between neuroprotective and neurotoxic effects, demonstrating how minor structural shifts can dramatically influence receptor binding, solubility, and metabolic stability [381]. By exploring molecules, like KYNA and QUIN, alongside analogues engineered for improved pharmacokinetics or selectivity, the synthesis showcases promising targets for conditions spanning neurodegenerative disorders and cancer [382]. Of particular note is the attention placed on bridging preclinical findings with translational barriers, including bioavailability challenges and safety concerns [383]. Overall, this review demonstrates that tuning the quinoline scaffold holds vast potential for advancing novel interventions, underscoring the need for continued interdisciplinary efforts to harness these insights for tangible patient benefits.
These four pillars provide a cohesive blueprint for taking KYNA-inspired quinoline derivatives from the lab to real-world treatments (Table 6). First, mapping disease pathways ensures that each structural motif addresses specific pathogenic processes—be it glutamate excitotoxicity in Alzheimer’s disease or immune dysregulation in rheumatoid arthritis. Next, meticulous SAR refinements guide rational changes in polarity, substituent placement, or prodrug modifications, thereby enhancing target engagement while curbing off-target interactions. Validating each candidate in context-specific models (like transgenic mouse lines for Parkinson’s or tumor xenografts) then confirms that the observed in vitro benefits actually translate into meaningful in vivo outcomes. Finally, integrating biomarker-based selection ensures that researchers can pinpoint which patient subgroups are most likely to benefit, ultimately increasing trial success rates and fostering more precise, personalized therapy. By weaving together these pillars, investigators build a robust framework for innovation—one that promotes both scientific rigor and practical translational impact in the development of next-generation quinoline-based interventions.
Quinoline–metal complexes, particularly 8-hydroxyquinoline chelators, play a crucial role in mitigating oxidative stress in Alzheimer’s disease [133,384,385]. By binding excess metal ions, such as copper and zinc, these compounds prevent the metal-catalyzed production of reactive oxygen species, thereby reducing neuroinflammation and amyloid aggregation [386]. Additionally, their ability to regulate the redox balance and modulate autophagy pathways highlights their potential as multi-target neuroprotective agents [387]. Notably, recent breakthroughs in migraine therapeutics have underscored how targeting neuropeptides, such as calcitonin-gene-related peptide, pituitary adenylate cyclase-activating polypeptide, and vasoactive intestinal polypeptide, can reshape our understanding of neuroinflammation and pain transmission, further emphasizing the need to develop quinoline-based agents with pleiotropic mechanisms [388]. Future research should focus on optimizing their stability and brain permeability to enhance clinical applicability. Structural modifications of KYNA derivatives extend beyond receptor binding, influencing critical downstream pathways, such as mTOR and autophagy [389]. Minor alterations in functional groups can modulate phosphorylation events within the mTOR axis, potentially affecting neuronal survival and inflammatory responses [390]. Additionally, shifts in KYNA’s metabolic profile may alter autophagy-related signaling, impacting cellular homeostasis under oxidative stress conditions [391]. These mechanistic insights highlight the need for targeted modifications that optimize KYNA analogs for neuroprotective or immunomodulatory applications. Such synergy underlines the manuscript’s contribution to research.
Although the KYN pathway is often discussed in terms of glutamate receptor modulation, its complex interaction with serotonin metabolism and mitochondrial function deserves greater attention [23,392]. In particular, QUIN—a downstream metabolite—can significantly impact neuronal energy homeostasis by depleting NAD+ through the de novo salvage pathway [393]. When QUIN accumulates, it not only drives excitotoxic damage but also impairs mitochondrial electron transport, exacerbating oxidative stress and triggering deficits in ATP production [394]. This metabolic interplay may be especially critical in neurodegenerative disorders, where chronic inflammation and mitochondrial dysfunction intersect to accelerate disease progression [395]. Meanwhile, upstream metabolites within the KYN pathway compete for Trp, potentially altering serotonin synthesis and influencing mood regulation [392]. Such biochemical crosstalk helps to explain why patients with conditions like Parkinson’s disease or Alzheimer’s often present with both cognitive and affective symptoms [396,397]. By illuminating how QUIN’s NAD+-depleting properties converge with disrupted serotonin metabolism, researchers can identify new multi-target strategies—such as modulating key enzymes in each branch of Trp degradation. Ultimately, a deeper appreciation of these interconnected metabolic nodes can refine our understanding of disease mechanisms, paving the way for therapeutic approaches that tackle excitotoxicity, energy deficits, and mood dysregulation in a more holistic manner [398,399].
Beyond exploring prodrug designs and multi-target ligands, integrating emerging technologies—particularly CRISPR-based screening—could elevate the originality and impact of quinoline-focused research [400,401]. CRISPR knockouts or knock-ins enable the precise genetic manipulation of targets implicated in the KYN pathway, thereby uncovering how specific enzymes, transporters, or receptors influence the efficacy of quinoline derivatives [402]. This level of control helps to delineate off-target effects and refine SAR predictions, ensuring that lead compounds undergo more robust validation before advancing to clinical trials [403]. Additionally, CRISPR-based functional genomics can be paired with advanced phenotypic assays (e.g., high-content imaging or single-cell transcriptomics) to capture subtle changes in cell viability, immune signaling, or neuronal stress responses [404]. Such multimodal approaches can reveal novel synergy points—for instance, identifying an unanticipated metabolic bottleneck that amplifies or diminishes the therapeutic potency of KYNA analogues [405]. By validating these genetic interactions early in the drug-development pipeline, investigators can better prioritize candidates with the highest translational potential, thereby reducing costly late-stage failures [404]. Ultimately, the marriage of CRISPR technologies and quinoline-focused medicinal chemistry stands poised to yield a new generation of targeted interventions, leveraging enhanced mechanistic precision to tackle neurodegenerative, oncological, and inflammatory pathologies in a truly innovative manner.
The chemical identity extends well beyond a molecule’s basic formula or nomenclature [406]. Subtle shifts in substituents—such as halogens, hydroxyls, or carboxyl groups—can drastically change a quinoline compound’s lipophilicity, stability, and affinity for target receptors [407]. For instance, introducing a chlorine atom at a strategic position may improve BBB penetration, while adding a bulky moiety might decrease solubility or alter enzyme-mediated metabolism [408]. These seemingly minor alterations can pave the way for novel pharmacological profiles, sometimes swinging the balance between neuroprotection and toxicity [409]. Historically, researchers have leveraged rational design methods to develop promising quinoline analogues with heightened specificity, boosted potency, and reduced side effects [245]. Nonetheless, large-scale synthesis remains challenging when certain derivatives exhibit poor yields or require stringent reaction conditions, thus complicating purification steps [410]. Adding to these hurdles, positional isomerism in the fused ring system can generate multiple byproducts that confound analytical assessments and limit overall compound purity [411]. Recognizing such synthetic barriers is critical for streamlining production pipelines and ensuring that lead candidates meet rigorous quality standards [412]. Overall, mapping out how even the smallest functional group can shift a compound’s fate is key to optimizing existing scaffolds and propelling more targeted therapeutic discoveries.
Biological mechanisms rarely operate in isolation. When a derivative modulates a single receptor or enzyme, it often triggers a ripple effect through multiple pathways, especially in the context of the KYN route [413]. Some compounds, for instance, may tamp down neuroinflammation by directly attenuating excitotoxic signals, only to exert a secondary influence on immune cells by altering cytokine release [414]. Such dual or even multi-pronged interactions underscore the need to see beyond straightforward receptor-binding data [415]. For KYN pathway analogues, identifying exact positions in the metabolic cascade reveals potential consequences for the redox balance and neuronal homeostasis: a slight shift in enzyme activity could tip the scales from protective antioxidant mechanisms to detrimental oxidative stress [416]. Yet, despite these insights, many indirect targets remain elusive. Off-target interactions—whether they involve uncharacterized receptors or ancillary enzymes—can complicate both therapeutic efficacy and safety [417]. More rigorous mapping of these secondary pathways, using both in vitro and in vivo models, could pinpoint previously overlooked nodes that either amplify or mitigate a compound’s effects [418]. Bridging such gaps would open the door for more targeted preclinical evaluations and eventually guide clinical trials toward meaningful outcomes [419]. Ultimately, unraveling these intricate webs of interaction is crucial for fully harnessing the therapeutic potential of quinoline-based metabolites.
Recent research has propelled several quinoline-based metabolites from exploratory benchwork to more tangible therapeutic leads [251]. In vitro assays frequently highlight their ability to modulate neuroprotective pathways, suppress inflammatory signals, or even arrest tumor cell proliferation [121]. Moving to in vivo models, some of these candidates have displayed meaningful behavioral improvements in neurodegenerative disease paradigms or slowed the progression of certain cancers [420]. However, conflicting data occasionally arise, with a compound that shows potent in vitro efficacy falling short in animal studies due to issues like rapid metabolism or unanticipated toxicity [421]. Such discrepancies underscore the complexity of translating molecular promise into robust, clinically relevant benefits [422]. Despite these hurdles, certain agents continue to stand out [423]. For example, derivatives with enhanced BBB penetration appear to be especially compelling for psychiatric or neurodegenerative indications, offering a potential edge over older, less permeable analogues [424]. Moreover, targeted delivery systems—ranging from liposomal encapsulations to prodrug designs—are making inroads by bolstering tissue specificity and minimizing off-target effects [425]. Formulation breakthroughs aside, many programs remain in their early clinical stages, where gaps in ADME data or sporadic toxicity reports can stall progress [426]. Ultimately, bridging these translational gaps will hinge on ongoing refinements in compound design, alongside more systematic preclinical and clinical assessments. Overall, these findings underscore how minor structural modifications in quinoline scaffolds can profoundly influence both protective and pathogenic pathways. Strategic changes to receptor specificity, delivery methods, and enzyme targeting therefore emerge as crucial levers for optimizing their therapeutic applications. A coordinated effort—bridging medicinal chemistry, molecular biology, and clinical research—can propel these compounds toward broader utility in complex disorders.
A vital takeaway is that the design principles underpinning classical quinoline derivatives can enrich the ongoing innovation around KYNA-based therapeutics. Whereas older agents (e.g., quinine and fluoroquinolone antibiotics) illustrate how small substitutions affect targeting and safety profiles, the emergent KYNA analogues reveal new avenues for immunomodulatory and neuroprotective actions. By deliberately blending these two perspectives—one rooted in historical clinical success, the other grounded in advanced pathophysiological insights—we open the door to broader applications that transcend any single therapeutic area. This integrative view not only bridges perceived gaps between “old” and “new” quinolines but also sets a trajectory for the next generation of rationally designed, multifaceted treatments.
Though the therapeutic promise of these quinoline derivatives remains compelling, a series of limitations underscores the need for a cautious and well-strategized development path [250]. Low bioavailability, for instance, often impedes their ability to reach efficacious concentrations at the intended site of action, forcing scientists to explore advanced formulation strategies [421]. Prodrug designs and nanoparticle encapsulations stand out as potential solutions, yet they introduce new complexities in manufacturing and regulatory approvals [12]. Additionally, unresolved safety profiles can stall clinical progress, particularly when a compound influences multiple biological pathways that could spark off-target effects [427]. These concerns highlight the importance of rigorous toxicological screening early in the pipeline. Furthermore, complex synthesis methods may hamper large-scale production, driving up costs and limiting accessibility for broader research [428].
Although many quinoline derivatives exhibit substantial therapeutic promise—from antibiotics to neuroprotective agents—recent reports also underscore concerns about potential organ toxicity, off-target enzyme inhibition, and unpredictable drug–drug interactions. For instance, while sulfonamide-modified scaffolds (such as Gavestinel) enhance solubility and CNS penetration, they can also trigger undesired side effects if not thoroughly screened. Similarly, some halogenated derivatives may inadvertently increase cardiotoxic or hepatotoxic risks. Moving forward, a more systematic approach to toxicity evaluations should accompany all efficacy studies in disease models (e.g., stroke, Parkinson’s, oncology) to ensure that potent therapeutic actions are not overshadowed by damaging toxicities. By integrating in vitro cytotoxicity assays, targeted biomarker analyses, and comprehensive in vivo evaluations, researchers can better align these potent scaffolds with real-world clinical safety standards.
Intellectual property barriers can add another layer of difficulty, restricting collaborative efforts and slowing the pace of innovation [429]. In short, no single fix exists, requiring multifaceted strategies and cross-disciplinary collaboration. Recognizing these challenges is essential for shaping the path forward: unless researchers and stakeholders address these technical, financial, and regulatory constraints, the full clinical impact of quinoline-based therapeutics may remain unrealized. By confronting these obstacles head-on, however, the scientific community stands poised to refine existing designs, explore novel delivery routes, and ultimately translate these promising molecules into real-world treatments.
In light of the diverse roles played by KYN-pathway enzymes, such as IDOs and KMO, incorporating robust biomarker strategies into clinical trial designs becomes essential [430]. This shift aligns with emerging paradigms in mental health diagnostics that emphasize biobehavioral precision over categorical rigidity [431,432]. By measuring baseline gene polymorphisms and enzyme expression levels, clinicians can stratify patients according to their potential responsiveness to quinoline-based agents [433]. This approach mirrors successes in targeted oncology, where genomic profiling matches treatments to precise molecular subtypes [434,435]. In the context of neurological and inflammatory diseases, leveraging a similar paradigm could help avoid the ‘one-size-fits-all’ pitfall, ensuring that individuals most likely to benefit—based on their enzyme activity profiles—are prioritized for quinoline therapies [126]. A growing body of research underscores how biomarker-driven stratification can transform trial outcomes for quinoline therapies. Measuring IDO/KMO activity levels or tracking KYN/QUIN ratios can pinpoint which patients exhibit heightened neuroinflammatory or excitotoxic pathways. Similarly, profiling cytokines (like TNF-α, IL-6) may reveal inflammatory ‘hot spots’ where KYNA-based interventions are most effective. Studies in oncology already illustrate how matching patients to treatments based on genetic markers can significantly enhance response rates. Applying a similar approach to neurodegenerative or autoimmune conditions ensures therapies reach those who stand to benefit most. As such, routine biomarker assessments not only accelerate clinical success but also pave the way for personalized, high-impact interventions. Such enrichment not only boosts the probability of clinical efficacy but also reduces the risk of adverse reactions in patients whose metabolic idiosyncrasies lead to off-target toxicity [436]. As studies of KYN pathway polymorphisms progress, stratification methods should become more refined. This refinement can guide the development of targeted interventions that harness the immunoregulatory and neuroprotective power of quinoline derivatives.
Moreover, polymorphisms in IDOs or KMO can drastically alter flux through the KYN pathway, impacting both the magnitude of neuroprotective metabolites (e.g., KYNA) and the generation of excitotoxic compounds (e.g., QUIN) [437]. This genetic variability underscores why certain patient subgroups respond differently—even within the same clinical trial cohort [438]. Incorporating routine genotyping or enzyme activity assays into trial protocols can clarify which individuals possess ‘favorable’ polymorphisms conducive to enhanced drug efficacy [439]. Conversely, identifying high-risk genotypes might prompt dose adjustments or the addition of secondary agents—like antioxidants or immune modulators—to mitigate potential side effects [440]. By interweaving this biomarker intelligence with standard clinical endpoints, researchers can implement adaptive trial designs that dynamically optimize treatment regimens [441]. This personalized approach stands to accelerate the clinical translation of quinoline-based therapies while reducing attrition rates, effectively bridging the gap between compelling preclinical data and consistent patient outcomes.
Despite this manuscript’s thorough exploration of quinoline-based strategies, a comparative perspective on non-quinoline neuroprotectants—such as indole-based IDO inhibitors or flavonoid antioxidants—could strengthen the rationale for prioritizing quinoline scaffolds [442]. Indole frameworks, while effective at modulating certain enzymes in the KYN pathway, often struggle with limited metabolic stability or suboptimal blood–brain barrier penetration [443]. Likewise, flavonoid antioxidants can offer substantial free-radical scavenging capabilities, yet their broad-spectrum activity and relatively weak target specificity can sometimes lead to off-target effects or insufficient impact on key regulatory nodes [444,445]. In contrast, quinoline cores frequently exhibit a more favorable balance of selective receptor binding and pharmacokinetic versatility, enabling the targeted modulation of both excitotoxic and inflammatory cascades [446]. By systematically juxtaposing these scaffold types, researchers would be better equipped to identify which structural features—such as ring fusion motifs or substituent placement—truly confer an edge in neurodegenerative or inflammatory models [447]. Moreover, head-to-head comparisons could clarify cost-effectiveness, synthesis scalability, and the likelihood of off-target liabilities [448]. Ultimately, a robust assessment of non-quinoline contenders would confirm whether quinolines indeed offer the most promising route to next-generation neuroprotectants, thereby ensuring that future research and development efforts are directed toward the scaffolds most likely to yield significant therapeutic benefits.
Looking ahead, several avenues present themselves for propelling this research toward tangible clinical gains. Novel synthetic strategies hold particular promise, as improving the yield or specificity of key intermediates could streamline large-scale production and expedite trials [449]. Equally intriguing is the potential for combining these quinoline derivatives with other pharmacological agents, such as immunomodulators or antioxidants, to tackle multifaceted diseases like neurodegeneration or cancer [450]. These multi-target regimens could not only boost the therapeutic impact but also mitigate resistance or relapse [451]. High-priority experiments will likely involve both in vivo and ex vivo models to confirm mechanistic pathways, refine optimal dosing, and uncover synergistic effects [452]. Biomarker-driven assessments that correlate molecular actions with patient outcomes will be vital, offering a clearer rationale for translating preclinical successes into human studies [453]. Ultimately, refining structural features while embracing combination strategies provides a balanced roadmap for researchers eager to unlock the full therapeutic potential of this diverse molecular class.
Each discussion subsection effectively acts as a mini-review that weaves together foundational knowledge, underscores clinical significance, and illuminates new directions. By clustering these quinoline-based compounds according to their structural traits or metabolic origins—be they endogenous, synthetic, or analogues—the overarching narrative becomes clearer. Patterns in receptor engagement, bioavailability, and therapeutic windows emerge, helping researchers pinpoint critical design features for improvement [454,455,456]. This bird’s-eye perspective also reveals shared challenges, such as scalability and off-target toxicity, which might impede clinical translation [457]. Yet, these pitfalls can guide smarter modifications in functional groups or combined treatment strategies, driving progress [455]. Ultimately, evaluating these diverse compounds side by side creates a roadmap of what works, what fails, and how the field can advance. The collaborative spirit behind collecting these insights promises to transform individual leads into robust, evidence-based interventions grounded in the science of quinoline-based therapeutics. Harnessing these consolidated findings can chart new approaches for KYNA-inspired strategies. Moving forward, researchers should aim to test these compounds in a broad range of disease models, thoroughly assess their long-term safety, and employ biomarker-guided patient selection to personalize treatments. By merging refined chemical modifications with cutting-edge formulation strategies, it becomes possible to fully exploit the immunomodulatory, neuroprotective, and anticancer promise of this emerging quinoline class.

7. Conclusions

The structural complexity of quinoline-based metabolites underpins their remarkable versatility across neurodegenerative, inflammatory, and oncological contexts, thereby making them prime candidates for drug development. This review has shown how carefully tuned substitutions—chlorination, fluorination, and esterification—can bolster receptor affinity, metabolic stability, and bioavailability, as demonstrated by derivatives, such as 7-CKA (enhanced NMDA blockade) and SZR-81 (improved oral absorption and GPR35 selectivity). Nonetheless, ongoing hurdles—limited BBB penetration, off-target effects (such as SZR-105’s kinase inhibition), and rapid clearance (e.g., SZR-73)—show that minor modifications may not fully realize these compounds’ therapeutic potential. Moving beyond basic prodrug strategies, a more integrative approach is warranted—one that simultaneously harnesses advanced computational modeling, combinatorial screening, and emerging research on epigenetic modulators. For instance, coupling KYNA analogues with IDO inhibitors could offer synergistic regulation of the KYN pathway, while multi-target ligands, like tasquinimod, highlight the promise of convergent pharmacological mechanisms. Further, employing novel formulation technologies (e.g., nanoparticle-based delivery) and improved in vivo models can strengthen the translational accuracy, fostering therapies that more closely match human pathophysiology. Ultimately, it is by weaving these interdisciplinary strategies into the existing foundation of SAR insights that the next generation of quinoline-based interventions will realize their broad therapeutic potential.

8. Patents

P1500356; PCT/HU2017?000014; EP17759330.8A; US15/082,099: WO 2017149333; A1 2017090. P1500356; PCT/HU2016/050034; US 10,857,236B2 (2020); WO2017021748 (EU).

Author Contributions

Conceptualization, M.T. and L.V.; methodology, M.T.; software, M.T.; validation, M.T.; formal analysis, M.T.; investigation, M.T.; resources, M.T.; data curation, M.T.; writing—original draft preparation, M.T.; writing—review and editing, M.T., I.S. and L.V.; visualization, M.T.; supervision, L.V.; project administration, L.V.; funding acquisition, L.V. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Research, Development, and Innovation Office—NKFIH K138125, SZTE SZAOK-KKA No: 2022/5S729, and the HUN-REN Hungarian Research Network.

Acknowledgments

The graphical abstract was created using biorender.com.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

BBBblood–brain barrier
7-CKA7-chlorokynurenic acid
DPAdipicolinic acid
GPR35G-protein-coupled receptor 35
HDAChistone deacetylase
IDOsindoleamine 2,3-dioxygenases
INAisonicotinic acid
KMOkynurenine 3-monooxygenase
KYNkynurenine
KYNAkynurenic acid
NAnicotinic acid
NMDAN-methyl-D-aspartate
QUINquinolinic acid
SARstructure–activity relationship
Trptryptophan
TDOtryptophan 2,3-dioxygenase
XAxanthurenic acid

References

  1. Stone, T.W.; Williams, R.O. Tryptophan metabolism as a ‘reflex’ feature of neuroimmune communication: Sensor and effector functions for the indoleamine-2,3-dioxygenase kynurenine pathway. J. Neurochem. 2024, 168, 3333–3357. [Google Scholar] [CrossRef]
  2. Lovelace, M.D.; Varney, B.; Sundaram, G.; Lennon, M.J.; Lim, C.K.; Jacobs, K.; Guillemin, G.J.; Brew, B.J. Recent evidence for an expanded role of the kynurenine pathway of tryptophan metabolism in neurological diseases. Neuropharmacology 2017, 112, 373–388. [Google Scholar] [CrossRef]
  3. Cervenka, I.; Agudelo, L.Z.; Ruas, J.L. Kynurenines: Tryptophan’s metabolites in exercise, inflammation, and mental health. Science 2017, 357, eaaf9794. [Google Scholar] [CrossRef] [PubMed]
  4. Roth, W.; Zadeh, K.; Vekariya, R.; Ge, Y.; Mohamadzadeh, M. Tryptophan metabolism and gut-brain homeostasis. Int. J. Mol. Sci. 2021, 22, 2973. [Google Scholar] [CrossRef] [PubMed]
  5. Li, D.; Yu, S.; Long, Y.; Shi, A.; Deng, J.; Ma, Y.; Wen, J.; Li, X.; Liu, S.; Zhang, Y. Tryptophan metabolism: Mechanism-oriented therapy for neurological and psychiatric disorders. Front. Immunol. 2022, 13, 985378. [Google Scholar] [CrossRef]
  6. Badawy, A.A. Kynurenine pathway of tryptophan metabolism: Regulatory and functional aspects. Int. J. Tryptophan Res. 2017, 10, 1178646917691938. [Google Scholar] [CrossRef] [PubMed]
  7. Wang, Q.; Liu, D.; Song, P.; Zou, M.-H. Deregulated tryptophan-kynurenine pathway is linked to inflammation, oxidative stress, and immune activation pathway in cardiovascular diseases. Front. Biosci. 2015, 20, 1116. [Google Scholar]
  8. Tanaka, M.; Szabó, Á.; Vécsei, L. Redefining roles: A paradigm shift in tryptophan–kynurenine metabolism for innovative clinical applications. Int. J. Mol. Sci. 2024, 25, 12767. [Google Scholar] [CrossRef]
  9. Zhao, Y.-Q.; Li, X.; Guo, H.-Y.; Shen, Q.-K.; Quan, Z.-S.; Luan, T. Application of quinoline ring in structural modification of natural products. Molecules 2023, 28, 6478. [Google Scholar] [CrossRef]
  10. Ajani, O.O.; Iyaye, K.T.; Ademosun, O.T. Recent advances in chemistry and therapeutic potential of functionalized quinoline motifs—A review. RSC Adv. 2022, 12, 18594–18614. [Google Scholar] [CrossRef]
  11. Sehlangia, S.; Nayak, N.; Garg, N.; Pradeep, C.P. Substituent-controlled structural, supramolecular, and cytotoxic properties of a series of 2-Styryl-8-nitro and 2-Styryl-8-hydroxy quinolines. ACS Omega 2022, 7, 24838–24850. [Google Scholar] [CrossRef] [PubMed]
  12. Hernández-Ayala, L.F.; Guzmán-López, E.G.; Galano, A. Quinoline Derivatives: Promising Antioxidants with Neuroprotective Potential. Antioxidants 2023, 12, 1853. [Google Scholar] [CrossRef] [PubMed]
  13. Martorana, A.; La Monica, G.; Lauria, A. Quinoline-based molecules targeting c-Met, EGF, and VEGF receptors and the proteins involved in related carcinogenic pathways. Molecules 2020, 25, 4279. [Google Scholar] [CrossRef]
  14. Shoji, T.; Takeuchi, M.; Uda, M.; Ariga, Y.; Yamazaki, A.; Sekiguchi, R.; Ito, S. Synthesis of Azuleno [2,1-b] quinolones and Quinolines via Brønsted Acid-Catalyzed Cyclization of 2-Arylaminoazulenes. Molecules 2023, 28, 5785. [Google Scholar] [CrossRef]
  15. Yadav, V.; Reang, J.; Sharma, V.; Majeed, J.; Sharma, P.C.; Sharma, K.; Giri, N.; Kumar, A.; Tonk, R.K. Quinoline-derivatives as privileged scaffolds for medicinal and pharmaceutical chemists: A comprehensive review. Chem. Biol. Drug Des. 2022, 100, 389–418. [Google Scholar] [CrossRef]
  16. Lanz, T.V.; Williams, S.K.; Stojic, A.; Iwantscheff, S.; Sonner, J.K.; Grabitz, C.; Becker, S.; Böhler, L.-I.; Mohapatra, S.R.; Sahm, F. Tryptophan-2, 3-Dioxygenase (TDO) deficiency is associated with subclinical neuroprotection in a mouse model of multiple sclerosis. Sci. Rep. 2017, 7, 41271. [Google Scholar] [CrossRef]
  17. Maya, S.; Prakash, T.; Goli, D. Effect of wedelolactone and gallic acid on quinolinic acid-induced neurotoxicity and impaired motor function: Significance to sporadic amyotrophic lateral sclerosis. Neurotoxicology 2018, 68, 1–12. [Google Scholar]
  18. Platten, M.; Friedrich, M.; Wainwright, D.A.; Panitz, V.; Opitz, C.A. Tryptophan metabolism in brain tumors—IDO and beyond. Curr. Opin. Immunol. 2021, 70, 57–66. [Google Scholar] [CrossRef]
  19. Lovelace, M.D.; Varney, B.; Sundaram, G.; Franco, N.F.; Ng, M.L.; Pai, S.; Lim, C.K.; Guillemin, G.J.; Brew, B.J. Current evidence for a role of the kynurenine pathway of tryptophan metabolism in multiple sclerosis. Front. Immunol. 2016, 7, 246. [Google Scholar] [CrossRef]
  20. Krause, D.; Suh, H.S.; Tarassishin, L.; Cui, Q.L.; Durafourt, B.A.; Choi, N.; Bauman, A.; Cosenza-Nashat, M.; Antel, J.P.; Zhao, M.L.; et al. The tryptophan metabolite 3-hydroxyanthranilic acid plays anti-inflammatory and neuroprotective roles during inflammation: Role of hemeoxygenase-1. Am. J. Pathol. 2011, 179, 1360–1372. [Google Scholar] [CrossRef]
  21. Zádori, D.; Veres, G.; Szalárdy, L.; Klivényi, P.; Vécsei, L. Alzheimer’s disease: Recent concepts on the relation of mitochondrial disturbances, excitotoxicity, neuroinflammation, and kynurenines. J. Alzheimer’s Dis. 2018, 62, 523–547. [Google Scholar] [CrossRef]
  22. Phillips, R.S.; Iradukunda, E.C.; Hughes, T.; Bowen, J.P. Modulation of enzyme activity in the kynurenine pathway by kynurenine monooxygenase inhibition. Front. Mol. Biosci. 2019, 6, 3. [Google Scholar] [CrossRef] [PubMed]
  23. Valotto Neto, L.J.; Reverete de Araujo, M.; Moretti Junior, R.C.; Mendes Machado, N.; Joshi, R.K.; Dos Santos Buglio, D.; Barbalho Lamas, C.; Direito, R.; Fornari Laurindo, L.; Tanaka, M.; et al. Investigating the Neuroprotective and Cognitive-Enhancing Effects of Bacopa monnieri: A Systematic Review Focused on Inflammation, Oxidative Stress, Mitochondrial Dysfunction, and Apoptosis. Antioxidants 2024, 13, 393. [Google Scholar] [CrossRef] [PubMed]
  24. Michaudel, C.; Danne, C.; Agus, A.; Magniez, A.; Aucouturier, A.; Spatz, M.; Lefevre, A.; Kirchgesner, J.; Rolhion, N.; Wang, Y. Rewiring the altered tryptophan metabolism as a novel therapeutic strategy in inflammatory bowel diseases. Gut 2023, 72, 1296–1307. [Google Scholar] [CrossRef]
  25. Opitz, C.A.; Somarribas Patterson, L.F.; Mohapatra, S.R.; Dewi, D.L.; Sadik, A.; Platten, M.; Trump, S. The therapeutic potential of targeting tryptophan catabolism in cancer. Br. J. Cancer 2020, 122, 30–44. [Google Scholar] [CrossRef]
  26. Tang, K.; Wu, Y.-H.; Song, Y.; Yu, B. Indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors in clinical trials for cancer immunotherapy. J. Hematol. Oncol. 2021, 14, 68. [Google Scholar] [CrossRef] [PubMed]
  27. El-Zahabi, M.A.A.; Abouzeid, M.Y.; Eissa, I.H.; Taghour, M.S. Dual Inhibitors of Indoleamine-2,3-Dioxygenase (IDO) and Tryptophan-2,3-Dioxygenase (TDO) as Anti-Tumor Immune Modulators. Al-Azhar J. Pharm. Sci. 2024, 69, 38–61. [Google Scholar] [CrossRef]
  28. Plitman, E.; Iwata, Y.; Caravaggio, F.; Nakajima, S.; Chung, J.K.; Gerretsen, P.; Kim, J.; Takeuchi, H.; Chakravarty, M.M.; Remington, G. Kynurenic acid in schizophrenia: A systematic review and meta-analysis. Schizophr. Bull. 2017, 43, 764–777. [Google Scholar] [CrossRef]
  29. Yang, P.; Zhang, J. Indoleamine 2,3-dioxygenase (IDO) activity: A perspective biomarker for laboratory determination in tumor immunotherapy. Biomedicines 2023, 11, 1988. [Google Scholar] [CrossRef]
  30. Stone, T.W. Does kynurenic acid act on nicotinic receptors? An assessment of the evidence. J. Neurochem. 2020, 152, 627–649. [Google Scholar] [CrossRef]
  31. Fukuwatari, T. Possibility of amino acid treatment to prevent the psychiatric disorders via modulation of the production of tryptophan metabolite kynurenic acid. Nutrients 2020, 12, 1403. [Google Scholar] [CrossRef] [PubMed]
  32. Bauzon, J.; Lee, G.; Cummings, J. Repurposed agents in the Alzheimer’s disease drug development pipeline. Alzheimer’s Res. Ther. 2020, 12, 98. [Google Scholar] [CrossRef]
  33. Saranraj, K.; Kiran, P.U. Drug repurposing: Clinical practices and regulatory pathways. Perspect. Clin. Res. 2025, 16, 61–68. [Google Scholar] [CrossRef]
  34. Patrignani, P.; Contursi, A.; Tacconelli, S.; Steinhilber, D. The future of pharmacology and therapeutics of the arachidonic acid cascade in the next decade: Innovative advancements in drug repurposing. Front. Pharmacol. 2024, 15, 1472396. [Google Scholar] [CrossRef]
  35. Panchal, N.B.; Vaghela, V.M. From Molecules to Medicine: The Remarkable Pharmacological Odyssey of Quinoline and It’s Derivatives. Orient. J. Chem. 2023, 39, 546–567. [Google Scholar] [CrossRef]
  36. Atukuri, D.; Vijayalaxmi, S.; Sanjeevamurthy, R.; Vidya, L.; Prasannakumar, R.; Raghavendra, M. Identification of quinoline-chalcones and heterocyclic chalcone-appended quinolines as broad-spectrum pharmacological agents. Bioorganic Chem. 2020, 105, 104419. [Google Scholar] [CrossRef]
  37. Elebiju, O.F.; Ajani, O.O.; Oduselu, G.O.; Ogunnupebi, T.A.; Adebiyi, E. Recent advances in functionalized quinoline scaffolds and hybrids—Exceptional pharmacophore in therapeutic medicine. Front. Chem. 2023, 10, 1074331. [Google Scholar] [CrossRef]
  38. Fukushima, T.; Umino, M.; Sakamoto, T.; Onozato, M. A review of chromatographic methods for bioactive tryptophan metabolites, kynurenine, kynurenic acid, quinolinic acid, and others, in biological fluids. Biomed. Chromatogr. 2022, 36, e5308. [Google Scholar] [CrossRef]
  39. Ostapiuk, A.; Urbanska, E.M. Kynurenic acid in neurodegenerative disorders—Unique neuroprotection or double-edged sword? CNS Neurosci. Ther. 2022, 28, 19–35. [Google Scholar] [CrossRef]
  40. Biernacki, T.; Sandi, D.; Bencsik, K.; Vécsei, L. Kynurenines in the pathogenesis of multiple sclerosis: Therapeutic perspectives. Cells 2020, 9, 1564. [Google Scholar] [CrossRef]
  41. Vécsei, L.; Szalárdy, L.; Fülöp, F.; Toldi, J. Kynurenines in the CNS: Recent advances and new questions. Nat. Rev. Drug Discov. 2013, 12, 64–82. [Google Scholar] [CrossRef] [PubMed]
  42. Mor, A.; Tankiewicz-Kwedlo, A.; Krupa, A.; Pawlak, D. Role of kynurenine pathway in oxidative stress during neurodegenerative disorders. Cells 2021, 10, 1603. [Google Scholar] [CrossRef] [PubMed]
  43. Houldsworth, A. Role of oxidative stress in neurodegenerative disorders: A review of reactive oxygen species and prevention by antioxidants. Brain Commun. 2024, 6, fcad356. [Google Scholar] [CrossRef]
  44. Liu, Z.; Zhou, T.; Ziegler, A.C.; Dimitrion, P.; Zuo, L. Oxidative stress in neurodegenerative diseases: From molecular mechanisms to clinical applications. Oxidative Med. Cell. Longev. 2017, 2017, 2525967. [Google Scholar] [CrossRef]
  45. Olufunmilayo, E.O.; Gerke-Duncan, M.B.; Holsinger, R.D. Oxidative stress and antioxidants in neurodegenerative disorders. Antioxidants 2023, 12, 517. [Google Scholar] [CrossRef]
  46. Tóth, F.; Cseh, E.K.; Vécsei, L. Natural molecules and neuroprotection: Kynurenic acid, pantethine and α-lipoic acid. Int. J. Mol. Sci. 2021, 22, 403. [Google Scholar] [CrossRef] [PubMed]
  47. Ferreira, F.S.; Schmitz, F.; Marques, E.P.; Siebert, C.; Wyse, A.T. Intrastriatal quinolinic acid administration impairs redox homeostasis and induces inflammatory changes: Prevention by kynurenic acid. Neurotox. Res. 2020, 38, 50–58. [Google Scholar] [CrossRef]
  48. Maitre, M.; Taleb, O.; Jeltsch-David, H.; Klein, C.; Mensah-Nyagan, A.G. Xanthurenic acid: A role in brain intercellular signaling. J. Neurochem. 2024, 168, 2303–2315. [Google Scholar] [CrossRef]
  49. Skorobogatov, K.; Autier, V.; Foiselle, M.; Richard, J.-R.; Boukouaci, W.; Wu, C.-L.; Raynal, S.; Carbonne, C.; Laukens, K.; Meysman, P. Kynurenine pathway abnormalities are state-specific but not diagnosis-specific in schizophrenia and bipolar disorder. Brain Behav. Immun. Health 2023, 27, 100584. [Google Scholar] [CrossRef]
  50. Bonaparte, J.; Cook, M.; Dixon, S.; Wright, C.; Hill, W.; Wang, G. Kynurenine Metabolites Induce Microglial Cell Senescence and Stimulate Neuroinflammation. Innov. Aging 2023, 7, 1073. [Google Scholar] [CrossRef]
  51. Chen, C.-M.; Huang, C.-Y.; Lai, C.-H.; Chen, Y.-C.; Hwang, Y.-T.; Lin, C.-Y. Neuroprotection effects of kynurenic acid-loaded micelles for the Parkinson’s disease models. J. Liposome Res. 2024, 34, 593–604. [Google Scholar] [CrossRef] [PubMed]
  52. Hertelendy, P.; Toldi, J.; Fülöp, F.; Vécsei, L. Ischemic stroke and kynurenines: Medicinal chemistry aspects. Curr. Med. Chem. 2018, 25, 5945–5957. [Google Scholar] [CrossRef]
  53. Martín-Hernández, D.; Tendilla-Beltrán, H.; Madrigal, J.L.; García-Bueno, B.; Leza, J.C.; Caso, J.R. Chronic mild stress alters kynurenine pathways changing the glutamate neurotransmission in frontal cortex of rats. Mol. Neurobiol. 2019, 56, 490–501. [Google Scholar] [CrossRef]
  54. Wirthgen, E.; Hoeflich, A.; Rebl, A.; Günther, J. Kynurenic acid: The Janus-faced role of an immunomodulatory tryptophan metabolite and its link to pathological conditions. Front. Immunol. 2018, 8, 1957. [Google Scholar] [CrossRef]
  55. Savitz, J. The kynurenine pathway: A finger in every pie. Mol. Psychiatry 2020, 25, 131–147. [Google Scholar] [CrossRef] [PubMed]
  56. Molnár, K.; Lőrinczi, B.; Fazakas, C.; Szatmári, I.; Fülöp, F.; Kmetykó, N.; Berkecz, R.; Ilisz, I.; Krizbai, I.A.; Wilhelm, I. Szr-104, a novel kynurenic acid analogue with high permeability through the blood–brain barrier. Pharmaceutics 2021, 13, 61. [Google Scholar] [CrossRef] [PubMed]
  57. Juhász, Á.; Ungor, D.; Varga, N.; Katona, G.; Balogh, G.T.; Csapó, E. Lipid-Based nanocarriers for delivery of neuroprotective kynurenic acid: Preparation, characterization, and BBB transport. Int. J. Mol. Sci. 2023, 24, 14251. [Google Scholar] [CrossRef]
  58. Varga, N.; Csapó, E.; Majláth, Z.; Ilisz, I.; Krizbai, I.A.; Wilhelm, I.; Knapp, L.; Toldi, J.; Vécsei, L.; Dékány, I. Targeting of the kynurenic acid across the blood–brain barrier by core-shell nanoparticles. Eur. J. Pharm. Sci. 2016, 86, 67–74. [Google Scholar] [CrossRef]
  59. Hara, N.; Yamada, K.; Shibata, T.; Osago, H.; Hashimoto, T.; Tsuchiya, M. Elevation of cellular NAD levels by nicotinic acid and involvement of nicotinic acid phosphoribosyltransferase in human cells. J. Biol. Chem. 2007, 282, 24574–24582. [Google Scholar] [CrossRef]
  60. Xiao, W.; Wang, R.-S.; Handy, D.E.; Loscalzo, J. NAD (H) and NADP (H) redox couples and cellular energy metabolism. Antioxid. Redox Signal. 2018, 28, 251–272. [Google Scholar] [CrossRef]
  61. Oyama, T.; Yamamoto, T.; Kameda, T.; Kamiya, T.; Abe, H.; Abe, T.; Tanuma, S.-I. Supplementation of nicotinic acid and its derivatives up-regulates cellular NAD+ level rather than nicotinamide derivatives in cultured normal human epidermal keratinocytes. Life 2024, 14, 413. [Google Scholar] [CrossRef] [PubMed]
  62. Carlson, L.A. Nicotinic acid: The broad-spectrum lipid drug. A 50th anniversary review. J. Intern. Med. 2005, 258, 94–114. [Google Scholar] [CrossRef]
  63. Schachter, M. Strategies for modifying high-density lipoprotein cholesterol: A role for nicotinic acid. Cardiovasc. Drugs Ther. 2005, 19, 415–422. [Google Scholar] [CrossRef] [PubMed]
  64. Figge, H.L.; Figge, J.; Souney, P.F.; Mutnick, A.H.; Sacks, F. Nicotinic acid: A review of its clinical use in the treatment of lipid disorders. Pharmacother. J. Hum. Pharmacol. Drug Ther. 1988, 8, 287–294. [Google Scholar] [CrossRef]
  65. Cheng, K.; Wu, T.-J.; Wu, K.K.; Sturino, C.; Metters, K.; Gottesdiener, K.; Wright, S.D.; Wang, Z.; O’Neill, G.; Lai, E. Antagonism of the prostaglandin D2 receptor 1 suppresses nicotinic acid-induced vasodilation in mice and humans. Proc. Natl. Acad. Sci. USA 2006, 103, 6682–6687. [Google Scholar] [CrossRef]
  66. Benyo, Z.; Gille, A.; Bennett, C.L.; Clausen, B.E.; Offermanns, S. Nicotinic acid-induced flushing is mediated by activation of epidermal langerhans cells. Mol. Pharmacol. 2006, 70, 1844–1849. [Google Scholar] [CrossRef]
  67. Benyó, Z.; Gille, A.; Kero, J.; Csiky, M.; Suchánková, M.C.; Nüsing, R.M.; Moers, A.; Pfeffer, K.; Offermanns, S. GPR109A (PUMA-G/HM74A) mediates nicotinic acid–induced flushing. J. Clin. Investig. 2005, 115, 3634–3640. [Google Scholar] [CrossRef]
  68. Braidy, N.; Grant, R.; Adams, S.; Brew, B.J.; Guillemin, G.J. Mechanism for quinolinic acid cytotoxicity in human astrocytes and neurons. Neurotox. Res. 2009, 16, 77–86. [Google Scholar] [CrossRef] [PubMed]
  69. Lugo-Huitrón, R.; Ugalde Muñiz, P.; Pineda, B.; Pedraza-Chaverrí, J.; Ríos, C.; Pérez-de la Cruz, V. Quinolinic acid: An endogenous neurotoxin with multiple targets. Oxidative Med. Cell. Longev. 2013, 2013, 104024. [Google Scholar] [CrossRef]
  70. Schurr, A.; West, C.A.; Rigor, B. Neurotoxicity of quinolinic acid and its derivatives in hypoxic rat hippocampal slices. Brain Res. 1991, 568, 199–204. [Google Scholar] [CrossRef]
  71. La Cruz, V.P.-D.; Carrillo-Mora, P.; Santamaría, A. Quinolinic acid, an endogenous molecule combining excitotoxicity, oxidative stress and other toxic mechanisms. Int. J. Tryptophan Res. 2012, 5, IJTR.S8158. [Google Scholar] [CrossRef] [PubMed]
  72. Leipnitz, G.; Schumacher, C.; Scussiato, K.; Dalcin, K.B.; Wannmacher, C.M.; Wyse, A.T.; Dutra-Filho, C.S.; Wajner, M.; Latini, A. Quinolinic acid reduces the antioxidant defenses in cerebral cortex of young rats. Int. J. Dev. Neurosci. 2005, 23, 695–701. [Google Scholar] [CrossRef] [PubMed]
  73. Pérez-Severiano, F.; Rodríguez-Pérez, M.; Pedraza-Chaverrí, J.; Maldonado, P.D.; Medina-Campos, O.N.; Ortíz-Plata, A.; Sánchez-García, A.; Villeda-Hernández, J.; Galván-Arzate, S.; Aguilera, P. S-Allylcysteine, a garlic-derived antioxidant, ameliorates quinolinic acid-induced neurotoxicity and oxidative damage in rats. Neurochem. Int. 2004, 45, 1175–1183. [Google Scholar] [CrossRef]
  74. Schwarcz, R.; Whetsell, W.O., Jr.; Mangano, R.M. Quinolinic acid: An endogenous metabolite that produces axon-sparing lesions in rat brain. Science 1983, 219, 316–318. [Google Scholar] [CrossRef] [PubMed]
  75. Hestad, K.; Alexander, J.; Rootwelt, H.; Aaseth, J.O. The role of tryptophan dysmetabolism and quinolinic acid in depressive and neurodegenerative diseases. Biomolecules 2022, 12, 998. [Google Scholar] [CrossRef]
  76. Seal, C.J.; Heaton, F.W. Effect of dietary picolinic acid on the metabolism of exogenous and endogenous zinc in the rat. J. Nutr. 1985, 115, 986–993. [Google Scholar] [CrossRef]
  77. Testa, U.; Louache, F.; Titeux, M.; Thomopoulos, P.; Rochant, H. The iron-chelating agent picolinic acid enhances transferrin receptors expression in human erythroleukaemic cell lines. Br. J. Haematol. 1985, 60, 491–502. [Google Scholar] [CrossRef]
  78. Prodinger, J.; Loacker, L.J.; Schmidt, R.L.; Ratzinger, F.; Greiner, G.; Witzeneder, N.; Hoermann, G.; Jutz, S.; Pickl, W.F.; Steinberger, P. The tryptophan metabolite picolinic acid suppresses proliferation and metabolic activity of CD4+ T cells and inhibits c-Myc activation. J. Leucoc. Biol. 2016, 99, 583–594. [Google Scholar] [CrossRef]
  79. Tomioka, H.; Shimizu, T.; Tatano, Y. Effects of picolinic acid on the antimicrobial functions of host macrophages against Mycobacterium avium complex. Int. J. Antimicrob. Agents 2007, 29, 460–464. [Google Scholar] [CrossRef]
  80. Cai, S.; Sato, K.; Shimizu, T.; Yamabe, S.; Hiraki, M.; Sano, C.; Tomioka, H. Antimicrobial activity of picolinic acid against extracellular and intracellular Mycobacterium avium complex and its combined activity with clarithromycin, rifampicin and fluoroquinolones. J. Antimicrob. Chemother. 2006, 57, 85–93. [Google Scholar] [CrossRef]
  81. Bosco, M.C.; Rapisarda, A.; Massazza, S.; Melillo, G.; Young, H.; Varesio, L. The tryptophan catabolite picolinic acid selectively induces the chemokines macrophage inflammatory protein-1α and-1β in macrophages. J. Immunol. 2000, 164, 3283–3291. [Google Scholar] [CrossRef] [PubMed]
  82. Grant, R.; Coggan, S.; Smythe, G.A. The physiological action of picolinic acid in the human brain. Int. J. Tryptophan Res. 2009, 2, IJTR.S2469. [Google Scholar] [CrossRef] [PubMed]
  83. Cockhill, J.; Jhamandas, K.; Boegman, R.; Beninger, R. Action of picolinic acid and structurally related pyridine carboxylic acids on quinolinic acid-induced cortical cholinergic damage. Brain Res. 1992, 599, 57–63. [Google Scholar] [CrossRef] [PubMed]
  84. Kalisch, B.E.; Jhamandas, K.; Boegman, R.J.; Beninger, R.J. Picolinic acid protects against quinolinic acid-induced depletion of NADPH diaphorase containing neurons in the rat striatum. Brain Res. 1994, 668, 1–8. [Google Scholar] [CrossRef]
  85. Taleb, O.; Maammar, M.; Klein, C.; Maitre, M.; Mensah-Nyagan, A.G. A role for xanthurenic acid in the control of brain dopaminergic activity. Int. J. Mol. Sci. 2021, 22, 6974. [Google Scholar] [CrossRef]
  86. Fazio, F.; Lionetto, L.; Curto, M.; Iacovelli, L.; Copeland, C.S.; Neale, S.A.; Bruno, V.; Battaglia, G.; Salt, T.E.; Nicoletti, F. Cinnabarinic acid and xanthurenic acid: Two kynurenine metabolites that interact with metabotropic glutamate receptors. Neuropharmacology 2017, 112, 365–372. [Google Scholar] [CrossRef]
  87. Murakami, K.; Haneda, M.; Yoshino, M. Prooxidant action of xanthurenic acid and quinoline compounds: Role of transition metals in the generation of reactive oxygen species and enhanced formation of 8-hydroxy-2′-deoxyguanosine in DNA. Biometals 2006, 19, 429–435. [Google Scholar] [CrossRef]
  88. Murakami, K.; Ito, M.; Yoshino, M. Xanthurenic acid inhibits metal ion-induced lipid peroxidation and protects NADP-isocitrate dehydrogenase from oxidative inactivation. J. Nutr. Sci. Vitaminol. 2001, 47, 306–310. [Google Scholar] [CrossRef]
  89. Lima, V.L.; Dias, F.; Nunes, R.D.; Pereira, L.O.; Santos, T.S.; Chiarini, L.B.; Ramos, T.D.; Silva-Mendes, B.J.; Perales, J.; Valente, R.H. The antioxidant role of xanthurenic acid in the Aedes aegypti midgut during digestion of a blood meal. PLoS ONE 2012, 7, e38349. [Google Scholar] [CrossRef]
  90. Nowicka-Stążka, P.; Langner, E.; Turski, W.; Rzeski, W.; Parada-Turska, J. Quinaldic acid in synovial fluid of patients with rheumatoid arthritis and osteoarthritis and its effect on synoviocytes in vitro. Pharmacol. Rep. 2018, 70, 277–283. [Google Scholar] [CrossRef]
  91. Okamoto, H.; Miyamoto, S.; Mabuchi, H.; Yoneyama, Y.; Takeda, R. Insulin-releasing effect of quinaldic acid and its relatives on isolated Langerhans islets. Biochem. Biophys. Res. Commun. 1973, 53, 1297–1303. [Google Scholar] [CrossRef] [PubMed]
  92. Jhamandas, K.H.; Boegman, R.J.; Beninger, R.J.; Miranda, A.F.; Lipic, K.A. Excitotoxicity of quinolinic acid: Modulation by endogenous antagonists. Neurotox. Res. 2000, 2, 139–155. [Google Scholar] [CrossRef]
  93. Bala, M.; Radhakrishnan, T.; Kumar, A.; Mishra, G.P.; Dobraia, J.R.; Kirti, P.B. Erratum: Overexpression of a fusion defensin gene from radish and fenugreek improves resistance against leaf spot diseases caused by Cercospora arachidicola and Phaeoisariopsis personata in peanut. Turk. J. Biol. 2019, 43, 154. [Google Scholar] [CrossRef]
  94. Zheng, Q.; Wang, Q.; Wang, S.; Wu, J.; Gao, Q.; Liu, W. Thiopeptide Antibiotics Exhibit a Dual Mode of Action against Intracellular Pathogens by Affecting Both Host and Microbe. Chem. Biol. 2015, 22, 1002–1007. [Google Scholar] [CrossRef] [PubMed]
  95. Jhamandas, K.; Boegman, R.J.; Beninger, R.J.; Bialik, M. Quinolinate-induced cortical cholinergic damage: Modulation by tryptophan metabolites. Brain Res. 1990, 529, 185–191. [Google Scholar] [CrossRef] [PubMed]
  96. Anderson, E.W.; Fishbein, J.; Hong, J.; Roeser, J.; Furie, R.A.; Aranow, C.; Volpe, B.T.; Diamond, B.; Mackay, M. Quinolinic acid, a kynurenine/tryptophan pathway metabolite, associates with impaired cognitive test performance in systemic lupus erythematosus. Lupus Sci. Med. 2021, 8, e000559. [Google Scholar] [CrossRef]
  97. Sawada, Y.; Kayakiri, H.; Abe, Y.; Mizutani, T.; Inamura, N.; Asano, M.; Hatori, C.; Aramori, I.; Oku, T.; Tanaka, H. Discovery of the first non-peptide full agonists for the human bradykinin B2 receptor incorporating 4-(2-picolyloxy)quinoline and 1-(2-picolyl)benzimidazole frameworks. J. Med. Chem. 2004, 47, 2853–2863. [Google Scholar] [CrossRef]
  98. Antosiewicz, J.M.; Kane, P.M. Editorial: Intracellular Molecular Processes Affected by pH. Front. Mol. Biosci. 2022, 9, 891533. [Google Scholar] [CrossRef]
  99. Ghorai, P.; Pal, K.; Karmakar, P.; Saha, A. The development of two fluorescent chemosensors for the selective detection of Zn2+ and Al3+ ions in a quinoline platform by tuning the substituents in the receptor part: Elucidation of the structures of the metal-bound chemosensors and biological studies. Dalton Trans. 2020, 49, 4758–4773. [Google Scholar] [CrossRef]
  100. Mikata, Y. Quinoline- and isoquinoline-derived ligand design on TQEN (N,N,N′,N′-tetrakis(2-quinolylmethyl)ethylenediamine) platform for fluorescent sensing of specific metal ions and phosphate species. Dalton Trans. 2020, 49, 17494–17504. [Google Scholar] [CrossRef]
  101. Peterson, L.R. Quinolone molecular structure-activity relationships: What we have learned about improving antimicrobial activity. Clin. Infect. Dis. 2001, 33 (Suppl. S3), S180–S186. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, Y.; Xiao, F.; Jin, G. Structural basis of quinolone derivatives, inhibition of type I and II topoisomerases and inquiry into the relevance of bioactivity in odd or even branches with molecular docking study. J. Mol. Struct. 2020, 1221, 128869. [Google Scholar] [CrossRef]
  103. Cramer, J.; Sager, C.P.; Ernst, B. Hydroxyl Groups in Synthetic and Natural-Product-Derived Therapeutics: A Perspective on a Common Functional Group. J. Med. Chem. 2019, 62, 8915–8930. [Google Scholar] [CrossRef] [PubMed]
  104. Fray, M.J.; Bull, D.J.; Carr, C.L.; Gautier, E.C.; Mowbray, C.E.; Stobie, A. Structure-activity relationships of 1,4-dihydro-(1H,4H)-quinoxaline-2,3-diones as N-methyl-D-aspartate (glycine site) receptor antagonists. 1. Heterocyclic substituted 5-alkyl derivatives. J. Med. Chem. 2001, 44, 1951–1962. [Google Scholar] [CrossRef]
  105. Wei, L.; Hou, T.; Li, J.; Zhang, X.; Zhou, H.; Wang, Z.; Cheng, J.; Xiang, K.; Wang, J.; Zhao, Y.; et al. Structure-Activity Relationship Studies of Coumarin-like Diacid Derivatives as Human G Protein-Coupled Receptor-35 (hGPR35) Agonists and a Consequent New Design Principle. J. Med. Chem. 2021, 64, 2634–2647. [Google Scholar] [CrossRef]
  106. Hou, Z.; Vanecek, A.S.; Tepe, J.J.; Odom, A.L. Synthesis, structure, properties, and cytotoxicity of a (quinoline)RuCp+ complex. Dalton Trans. 2023, 52, 721–730. [Google Scholar] [CrossRef] [PubMed]
  107. Liu, J.Y.; Wellen, K.E. Advances into understanding metabolites as signaling molecules in cancer progression. Curr. Opin. Cell Biol. 2020, 63, 144–153. [Google Scholar] [CrossRef]
  108. Liloia, D.; Zamfira, D.A.; Tanaka, M.; Manuello, J.; Crocetta, A.; Keller, R.; Cozzolino, M.; Duca, S.; Cauda, F.; Costa, T. Disentangling the role of gray matter volume and concentration in autism spectrum disorder: A meta-analytic investigation of 25 years of voxel-based morphometry research. Neurosci. Biobehav. Rev. 2024, 164, 105791. [Google Scholar] [CrossRef]
  109. Boros, F.; Vécsei, L. Progress in the development of kynurenine and quinoline-3-carboxamide-derived drugs. Expert Opin. Investig. Drugs 2020, 29, 1223–1247. [Google Scholar] [CrossRef]
  110. Stone, T.W. Neuropharmacology of quinolinic and kynurenic acids. Pharmacol. Rev. 1993, 45, 309–379. [Google Scholar] [CrossRef]
  111. Stone, T.W. Kynurenines in the CNS: From endogenous obscurity to therapeutic importance. Prog. Neurobiol. 2001, 64, 185–218. [Google Scholar] [CrossRef]
  112. Stone, T.W.; Stoy, N.; Darlington, L.G. An expanding range of targets for kynurenine metabolites of tryptophan. Trends Pharmacol. Sci. 2013, 34, 136–143. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, Z.; Lamson, A.R.; Shelley, M.; Troyanskaya, O. Interpretable neural architecture search and transfer learning for understanding CRISPR-Cas9 off-target enzymatic reactions. Nat. Comput. Sci. 2023, 3, 1056–1066. [Google Scholar] [CrossRef] [PubMed]
  114. Vieira, P.A.; Shin, C.B.; Arroyo-Currás, N.; Ortega, G.; Li, W.; Keller, A.A.; Plaxco, K.W.; Kippin, T.E. Ultra-High-Precision, in-vivo Pharmacokinetic Measurements Highlight the Need for and a Route Toward More Highly Personalized Medicine. Front. Mol. Biosci. 2019, 6, 69. [Google Scholar] [CrossRef]
  115. Tanaka, M.; Vécsei, L. Monitoring the kynurenine system: Concentrations, ratios or what else? Adv. Clin. Exp. Med. 2021, 30, 775–778. [Google Scholar] [CrossRef] [PubMed]
  116. Zeng, M.; Yang, L.; He, D.; Li, Y.; Shi, M.; Zhang, J. Metabolic pathways and pharmacokinetics of natural medicines with low permeability. Drug Metab. Rev. 2017, 49, 464–476. [Google Scholar] [CrossRef]
  117. Al-Karagholi, M.A.M.; Hansen, J.M.; Abou-Kassem, D.; Hansted, A.K.; Ubhayasekera, K.; Bergquist, J.; Vécsei, L.; Jansen-Olesen, I.; Ashina, M. Phase 1 study to access safety, tolerability, pharmacokinetics, and pharmacodynamics of kynurenine in healthy volunteers. Pharmacol. Res. Perspect. 2021, 9, e00741. [Google Scholar] [CrossRef]
  118. Matada, B.S.; Pattanashettar, R.; Yernale, N.G. A comprehensive review on the biological interest of quinoline and its derivatives. Bioorg. Med. Chem. 2021, 32, 115973. [Google Scholar] [CrossRef]
  119. Solomon, V.R.; Lee, H. Quinoline as a privileged scaffold in cancer drug discovery. Curr. Med. Chem. 2011, 18, 1488–1508. [Google Scholar] [CrossRef]
  120. Siddique, S.; Hussain, K.; Shehzadi, N.; Arshad, M.; Arshad, M.N.; Iftikhar, S.; Saghir, F.; Shaukat, A.; Sarfraz, M.; Ahmed, N. Design, synthesis, biological evaluation and molecular docking studies of quinoline-anthranilic acid hybrids as potent anti-inflammatory drugs. Org. Biomol. Chem. 2024, 22, 3708–3724. [Google Scholar] [CrossRef]
  121. Arabiyat, S.; Alzoubi, A.; Al-Daghistani, H.; Al-Hiari, Y.; Kasabri, V.; Alkhateeb, R. Evaluation of Quinoline-Related Carboxylic Acid Derivatives as Prospective Differentially Antiproliferative, Antioxidative, and Anti-Inflammatory Agents. Chem. Biol. Drug Des. 2024, 104, e14615. [Google Scholar] [CrossRef] [PubMed]
  122. Liang, X.; Xie, Y.; Liu, X.; Xu, H.; Ren, H.; Tang, S.; Liu, Q.; Huang, M.; Shao, X.; Li, C.; et al. Discovery of Novel Imidazo[4,5-c]quinoline Derivatives to Treat Inflammatory Bowel Disease (IBD) by Inhibiting Multiple Proinflammatory Signaling Pathways and Restoring Intestinal Homeostasis. J. Med. Chem. 2022, 65, 11949–11969. [Google Scholar] [CrossRef]
  123. Shang, X.F.; Morris-Natschke, S.L.; Liu, Y.Q.; Guo, X.; Xu, X.S.; Goto, M.; Li, J.C.; Yang, G.Z.; Lee, K.H. Biologically active quinoline and quinazoline alkaloids part I. Med. Res. Rev. 2018, 38, 775–828. [Google Scholar] [CrossRef] [PubMed]
  124. Zhao, Y.; Zhang, C.; Liu, W.; Guo, Z.; Zhang, Y.; Wu, Y.; Wei, C.; Wu, J.; Yang, X. Quinolines: A Promising Heterocyclic Scaffold for Cancer Therapeutics. Curr. Med. Chem. 2025, 32, 958–973. [Google Scholar] [CrossRef]
  125. Park, L.T.; Kadriu, B.; Gould, T.D.; Zanos, P.; Greenstein, D.; Evans, J.W.; Yuan, P.; Farmer, C.A.; Oppenheimer, M.; George, J.M. A randomized trial of the N-methyl-d-aspartate receptor glycine site antagonist prodrug 4-chlorokynurenine in treatment-resistant depression. Int. J. Neuropsychopharmacol. 2020, 23, 417–425. [Google Scholar] [CrossRef] [PubMed]
  126. Heyes, M.P.; Saito, K.; Crowley, J.S.; Davis, L.E.; Demitrack, M.A.; Der, M.; Dilling, L.A.; Elia, J.; Kruesi, M.J.; Lackner, A.; et al. Quinolinic acid and kynurenine pathway metabolism in inflammatory and non-inflammatory neurological disease. Brain 1992, 115 Pt 5, 1249–1273. [Google Scholar] [CrossRef]
  127. Kadela-Tomanek, M.; Jastrzębska, M.; Chrobak, E.; Bębenek, E. Lipophilicity and ADMET Analysis of Quinoline-1,4-quinone Hybrids. Pharmaceutics 2022, 15, 34. [Google Scholar] [CrossRef] [PubMed]
  128. Kadela-Tomanek, M.; Jastrzębska, M.; Chrobak, E.; Bębenek, E.; Latocha, M. Hybrids of 1,4-Quinone with Quinoline Derivatives: Synthesis, Biological Activity, and Molecular Docking with DT-Diaphorase (NQO1). Molecules 2022, 27, 6206. [Google Scholar] [CrossRef]
  129. Platten, M.; Nollen, E.A.A.; Röhrig, U.F.; Fallarino, F.; Opitz, C.A. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat. Rev. Drug Discov. 2019, 18, 379–401. [Google Scholar] [CrossRef]
  130. Chiarugi, A.; Meli, E.; Moroni, F. Similarities and differences in the neuronal death processes activated by 3OH-kynurenine and quinolinic acid. J. Neurochem. 2001, 77, 1310–1318. [Google Scholar] [CrossRef]
  131. Yang, D.; Zhang, S.; Fang, X.; Guo, L.; Hu, N.; Guo, Z.; Li, X.; Yang, S.; He, J.C.; Kuang, C.; et al. N-Benzyl/Aryl Substituted Tryptanthrin as Dual Inhibitors of Indoleamine 2,3-Dioxygenase and Tryptophan 2,3-Dioxygenase. J. Med. Chem. 2019, 62, 9161–9174. [Google Scholar] [CrossRef] [PubMed]
  132. Dolšak, A.; Gobec, S.; Sova, M. Indoleamine and tryptophan 2,3-dioxygenases as important future therapeutic targets. Pharmacol. Ther. 2021, 221, 107746. [Google Scholar] [CrossRef]
  133. Barbalho, S.M.; Leme Boaro, B.; da Silva Camarinha Oliveira, J.; Patočka, J.; Barbalho Lamas, C.; Tanaka, M.; Laurindo, L.F. Molecular Mechanisms Underlying Neuroinflammation Intervention with Medicinal Plants: A Critical and Narrative Review of the Current Literature. Pharmaceuticals 2025, 18, 133. [Google Scholar] [CrossRef]
  134. Tanaka, M.; Vécsei, L. Monitoring the Redox Status in Multiple Sclerosis. Biomedicines 2020, 8, 406. [Google Scholar] [CrossRef] [PubMed]
  135. Löscher, W.; Gericke, B. Novel Intrinsic Mechanisms of Active Drug Extrusion at the Blood-Brain Barrier: Potential Targets for Enhancing Drug Delivery to the Brain? Pharmaceutics 2020, 12, 966. [Google Scholar] [CrossRef]
  136. Zuchero, Y.J.; Chen, X.; Bien-Ly, N.; Bumbaca, D.; Tong, R.K.; Gao, X.; Zhang, S.; Hoyte, K.; Luk, W.; Huntley, M.A.; et al. Discovery of Novel Blood-Brain Barrier Targets to Enhance Brain Uptake of Therapeutic Antibodies. Neuron 2016, 89, 70–82. [Google Scholar] [CrossRef] [PubMed]
  137. Terstappen, G.C.; Meyer, A.H.; Bell, R.D.; Zhang, W. Strategies for delivering therapeutics across the blood-brain barrier. Nat. Rev. Drug Discov. 2021, 20, 362–383. [Google Scholar] [CrossRef]
  138. Dounay, A.B.; Tuttle, J.B.; Verhoest, P.R. Challenges and Opportunities in the Discovery of New Therapeutics Targeting the Kynurenine Pathway. J. Med. Chem. 2015, 58, 8762–8782. [Google Scholar] [CrossRef]
  139. Shen, L.L.; Pernet, A.G. Mechanism of inhibition of DNA gyrase by analogues of nalidixic acid: The target of the drugs is DNA. Proc. Natl. Acad. Sci. USA 1985, 82, 307–311. [Google Scholar] [CrossRef]
  140. Walker, H.A.; Wilson, S.; Farrar, C.; Richardson, A.P. The reflex respiratory and circulatory actions of some cinchoninic acid derivatives and a number of unrelated compounds. J. Pharmacol. Exp. Ther. 1952, 104, 211–218. [Google Scholar] [CrossRef]
  141. de Lima, E.P.; Laurindo, L.F.; Catharin, V.C.S.; Direito, R.; Tanaka, M.; Jasmin Santos German, I.; Lamas, C.B.; Guiguer, E.L.; Araújo, A.C.; Fiorini, A.M.R. Polyphenols, Alkaloids, and Terpenoids Against Neurodegeneration: Evaluating the Neuroprotective Effects of Phytocompounds Through a Comprehensive Review of the Current Evidence. Metabolites 2025, 15, 124. [Google Scholar] [CrossRef] [PubMed]
  142. Herraiz, T.; Guillén, H.; González-Peña, D.; Arán, V.J. Antimalarial Quinoline Drugs Inhibit β-Hematin and Increase Free Hemin Catalyzing Peroxidative Reactions and Inhibition of Cysteine Proteases. Sci. Rep. 2019, 9, 15398. [Google Scholar] [CrossRef]
  143. Kapishnikov, S.; Staalsø, T.; Yang, Y.; Lee, J.; Pérez-Berná, A.J.; Pereiro, E.; Yang, Y.; Werner, S.; Guttmann, P.; Leiserowitz, L.; et al. Mode of action of quinoline antimalarial drugs in red blood cells infected by Plasmodium falciparum revealed in vivo. Proc. Natl. Acad. Sci. USA 2019, 116, 22946–22952. [Google Scholar] [CrossRef] [PubMed]
  144. Foley, M.; Tilley, L. Quinoline antimalarials: Mechanisms of action and resistance and prospects for new agents. Pharmacol. Ther. 1998, 79, 55–87. [Google Scholar] [CrossRef]
  145. Currie, J.P.; Brown, R.A.; Will, G. Observations on the treatment of rheumatoid arthritis with butazolidin. Ann. Rheum. Dis. 1953, 12, 88–94. [Google Scholar] [CrossRef]
  146. Gelfman, D.M. Reflections on quinine and its importance in dermatology today. Clin. Dermatol. 2021, 39, 900–903. [Google Scholar] [CrossRef] [PubMed]
  147. Mukusheva, G.K.; Zhasymbekova, A.R.; Seidakhmetova, R.B.; Nurkenov, O.A.; Akishina, E.A.; Petkevich, S.K.; Dikusar, E.A.; Potkin, V.I. Quinine Esters with 1,2-Azole, Pyridine and Adamantane Fragments. Molecules 2022, 27, 3476. [Google Scholar] [CrossRef]
  148. Man-Son-Hing, M.; Wells, G.; Lau, A. Quinine for nocturnal leg cramps: A meta-analysis including unpublished data. J. Gen. Intern. Med. 1998, 13, 600–606. [Google Scholar] [CrossRef]
  149. Gisselmann, G.; Alisch, D.; Welbers-Joop, B.; Hatt, H. Effects of Quinine, Quinidine and Chloroquine on Human Muscle Nicotinic Acetylcholine Receptors. Front. Pharmacol. 2018, 9, 1339. [Google Scholar] [CrossRef]
  150. Li, Z.; Jin, X.; Wu, T.; Huang, G.; Wu, K.; Lei, J.; Pan, X.; Yan, N. Structural Basis for Pore Blockade of the Human Cardiac Sodium Channel Nav1.5 by the Antiarrhythmic Drug Quinidine. Angew. Chem. Int. Ed. Engl. 2021, 60, 11474–11480. [Google Scholar] [CrossRef]
  151. Hill, R.J.; Duff, H.J.; Sheldon, R.S. Class I antiarrhythmic drug receptor: Biochemical evidence for state-dependent interaction with quinidine and lidocaine. Mol. Pharmacol. 1989, 36, 150–159. [Google Scholar] [CrossRef] [PubMed]
  152. Kodama, I.; Toyama, J.; Yamada, K. Open and inactivated sodium channel block by class-I antiarrhythmic drugs. Jpn. Heart J. 1986, 27 (Suppl. 1), 83–89. [Google Scholar] [CrossRef] [PubMed]
  153. Conn, H.L., Jr.; Luchi, R.J. Some Cellular and Metabolic Considerations Relating to the Action of Quinidine as a Prototype Antiarrhythmic Agent. Am. J. Med. 1964, 37, 685–699. [Google Scholar] [CrossRef]
  154. Yan, M.; Fan, P.; Shi, Y.; Feng, L.; Wang, J.; Zhan, G.; Li, B. Stereoselective Blockage of Quinidine and Quinine in the hERG Channel and the Effect of Their Rescue Potency on Drug-Induced hERG Trafficking Defect. Int. J. Mol. Sci. 2016, 17, 1648. [Google Scholar] [CrossRef]
  155. Brocks, D.R.; Mehvar, R. Stereoselectivity in the pharmacodynamics and pharmacokinetics of the chiral antimalarial drugs. Clin. Pharmacokinet. 2003, 42, 1359–1382. [Google Scholar] [CrossRef] [PubMed]
  156. Malik, M.; Camm, A.J. Evaluation of drug-induced QT interval prolongation: Implications for drug approval and labelling. Drug Saf. 2001, 24, 323–351. [Google Scholar] [CrossRef]
  157. Haeusler, I.L.; Chan, X.H.S.; Guérin, P.J.; White, N.J. The arrhythmogenic cardiotoxicity of the quinoline and structurally related antimalarial drugs: A systematic review. BMC Med. 2018, 16, 200. [Google Scholar] [CrossRef]
  158. Reiter, M.J.; Higgins, S.L.; Payne, A.G.; Mann, D.E. Effects of quinidine versus procainamide on the QT interval. Am. J. Cardiol. 1986, 58, 512–516. [Google Scholar] [CrossRef]
  159. Wang, X.; Zeng, Y.; Sheng, L.; Larson, P.; Liu, X.; Zou, X.; Wang, S.; Guo, K.; Ma, C.; Zhang, G.; et al. A Cinchona Alkaloid Antibiotic That Appears To Target ATP Synthase in Streptococcus pneumoniae. J. Med. Chem. 2019, 62, 2305–2332. [Google Scholar] [CrossRef]
  160. Boratyński, P.J.; Zielińska-Błajet, M.; Skarżewski, J. Cinchona Alkaloids-Derivatives and Applications. Alkaloids Chem. Biol. 2019, 82, 29–145. [Google Scholar] [CrossRef]
  161. Chajdaś, Z.; Kucharska, M.; Wesełucha-Birczyńska, A. Two-Dimensional Correlation Spectroscopy (2D-COS) Tracking of the Formation of Selected Transition Metal Compounds Cu(II) and Cd(II) with Cinchonine and Their Impact on Model Components of Erythrocytes. Appl. Spectrosc. 2024, 79, 37028241279434. [Google Scholar] [CrossRef] [PubMed]
  162. Jabłońska-Trypuć, A.; Wydro, U.; Wołejko, E.; Świderski, G.; Lewandowski, W. Biological Activity of New Cichoric Acid-Metal Complexes in Bacterial Strains, Yeast-Like Fungi, and Human Cell Cultures In Vitro. Nutrients 2020, 12, 154. [Google Scholar] [CrossRef]
  163. Ramić, A.; Skočibušić, M.; Odžak, R.; Čipak Gašparović, A.; Milković, L.; Mikelić, A.; Sović, K.; Primožič, I.; Hrenar, T. Antimicrobial Activity of Quasi-Enantiomeric Cinchona Alkaloid Derivatives and Prediction Model Developed by Machine Learning. Antibiotics 2021, 10, 659. [Google Scholar] [CrossRef] [PubMed]
  164. Parveen, S.; Maurya, N.; Meena, A.; Luqman, S. Cinchonine: A Versatile Pharmacological Agent Derived from Natural Cinchona Alkaloids. Curr. Top. Med. Chem. 2024, 24, 343–363. [Google Scholar] [CrossRef] [PubMed]
  165. Kalluraya, B.; Sreenivasa, S. Synthesis and pharmacological properties of some quinoline derivatives. Farmaco 1998, 53, 399–404. [Google Scholar] [CrossRef]
  166. Chen, C.R.; Malik, M.; Snyder, M.; Drlica, K. DNA gyrase and topoisomerase IV on the bacterial chromosome: Quinolone-induced DNA cleavage. J. Mol. Biol. 1996, 258, 627–637. [Google Scholar] [CrossRef]
  167. Drlica, K.; Zhao, X. DNA gyrase, topoisomerase IV, and the 4-quinolones. Microbiol. Mol. Biol. Rev. 1997, 61, 377–392. [Google Scholar] [CrossRef]
  168. Spencer, A.C.; Panda, S.S. DNA Gyrase as a Target for Quinolones. Biomedicines 2023, 11, 371. [Google Scholar] [CrossRef]
  169. Fu, H.G.; Li, Z.W.; Hu, X.X.; Si, S.Y.; You, X.F.; Tang, S.; Wang, Y.X.; Song, D.Q. Synthesis and Biological Evaluation of Quinoline Derivatives as a Novel Class of Broad-Spectrum Antibacterial Agents. Molecules 2019, 24, 548. [Google Scholar] [CrossRef]
  170. Khamkhenshorngphanuch, T.; Kulkraisri, K.; Janjamratsaeng, A.; Plabutong, N.; Thammahong, A.; Manadee, K.; Na Pombejra, S.; Khotavivattana, T. Synthesis and Antimicrobial Activity of Novel 4-Hydroxy-2-quinolone Analogs. Molecules 2020, 25, 3059. [Google Scholar] [CrossRef]
  171. Kania, A.; Tejchman, W.; Pawlak, A.M.; Mokrzyński, K.; Różanowski, B.; Musielak, B.M.; Greczek-Stachura, M. Preliminary Studies of Antimicrobial Activity of New Synthesized Hybrids of 2-Thiohydantoin and 2-Quinolone Derivatives Activated with Blue Light. Molecules 2022, 27, 1069. [Google Scholar] [CrossRef]
  172. Kostopoulou, I.; Tzani, A.; Chronaki, K.; Prousis, K.C.; Pontiki, E.; Hadjiplavlou-Litina, D.; Detsi, A. Novel Multi-Target Agents Based on the Privileged Structure of 4-Hydroxy-2-quinolinone. Molecules 2023, 29, 190. [Google Scholar] [CrossRef]
  173. Gach-Janczak, K.; Piekielna-Ciesielska, J.; Waśkiewicz, J.; Krakowiak, K.; Wtorek, K.; Janecka, A. Quinolin-4-ones: Methods of Synthesis and Application in Medicine. Molecules 2025, 30, 163. [Google Scholar] [CrossRef]
  174. Ahmed, M.; Kelley, S.O. Enhancing the Potency of Nalidixic Acid toward a Bacterial DNA Gyrase with Conjugated Peptides. ACS Chem. Biol. 2017, 12, 2563–2569. [Google Scholar] [CrossRef]
  175. Naeem, A.; Badshah, S.L.; Muska, M.; Ahmad, N.; Khan, K. The Current Case of Quinolones: Synthetic Approaches and Antibacterial Activity. Molecules 2016, 21, 268. [Google Scholar] [CrossRef] [PubMed]
  176. Fàbrega, A.; Madurga, S.; Giralt, E.; Vila, J. Mechanism of action of and resistance to quinolones. Microb. Biotechnol. 2009, 2, 40–61. [Google Scholar] [CrossRef] [PubMed]
  177. Fernandes, P.B. Mode of action, and in vitro and in vivo activities of the fluoroquinolones. J. Clin. Pharmacol. 1988, 28, 156–168. [Google Scholar] [CrossRef] [PubMed]
  178. Duckworth, G.J.; Williams, J.D. Frequency of appearance of resistant variants to norfloxacin and nalidixic acid. J. Antimicrob. Chemother. 1984, 13 (Suppl. B), 33–38. [Google Scholar] [CrossRef]
  179. Majalekar, P.P.; Shirote, P.J. Fluoroquinolones: Blessings or Curses. Curr. Drug Targets 2020, 21, 1354–1370. [Google Scholar] [CrossRef]
  180. Barlow, A.M. Nalidixic Acid in Infections of Urinary Tract. Laboratory and Clinical Investigations. Br. Med. J. 1963, 2, 1308–1310. [Google Scholar] [CrossRef]
  181. Reeves, D.S.; Lacey, R.W.; Mummery, R.V.; Mahendra, M.; Bint, A.J.; Newsom, S.W. Treatment of acute urinary infection by norfloxacin or nalidixic acid/citrate: A multi-centre comparative study. J. Antimicrob. Chemother. 1984, 13 (Suppl. B), 99–105. [Google Scholar] [CrossRef]
  182. Engle, E.C.; Manes, S.H.; Drlica, K. Differential effects of antibiotics inhibiting gyrase. J. Bacteriol. 1982, 149, 92–98. [Google Scholar] [CrossRef] [PubMed]
  183. Mizuuchi, K.; O’Dea, M.H.; Gellert, M. DNA gyrase: Subunit structure and ATPase activity of the purified enzyme. Proc. Natl. Acad. Sci. USA 1978, 75, 5960–5963. [Google Scholar] [CrossRef] [PubMed]
  184. Yang, H.L.; Heller, K.; Gellert, M.; Zubay, G. Differential sensitivity of gene expression in vitro to inhibitors of DNA gyrase. Proc. Natl. Acad. Sci. USA 1979, 76, 3304–3308. [Google Scholar] [CrossRef] [PubMed]
  185. Palma, E.; Tilocca, B.; Roncada, P. Antimicrobial resistance in veterinary medicine: An overview. Int. J. Mol. Sci. 2020, 21, 1914. [Google Scholar] [CrossRef]
  186. Martinez, M.N.; Watts, J.L.; Gilbert, J.M. Questions associated with the development of novel drugs intended for the treatment of bacterial infections in veterinary species. Vet. J. 2019, 248, 79–85. [Google Scholar] [CrossRef]
  187. Pomba, C.; Rantala, M.; Greko, C.; Baptiste, K.E.; Catry, B.; Van Duijkeren, E.; Mateus, A.; Moreno, M.A.; Pyörälä, S.; Ružauskas, M. Public health risk of antimicrobial resistance transfer from companion animals. J. Antimicrob. Chemother. 2017, 72, 957–968. [Google Scholar] [CrossRef]
  188. Wolfson, J.S.; Hooper, D.C. The fluoroquinolones: Structures, mechanisms of action and resistance, and spectra of activity in vitro. Antimicrob. Agents Chemother. 1985, 28, 581–586. [Google Scholar] [CrossRef]
  189. Chen, Y.; Xie, Y.; Li, Z. Room-Temperature Phosphorescence of Nicotinic Acid and Isonicotinic Acid: Efficient Intermolecular Hydrogen-Bond Interaction in Molecular Array. J. Phys. Chem. Lett. 2022, 13, 1652–1659. [Google Scholar] [CrossRef]
  190. Rajan, K.; Jaw, R.; Grecz, N. Role of chelation and water binding of calcium in dormancy and heat resistance of bacterial endospores. Bioinorg. Chem. 1978, 8, 477–491. [Google Scholar] [CrossRef]
  191. Ragab, A.E.; Badawy, E.T.; Aboukhatwa, S.M.; Abdel-Aziz, M.M.; Kabbash, A.; Abo Elseoud, K.A. Isonicotinic acid N-oxide, from isoniazid biotransformation by Aspergillus niger, as an InhA inhibitor antituberculous agent against multiple and extensively resistant strains supported by in silico docking and ADME prediction. Nat. Prod. Res. 2023, 37, 1687–1692. [Google Scholar] [CrossRef] [PubMed]
  192. Metcalfe, C.; Macdonald, I.K.; Murphy, E.J.; Brown, K.A.; Raven, E.L.; Moody, P.C. The tuberculosis prodrug isoniazid bound to activating peroxidases. J. Biol. Chem. 2008, 283, 6193–6200. [Google Scholar] [CrossRef]
  193. Volynets, G.P.; Tukalo, M.A.; Bdzhola, V.G.; Derkach, N.M.; Gumeniuk, M.I.; Tarnavskiy, S.S.; Yarmoluk, S.M. Novel isoniazid derivative as promising antituberculosis agent. Future Microbiol. 2020, 15, 869–879. [Google Scholar] [CrossRef]
  194. Judge, V.; Narasimhan, B.; Ahuja, M.; Sriram, D.; Yogeeswari, P.; De Clercq, E.; Pannecouque, C.; Balzarini, J. Synthesis, antimycobacterial, antiviral, antimicrobial activity and QSAR studies of N2-acyl isonicotinic acid hydrazide derivatives. Med. Chem. 2013, 9, 53–76. [Google Scholar] [CrossRef]
  195. Lewis, J.C. Germination of bacterial spores by calcium chelates of dipicolinic acid analogues. J. Biol. Chem. 1972, 247, 1861–1868. [Google Scholar] [CrossRef] [PubMed]
  196. Hintze, P.E.; Nicholson, W.L. Single-spore elemental analyses indicate that dipicolinic acid-deficient Bacillus subtilis spores fail to accumulate calcium. Arch. Microbiol. 2010, 192, 493–497. [Google Scholar] [CrossRef]
  197. Chen, A.Y.; Thomas, P.W.; Stewart, A.C.; Bergstrom, A.; Cheng, Z.; Miller, C.; Bethel, C.R.; Marshall, S.H.; Credille, C.V.; Riley, C.L.; et al. Dipicolinic Acid Derivatives as Inhibitors of New Delhi Metallo-β-lactamase-1. J. Med. Chem. 2017, 60, 7267–7283. [Google Scholar] [CrossRef] [PubMed]
  198. Church, B.D.; Halvorson, H. Dependence of the heat resistance of bacterial endospores on their dipicolinic acid content. Nature 1959, 183, 124–125. [Google Scholar] [CrossRef]
  199. Grecz, N.; Tang, T. Relation of dipicolinic acid to heat resistance of bacterial spores. J. Gen. Microbiol. 1970, 63, 303–310. [Google Scholar] [CrossRef]
  200. Setlow, B.; Atluri, S.; Kitchel, R.; Koziol-Dube, K.; Setlow, P. Role of dipicolinic acid in resistance and stability of spores of Bacillus subtilis with or without DNA-protective alpha/beta-type small acid-soluble proteins. J. Bacteriol. 2006, 188, 3740–3747. [Google Scholar] [CrossRef]
  201. Summers, K.L.; Pushie, M.J.; Sopasis, G.J.; James, A.K.; Dolgova, N.V.; Sokaras, D.; Kroll, T.; Harris, H.H.; Pickering, I.J.; George, G.N. Solution Chemistry of Copper(II) Binding to Substituted 8-Hydroxyquinolines. Inorg. Chem. 2020, 59, 13858–13874. [Google Scholar] [CrossRef]
  202. Pierre, J.L.; Baret, P.; Serratrice, G. Hydroxyquinolines as iron chelators. Curr. Med. Chem. 2003, 10, 1077–1084. [Google Scholar] [CrossRef]
  203. Pape, V.F.S.; May, N.V.; Gál, G.T.; Szatmári, I.; Szeri, F.; Fülöp, F.; Szakács, G.; Enyedy, É.A. Impact of copper and iron binding properties on the anticancer activity of 8-hydroxyquinoline derived Mannich bases. Dalton Trans. 2018, 47, 17032–17045. [Google Scholar] [CrossRef] [PubMed]
  204. Li, L.; Wu, H.; Wang, J.; Ji, Z.; Fang, T.; Lu, H.; Yan, L.; Shen, F.; Zhang, D.; Jiang, Y.; et al. Discovery of Novel 8-Hydroxyquinoline Derivatives with Potent In Vitro and In Vivo Antifungal Activity. J. Med. Chem. 2023, 66, 16364–16376. [Google Scholar] [CrossRef] [PubMed]
  205. Shen, A.Y.; Chen, C.P.; Roffler, S. A chelating agent possessing cytotoxicity and antimicrobial activity: 7-morpholinomethyl-8-hydroxyquinoline. Life Sci. 1999, 64, 813–825. [Google Scholar] [CrossRef]
  206. Cherdtrakulkiat, R.; Boonpangrak, S.; Sinthupoom, N.; Prachayasittikul, S.; Ruchirawat, S.; Prachayasittikul, V. Derivatives (halogen, nitro and amino) of 8-hydroxyquinoline with highly potent antimicrobial and antioxidant activities. Biochem. Biophys. Rep. 2016, 6, 135–141. [Google Scholar] [CrossRef] [PubMed]
  207. Prati, F.; Bergamini, C.; Fato, R.; Soukup, O.; Korabecny, J.; Andrisano, V.; Bartolini, M.; Bolognesi, M.L. Novel 8-Hydroxyquinoline Derivatives as Multitarget Compounds for the Treatment of Alzheimer’s Disease. ChemMedChem 2016, 11, 1284–1295. [Google Scholar] [CrossRef]
  208. Fernández-Bachiller, M.I.; Pérez, C.; González-Muñoz, G.C.; Conde, S.; López, M.G.; Villarroya, M.; García, A.G.; Rodríguez-Franco, M.I. Novel tacrine-8-hydroxyquinoline hybrids as multifunctional agents for the treatment of Alzheimer’s disease, with neuroprotective, cholinergic, antioxidant, and copper-complexing properties. J. Med. Chem. 2010, 53, 4927–4937. [Google Scholar] [CrossRef]
  209. Prachayasittikul, V.; Prachayasittikul, S.; Ruchirawat, S.; Prachayasittikul, V. 8-Hydroxyquinolines: A review of their metal chelating properties and medicinal applications. Drug Des. Dev. Ther. 2013, 7, 1157–1178. [Google Scholar] [CrossRef]
  210. Fazio, F.; Lionetto, L.; Curto, M.; Iacovelli, L.; Cavallari, M.; Zappulla, C.; Ulivieri, M.; Napoletano, F.; Capi, M.; Corigliano, V.; et al. Xanthurenic Acid Activates mGlu2/3 Metabotropic Glutamate Receptors and is a Potential Trait Marker for Schizophrenia. Sci. Rep. 2015, 5, 17799. [Google Scholar] [CrossRef]
  211. Wu, Q.; Li, L.; Zhang, Y.; Ming, X.; Feng, N. Measurement methods, influencing factors and applications of intercellular receptor-ligand binding kinetics in diseases. Prog. Biophys. Mol. Biol. 2024, 194, 43–54. [Google Scholar] [CrossRef] [PubMed]
  212. Taleb, O.; Maammar, M.; Brumaru, D.; Bourguignon, J.J.; Schmitt, M.; Klein, C.; Kemmel, V.; Maitre, M.; Mensah-Nyagan, A.G. Xanthurenic acid binds to neuronal G-protein-coupled receptors that secondarily activate cationic channels in the cell line NCB-20. PLoS ONE 2012, 7, e48553. [Google Scholar] [CrossRef]
  213. Haruki, H.; Hovius, R.; Pedersen, M.G.; Johnsson, K. Tetrahydrobiopterin Biosynthesis as a Potential Target of the Kynurenine Pathway Metabolite Xanthurenic Acid. J. Biol. Chem. 2016, 291, 652–657. [Google Scholar] [CrossRef]
  214. Nilsson, L.; Larsson, A.; Begum, A.; Iakovleva, I.; Carlsson, M.; Brännström, K.; Sauer-Eriksson, A.E.; Olofsson, A. Modifications of the 7-Hydroxyl Group of the Transthyretin Ligand Luteolin Provide Mechanistic Insights into Its Binding Properties and High Plasma Specificity. PLoS ONE 2016, 11, e0153112. [Google Scholar] [CrossRef] [PubMed]
  215. Kontermann, R.E. Half-life extended biotherapeutics. Expert Opin. Biol. Ther. 2016, 16, 903–915. [Google Scholar] [CrossRef] [PubMed]
  216. Contella, L.; Farrell, C.L.; Boccuto, L.; Litwin, A.; Snyder, M.L. Gene Variant Frequencies of IDO1, IDO2, TDO, and KMO in Substance Use Disorder Cohorts. Genes 2024, 15, 1388. [Google Scholar] [CrossRef]
  217. Procaccini, C.; Santopaolo, M.; Faicchia, D.; Colamatteo, A.; Formisano, L.; de Candia, P.; Galgani, M.; De Rosa, V.; Matarese, G. Role of metabolism in neurodegenerative disorders. Metabolism 2016, 65, 1376–1390. [Google Scholar] [CrossRef]
  218. Andrade, J.C.O.; do Vale, T.M.; Gomes, R.L.M.; Forezi, L.; de Souza, M.; Batalha, P.N.; Boechat, F. Exploring 4-quinolone-3-carboxamide derivatives: A versatile framework for emerging biological applications. Bioorg. Chem. 2025, 157, 108240. [Google Scholar] [CrossRef]
  219. Verma, S.; Lal, S.; Narang, R.; Sudhakar, K. Quinoline Hydrazide/Hydrazone Derivatives: Recent Insights on Antibacterial Activity and Mechanism of Action. ChemMedChem 2023, 18, e202200571. [Google Scholar] [CrossRef]
  220. Stone, T.W.; Forrest, C.M.; Darlington, L.G. Kynurenine pathway inhibition as a therapeutic strategy for neuroprotection. FEBS J. 2012, 279, 1386–1397. [Google Scholar] [CrossRef]
  221. Liu, X.; Lv, H.; Guo, Y.; Teka, T.; Wang, X.; Huang, Y.; Han, L.; Pan, G. Structure-Based Reactivity Profiles of Reactive Metabolites with Glutathione. Chem. Res. Toxicol. 2020, 33, 1579–1593. [Google Scholar] [CrossRef] [PubMed]
  222. Musiol, R. An overview of quinoline as a privileged scaffold in cancer drug discovery. Expert Opin. Drug Discov. 2017, 12, 583–597. [Google Scholar] [CrossRef]
  223. Chen, C.; Yang, Y.; Wang, Z.; Li, H.; Dong, C.; Zhang, X. Recent Advances in Pro-PROTAC Development to Address On-Target Off-Tumor Toxicity. J. Med. Chem. 2023, 66, 8428–8440. [Google Scholar] [CrossRef]
  224. Robles, O.; Romo, D. Chemo- and site-selective derivatizations of natural products enabling biological studies. Nat. Prod. Rep. 2014, 31, 318–334. [Google Scholar] [CrossRef] [PubMed]
  225. Ramann, G.A.; Cowen, B.J. Recent Advances in Metal-Free Quinoline Synthesis. Molecules 2016, 21, 986. [Google Scholar] [CrossRef]
  226. Liu, Z.; Fang, J.J.; Wang, Z.Y.; Xie, Y.P.; Lu, X. Structural diversity of copper(I) alkynyl cluster-based coordination polymers utilizing bifunctional pyridine carboxylic acid ligands. Nanoscale 2024, 16, 17817–17824. [Google Scholar] [CrossRef]
  227. Vitaku, E.; Smith, D.T.; Njardarson, J.T. Analysis of the structural diversity, substitution patterns, and frequency of nitrogen heterocycles among U.S. FDA approved pharmaceuticals. J. Med. Chem. 2014, 57, 10257–10274. [Google Scholar] [CrossRef] [PubMed]
  228. Garcia, C. Tuning receptor signaling through ligand engineering. FASEB J. 2020, 34, 1-1. [Google Scholar] [CrossRef]
  229. Abraham, M.H.; Acree, W.E., Jr. On the solubility of nicotinic acid and isonicotinic acid in water and organic solvents. J. Chem. Thermodyn. 2013, 61, 74–78. [Google Scholar] [CrossRef]
  230. Kotammagari, T.K.; Saleh, L.Y.; Lönnberg, T. Organometallic modification confers oligonucleotides new functionalities. Chem. Commun. 2024, 60, 3118–3128. [Google Scholar] [CrossRef]
  231. Murakami, K.; Ueda, T.; Morikawa, R.; Ito, M.; Haneda, M.; Yoshino, M. Antioxidant effect of dipicolinic acid on the metal-catalyzed lipid peroxidation and enzyme inactivation. Biomed. Res. 1998, 19, 205–208. [Google Scholar] [CrossRef]
  232. Dattatray Shinde, S.; Kumar Behera, S.; Kulkarni, N.; Dewangan, B.; Sahu, B. Bifunctional backbone modified squaramide dipeptides as amyloid beta (Aβ) aggregation inhibitors. Bioorg. Med. Chem. 2024, 97, 117538. [Google Scholar] [CrossRef]
  233. Bhat, S.A.; Henry, R.J.; Blanchard, A.C.; Stoica, B.A.; Loane, D.J.; Faden, A.I. Enhanced Akt/GSK-3β/CREB signaling mediates the anti-inflammatory actions of mGluR5 positive allosteric modulators in microglia and following traumatic brain injury in male mice. J. Neurochem. 2021, 156, 225–248. [Google Scholar] [CrossRef]
  234. Jiang, J.; Dingledine, R. Prostaglandin receptor EP2 in the crosshairs of anti-inflammation, anti-cancer, and neuroprotection. Trends Pharmacol. Sci. 2013, 34, 413–423. [Google Scholar] [CrossRef]
  235. Falcucci, R.M.; Wertz, R.; Green, J.L.; Meucci, O.; Salvino, J.; Fontana, A.C.K. Novel Positive Allosteric Modulators of Glutamate Transport Have Neuroprotective Properties in an in Vitro Excitotoxic Model. ACS Chem. Neurosci. 2019, 10, 3437–3453. [Google Scholar] [CrossRef] [PubMed]
  236. Wang, Q.; Li, W.X.; Dai, S.X.; Guo, Y.C.; Han, F.F.; Zheng, J.J.; Li, G.H.; Huang, J.F. Meta-Analysis of Parkinson’s Disease and Alzheimer’s Disease Revealed Commonly Impaired Pathways and Dysregulation of NRF2-Dependent Genes. J. Alzheimer’s Dis. 2017, 56, 1525–1539. [Google Scholar] [CrossRef]
  237. Yang, L.; Nao, J.; Dong, X. The Therapeutic Potential of Hydroxycinnamic Acid Derivatives in Parkinson’s Disease: Focus on In Vivo Research Advancements. J. Agric. Food Chem. 2023, 71, 10932–10951. [Google Scholar] [CrossRef]
  238. Pagotto, G.L.d.O.; Santos, L.M.O.d.; Osman, N.; Lamas, C.B.; Laurindo, L.F.; Pomini, K.T.; Guissoni, L.M.; Lima, E.P.d.; Goulart, R.d.A.; Catharin, V.M.S. Ginkgo biloba: A leaf of hope in the fight against Alzheimer’s dementia: Clinical trial systematic review. Antioxidants 2024, 13, 651. [Google Scholar] [CrossRef] [PubMed]
  239. Salomé, C.; Salomé-Grosjean, E.; Park, K.D.; Morieux, P.; Swendiman, R.; DeMarco, E.; Stables, J.P.; Kohn, H. Synthesis and anticonvulsant activities of (R)-N-(4’-substituted)benzyl 2-acetamido-3-methoxypropionamides. J. Med. Chem. 2010, 53, 1288–1305. [Google Scholar] [CrossRef]
  240. Sharifi, M.; Buzatu, D.; Harris, S.; Wilkes, J. Development of models for predicting Torsade de Pointes cardiac arrhythmias using perceptron neural networks. BMC Bioinform. 2017, 18, 497. [Google Scholar] [CrossRef]
  241. Assis de Oliveira, B.; Gonçalves de Oliveira, F.; de Assis Cruz, O.; Mendonça de Assis, P.; Glanzmann, N.; David da Silva, A.; Rezende Barbosa Raposo, N.; Zabala Capriles Goliatt, P.V. In Silico and In Vitro Evaluation of Quinoline Derivatives as Potential Inhibitors of AChE, BACE1, and GSK3β for Neurodegenerative Diseases Treatment. Chem. Biodivers. 2025, 22, e202401629. [Google Scholar] [CrossRef] [PubMed]
  242. Zindo, F.T.; Joubert, J.; Malan, S.F. Propargylamine as functional moiety in the design of multifunctional drugs for neurodegenerative disorders: MAO inhibition and beyond. Future Med. Chem. 2015, 7, 609–629. [Google Scholar] [CrossRef] [PubMed]
  243. Janse van Rensburg, H.D.; Terre′Blanche, G.; van der Walt, M.M.; Legoabe, L.J. 5-Substituted 2-benzylidene-1-tetralone analogues as A(1) and/or A(2A) antagonists for the potential treatment of neurological conditions. Bioorg. Chem. 2017, 74, 251–259. [Google Scholar] [CrossRef] [PubMed]
  244. Bongarzone, S.; Bolognesi, M.L. The concept of privileged structures in rational drug design: Focus on acridine and quinoline scaffolds in neurodegenerative and protozoan diseases. Expert Opin. Drug Discov. 2011, 6, 251–268. [Google Scholar] [CrossRef]
  245. Jun, J.V.; Petersson, E.J.; Chenoweth, D.M. Rational Design and Facile Synthesis of a Highly Tunable Quinoline-Based Fluorescent Small-Molecule Scaffold for Live Cell Imaging. J. Am. Chem. Soc. 2018, 140, 9486–9493. [Google Scholar] [CrossRef]
  246. Foster, A.C.; Kemp, J.A.; Leeson, P.D.; Grimwood, S.; Donald, A.E.; Marshall, G.R.; Priestley, T.; Smith, J.D.; Carling, R.W. Kynurenic acid analogues with improved affinity and selectivity for the glycine site on the N-methyl-D-aspartate receptor from rat brain. Mol. Pharmacol. 1992, 41, 914–922. [Google Scholar] [CrossRef]
  247. Kemp, J.A.; Priestley, T. Effects of (+)-HA-966 and 7-chlorokynurenic acid on the kinetics of N-methyl-D-aspartate receptor agonist responses in rat cultured cortical neurons. Mol. Pharmacol. 1991, 39, 666–670. [Google Scholar] [CrossRef]
  248. Kloog, Y.; Lamdani-Itkin, H.; Sokolovsky, M. The glycine site of the N-methyl-D-aspartate receptor channel: Differences between the binding of HA-966 and of 7-chlorokynurenic acid. J. Neurochem. 1990, 54, 1576–1583. [Google Scholar] [CrossRef]
  249. Wu, H.Q.; Salituro, F.G.; Schwarcz, R. Enzyme-catalyzed production of the neuroprotective NMDA receptor antagonist 7-chlorokynurenic acid in the rat brain in vivo. Eur. J. Pharmacol. 1997, 319, 13–20. [Google Scholar] [CrossRef]
  250. Yadav, P.; Shah, K. Quinolines, a perpetual, multipurpose scaffold in medicinal chemistry. Bioorg. Chem. 2021, 109, 104639. [Google Scholar] [CrossRef]
  251. Dolšak, A.; Švajger, U.; Lešnik, S.; Konc, J.; Gobec, S.; Sova, M. Selective Toll-like receptor 7 agonists with novel chromeno[3,4-d]imidazol-4(1H)-one and 2-(trifluoromethyl)quinoline/ quinazoline-4-amine scaffolds. Eur. J. Med. Chem. 2019, 179, 109–122. [Google Scholar] [CrossRef]
  252. Konieczny, J.; Ossowska, K.; Schulze, G.; Coper, H.; Wolfarth, S. L-701,324, a selective antagonist at the glycine site of the NMDA receptor, counteracts haloperidol-induced muscle rigidity in rats. Psychopharmacology 1999, 143, 235–243. [Google Scholar] [CrossRef] [PubMed]
  253. Kemp, J.A.; Foster, A.C.; Leeson, P.D.; Priestley, T.; Tridgett, R.; Iversen, L.L.; Woodruff, G.N. 7-Chlorokynurenic acid is a selective antagonist at the glycine modulatory site of the N-methyl-D-aspartate receptor complex. Proc. Natl. Acad. Sci. USA 1988, 85, 6547–6550. [Google Scholar] [CrossRef]
  254. Lehmann, J.C.; Procureur, D.; Wood, P.L. 7-Chlorokynurenate prevents NMDA-induced and kainate-induced striatal lesions. Brain Res. 1993, 620, 1–6. [Google Scholar] [CrossRef] [PubMed]
  255. Zhu, W.L.; Wang, S.J.; Liu, M.M.; Shi, H.S.; Zhang, R.X.; Liu, J.F.; Ding, Z.B.; Lu, L. Glycine site N-methyl-D-aspartate receptor antagonist 7-CTKA produces rapid antidepressant-like effects in male rats. J. Psychiatry Neurosci. 2013, 38, 306–316. [Google Scholar] [CrossRef] [PubMed]
  256. Koek, W.; Colpaert, F.C. Selective blockade of N-methyl-D-aspartate (NMDA)-induced convulsions by NMDA antagonists and putative glycine antagonists: Relationship with phencyclidine-like behavioral effects. J. Pharmacol. Exp. Ther. 1990, 252, 349–357. [Google Scholar] [CrossRef]
  257. Wood, E.R.; Bussey, T.J.; Phillips, A.G. A glycine antagonist 7-chlorokynurenic acid attenuates ischemia-induced learning deficits. Neuroreport 1993, 4, 151–154. [Google Scholar] [CrossRef]
  258. AdisInsight. Drug Profile AV101. Available online: https://adisinsight.springer.com/drugs/800019948 (accessed on 18 April 2025).
  259. Baron, B.M.; Harrison, B.L.; Miller, F.P.; McDonald, I.A.; Salituro, F.G.; Schmidt, C.J.; Sorensen, S.M.; White, H.S.; Palfreyman, M.G. Activity of 5,7-dichlorokynurenic acid, a potent antagonist at the N-methyl-D-aspartate receptor-associated glycine binding site. Mol. Pharmacol. 1990, 38, 554–561. [Google Scholar] [CrossRef]
  260. Baron, B.M.; Siegel, B.W.; Slone, A.L.; Harrison, B.L.; Palfreyman, M.G.; Hurt, S.D. [3H]5,7-dichlorokynurenic acid, a novel radioligand labels NMDA receptor-associated glycine binding sites. Eur. J. Pharmacol. 1991, 206, 149–154. [Google Scholar] [CrossRef]
  261. McNamara, D.; Smith, E.C.; Calligaro, D.O.; O’Malley, P.J.; McQuaid, L.A.; Dingledine, R. 5,7-Dichlorokynurenic acid, a potent and selective competitive antagonist of the glycine site on NMDA receptors. Neurosci. Lett. 1990, 120, 17–20. [Google Scholar] [CrossRef]
  262. Hurt, S.D.; Baron, B.M. [3H] 5,7-dichlorokynurenic acid, a high affinity ligand for the NMDA receptor glycine regulatory site. J. Recept. Res. 1991, 11, 215–220. [Google Scholar] [CrossRef] [PubMed]
  263. Corbett, R.; Dunn, R.W. Effects of 5,7 dichlorokynurenic acid on conflict, social interaction and plus maze behaviors. Neuropharmacology 1993, 32, 461–466. [Google Scholar] [CrossRef] [PubMed]
  264. Plaznik, A.; Palejko, W.; Nazar, M.; Jessa, M. Effects of antagonists at the NMDA receptor complex in two models of anxiety. Eur. Neuropsychopharmacol. 1994, 4, 503–512. [Google Scholar] [CrossRef] [PubMed]
  265. Hlinák, Z.; Krejci, I. Kynurenic acid and 5,7-dichlorokynurenic acids improve social and object recognition in male rats. Psychopharmacology 1995, 120, 463–469. [Google Scholar] [CrossRef]
  266. Priestley, T.; Laughton, P.; Macaulay, A.J.; Hill, R.G.; Kemp, J.A. Electrophysiological characterisation of the antagonist properties of two novel NMDA receptor glycine site antagonists, L-695,902 and L-701,324. Neuropharmacology 1996, 35, 1573–1581. [Google Scholar] [CrossRef]
  267. Dannhardt, G.; Kohl, B.K. The glycine site on the NMDA receptor: Structure-activity relationships and possible therapeutic applications. Curr. Med. Chem. 1998, 5, 253–263. [Google Scholar] [CrossRef]
  268. Kotlinska, J.; Liljequist, S. A characterization of anxiolytic-like actions induced by the novel NMDA/glycine site antagonist, L-701,324. Psychopharmacology 1998, 135, 175–181. [Google Scholar] [CrossRef]
  269. Potschka, H.; Löscher, W.; Wlaź, P.; Behl, B.; Hofmann, H.P.; Treiber, H.J.; Szabo, L. LU 73068, a new non-NMDA and glycine/NMDA receptor antagonist: Pharmacological characterization and comparison with NBQX and L-701,324 in the kindling model of epilepsy. Br. J. Pharmacol. 1998, 125, 1258–1266. [Google Scholar] [CrossRef]
  270. Bristow, L.J.; Flatman, K.L.; Hutson, P.H.; Kulagowski, J.J.; Leeson, P.D.; Young, L.; Tricklebank, M.D. The atypical neuroleptic profile of the glycine/N-methyl-D-aspartate receptor antagonist, L-701,324, in rodents. J. Pharmacol. Exp. Ther. 1996, 277, 578–585. [Google Scholar] [CrossRef]
  271. Christoph, T.; Reissmüller, E.; Schiene, K.; Englberger, W.; Chizh, B.A. Antiallodynic effects of NMDA glycine(B) antagonists in neuropathic pain: Possible peripheral mechanisms. Brain Res. 2005, 1048, 218–227. [Google Scholar] [CrossRef]
  272. Lees, K.R.; Asplund, K.; Carolei, A.; Davis, S.M.; Diener, H.-C.; Kaste, M.; Orgogozo, J.-M.; Whitehead, J. Glycine antagonist (gavestinel) in neuroprotection (GAIN International) in patients with acute stroke: A randomised controlled trial. Lancet 2000, 355, 1949–1954. [Google Scholar] [CrossRef]
  273. Banks, P.; Franks, N.P.; Dickinson, R. Competitive inhibition at the glycine site of the N-methyl-D-aspartate receptor mediates xenon neuroprotection against hypoxia-ischemia. Anesthesiology 2010, 112, 614–622. [Google Scholar] [CrossRef] [PubMed]
  274. Benneyworth, M.A.; Basu, A.C.; Coyle, J.T. Discordant behavioral effects of psychotomimetic drugs in mice with altered NMDA receptor function. Psychopharmacology 2011, 213, 143–153. [Google Scholar] [CrossRef] [PubMed]
  275. Haley, E.C., Jr.; Thompson, J.L.; Levin, B.; Davis, S.; Lees, K.R.; Pittman, J.G.; DeRosa, J.T.; Ordronneau, P.; Brown, D.L.; Sacco, R.L. Gavestinel does not improve outcome after acute intracerebral hemorrhage: An analysis from the GAIN International and GAIN Americas studies. Stroke 2005, 36, 1006–1010. [Google Scholar] [CrossRef]
  276. Sacco, R.L.; DeRosa, J.T.; Haley, E.C., Jr.; Levin, B.; Ordronneau, P.; Phillips, S.J.; Rundek, T.; Snipes, R.G.; Thompson, J.L.; Investigators, G.A. Glycine antagonist in neuroprotection for patients with acute stroke: GAIN Americas: A randomized controlled trial. JAMA 2001, 285, 1719–1728. [Google Scholar] [CrossRef] [PubMed]
  277. Jiang, T.; Zhang, W.; Wang, Z. Laquinimod protects against TNF-α-induced attachment of monocytes to human aortic endothelial cells (HAECs) by increasing the expression of KLF2. Drug Des. Dev. Ther. 2020, 14, 1683–1691. [Google Scholar] [CrossRef]
  278. Nessler, S.; Ott, M.; Wegner, C.; Hayardeny, L.; Ullrich, E.; Bruck, W. Laquinimod suppresses CNS autoimmunity by activation of natural killer cells (P1. 161). Neurology 2015, 84, P1–P161. [Google Scholar] [CrossRef]
  279. Ott, M.; Avendaño-Guzmán, E.; Ullrich, E.; Dreyer, C.; Strauss, J.; Harden, M.; Schön, M.; Schön, M.P.; Bernhardt, G.; Stadelmann, C. Laquinimod, a prototypic quinoline-3-carboxamide and aryl hydrocarbon receptor agonist, utilizes a CD155-mediated natural killer/dendritic cell interaction to suppress CNS autoimmunity. J. Neuroinflammation 2019, 16, 49. [Google Scholar] [CrossRef] [PubMed]
  280. Ellrichmann, G.; Blusch, A.; Fatoba, O.; Brunner, J.; Reick, C.; Hayardeny, L.; Hayden, M.; Sehr, D.; Winklhofer, K.F.; Saft, C. Laquinimod treatment in the R6/2 mouse model. Sci. Rep. 2017, 7, 4947. [Google Scholar] [CrossRef]
  281. Rothhammer, V.; Kenison, J.E.; Li, Z.; Tjon, E.; Takenaka, M.C.; Chao, C.-C.; Alves de Lima, K.; Borucki, D.M.; Kaye, J.; Quintana, F.J. Aryl hydrocarbon receptor activation in astrocytes by laquinimod ameliorates autoimmune inflammation in the CNS. Neurol. Neuroimmunol. Neuroinflammation 2021, 8, e946. [Google Scholar] [CrossRef]
  282. Comi, G.; Jeffery, D.; Kappos, L.; Montalban, X.; Boyko, A.; Rocca, M.A.; Filippi, M. Placebo-controlled trial of oral laquinimod for multiple sclerosis. N. Engl. J. Med. 2012, 366, 1000–1009. [Google Scholar] [CrossRef] [PubMed]
  283. Vollmer, T.; Sorensen, P.; Selmaj, K.; Zipp, F.; Havrdova, E.; Cohen, J.; Sasson, N.; Gilgun-Sherki, Y.; Arnold, D.; Group, B.S. A randomized placebo-controlled phase III trial of oral laquinimod for multiple sclerosis. J. Neurol. 2014, 261, 773–783. [Google Scholar] [CrossRef] [PubMed]
  284. Comi, G.; Dadon, Y.; Sasson, N.; Steinerman, J.R.; Knappertz, V.; Vollmer, T.L.; Boyko, A.; Vermersch, P.; Ziemssen, T.; Montalban, X. CONCERTO: A randomized, placebo-controlled trial of oral laquinimod in relapsing-remitting multiple sclerosis. Mult. Scler. J. 2022, 28, 608–619. [Google Scholar] [CrossRef]
  285. Isaacs, J.T. The long and winding road for the development of tasquinimod as an oral second-generation quinoline-3-carboxamide antiangiogenic drug for the treatment of prostate cancer. Expert Opin. Investig. Drugs 2010, 19, 1235–1243. [Google Scholar] [CrossRef] [PubMed]
  286. Shen, L.; Sundstedt, A.; Ciesielski, M.; Miles, K.M.; Celander, M.; Adelaiye, R.; Orillion, A.; Ciamporcero, E.; Ramakrishnan, S.; Ellis, L. Tasquinimod modulates suppressive myeloid cells and enhances cancer immunotherapies in murine models. Cancer Immunol. Res. 2015, 3, 136–148. [Google Scholar] [CrossRef]
  287. Wobus, M.; Balaian, E.; Oelschlaegel, U.; Towers, R.; Möbus, K.; Habermann, I.; Wehner, R.; Stölzel, F.; Chavakis, T.; Törngren, M. Targeting the Inflammatory Niche in MDS By Tasquinimod Restores Hematopoietic Support and Suppresses Immune-Checkpoint Expression in Vitro. Blood 2021, 138, 2596. [Google Scholar] [CrossRef]
  288. Li, Z.; Zhang, X.; Liu, C.; Peng, Q.; Wu, Y.; Wen, Y.; Zheng, R.; Yan, Q.; Ma, J. Macrophage-Biomimetic Nanoparticles Ameliorate Ulcerative Colitis through Reducing Inflammatory Factors Expression. J. Innate Immun. 2022, 14, 380–392. [Google Scholar] [CrossRef]
  289. Cristóvão, J.S.; Gomes, C.M. S100 Proteins in Alzheimer’s Disease. Front. Neurosci. 2019, 13, 463. [Google Scholar] [CrossRef]
  290. Leimkühler, N.B.; Gleitz, H.F.E.; Ronghui, L.; Snoeren, I.A.M.; Fuchs, S.N.R.; Nagai, J.S.; Banjanin, B.; Lam, K.H.; Vogl, T.; Kuppe, C.; et al. Heterogeneous bone-marrow stromal progenitors drive myelofibrosis via a druggable alarmin axis. Cell Stem Cell 2021, 28, 637–652.e638. [Google Scholar] [CrossRef]
  291. Xu, K.; Dai, X.L.; Huang, H.C.; Jiang, Z.F. Targeting HDACs: A promising therapy for Alzheimer’s disease. Oxid. Med. Cell. Longev. 2011, 2011, 143269. [Google Scholar] [CrossRef]
  292. Ganai, S.A.; Ramadoss, M.; Mahadevan, V. Histone Deacetylase (HDAC) Inhibitors—Emerging roles in neuronal memory, learning, synaptic plasticity and neural regeneration. Curr. Neuropharmacol. 2016, 14, 55–71. [Google Scholar] [CrossRef] [PubMed]
  293. Singh, Y.; Bhatia, N.; Biharee, A.; Kulkarni, S.; Thareja, S.; Monga, V. Developing our knowledge of the quinolone scaffold and its value to anticancer drug design. Expert Opin. Drug Discov. 2023, 18, 1151–1167. [Google Scholar] [CrossRef]
  294. Kleckner, N.; Dingledine, R. Selectivity of quinoxalines and kynurenines as antagonists of the glycine site on N-methyl-D-aspartate receptors. Mol. Pharmacol. 1989, 36, 430–436. [Google Scholar] [CrossRef] [PubMed]
  295. Culletta, G.; Tutone, M.; Zappalà, M.; Almerico, A.M. Sulfonamide moiety as “molecular chimera” in the design of new drugs. Curr. Med. Chem. 2023, 30, 128–163. [Google Scholar] [CrossRef]
  296. Brnardic, E.J.; Ye, G.; Brooks, C.; Donatelli, C.; Barton, L.; McAtee, J.; Sanchez, R.M.; Shu, A.; Erhard, K.; Terrell, L. Discovery of pyrrolidine sulfonamides as selective and orally bioavailable antagonists of transient receptor potential vanilloid-4 (TRPV4). J. Med. Chem. 2018, 61, 9738–9755. [Google Scholar] [CrossRef] [PubMed]
  297. Yu, Y.; Wang, C.; Zhang, X.; Zhu, J.; Wang, L.; Ji, M.; Zhang, Z.; Ji, X.-M.; Wang, S.-L. Perfluorooctane sulfonate disrupts the blood brain barrier through the crosstalk between endothelial cells and astrocytes in mice. Environ. Pollut. 2020, 256, 113429. [Google Scholar] [CrossRef]
  298. Ma, H.; Brust, T.; Frankowski, K.J.; Lovell, K.M.; Cameron, M.D.; Bohn, L.M.; Aube, J. Advances in Sulfonamide Kappa Opioid Receptor Antagonists: Structural Refinement and Evaluation of CNS Clearance. ACS Chem. Neurosci. 2022, 13, 1315–1332. [Google Scholar] [CrossRef]
  299. Zong, P.; Feng, J.; Yue, Z.; Li, Y.; Wu, G.; Sun, B.; He, Y.; Miller, B.; Albert, S.Y.; Su, Z. Functional coupling of TRPM2 and extrasynaptic NMDARs exacerbates excitotoxicity in ischemic brain injury. Neuron 2022, 110, 1944–1958.e1948. [Google Scholar] [CrossRef]
  300. Shen, Z.; Xiang, M.; Chen, C.; Ding, F.; Wang, Y.; Shang, C.; Xin, L.; Zhang, Y.; Cui, X. Glutamate excitotoxicity: Potential therapeutic target for ischemic stroke. Biomed. Pharmacother. 2022, 151, 113125. [Google Scholar] [CrossRef]
  301. Butcher, S.P.; Henshall, D.C.; Teramura, Y.; Iwasaki, K.; Sharkey, J. Neuroprotective actions of FK506 in experimental stroke: In vivo evidence against an antiexcitotoxic mechanism. J. Neurosci. 1997, 17, 6939–6946. [Google Scholar] [CrossRef]
  302. Singh, D.; Wasan, H.; Reeta, K. Preclinical stroke research and translational failure: A bird’s eye view on preventable variables. Cell. Mol. Neurobiol. 2022, 42, 2003–2017. [Google Scholar] [CrossRef] [PubMed]
  303. Cui, L.l.; Golubczyk, D.; Tolppanen, A.M.; Boltze, J.; Jolkkonen, J. Cell therapy for ischemic stroke: Are differences in preclinical and clinical study design responsible for the translational loss of efficacy? Ann. Neurol. 2019, 86, 5–16. [Google Scholar] [CrossRef] [PubMed]
  304. Dirnagl, U.; Fisher, M. International, multicenter randomized preclinical trials in translational stroke research: It’s time to act. J. Cereb. Blood Flow Metab. 2012, 32, 933–935. [Google Scholar] [CrossRef]
  305. Le Naour, J.; Galluzzi, L.; Zitvogel, L.; Kroemer, G.; Vacchelli, E. Trial watch: IDO inhibitors in cancer therapy. Oncoimmunology 2020, 9, 1777625. [Google Scholar] [CrossRef] [PubMed]
  306. Van den Eynde, B.J.; van Baren, N.; Baurain, J.-F. Is there a clinical future for IDO1 inhibitors after the failure of epacadostat in melanoma? Annu. Rev. Cancer Biol. 2020, 4, 241–256. [Google Scholar] [CrossRef]
  307. Muller, A.J.; Manfredi, M.G.; Zakharia, Y.; Prendergast, G.C. Inhibiting IDO pathways to treat cancer: Lessons from the ECHO-301 trial and beyond. Semin. Immunopathol. 2019, 41, 41–48. [Google Scholar] [CrossRef]
  308. Fujiwara, Y.; Kato, S.; Nesline, M.K.; Conroy, J.M.; DePietro, P.; Pabla, S.; Kurzrock, R. Indoleamine 2,3-dioxygenase (IDO) inhibitors and cancer immunotherapy. Cancer Treat. Rev. 2022, 110, 102461. [Google Scholar] [CrossRef]
  309. Yan, Y.; Kumar, A.B.; Finnes, H.; Markovic, S.N.; Park, S.; Dronca, R.S.; Dong, H. Combining immune checkpoint inhibitors with conventional cancer therapy. Front. Immunol. 2018, 9, 1739. [Google Scholar] [CrossRef]
  310. Desnoyer, A.; Broutin, S.; Delahousse, J.; Maritaz, C.; Blondel, L.; Mir, O.; Chaput, N.; Paci, A. Pharmacokinetic/pharmacodynamic relationship of therapeutic monoclonal antibodies used in oncology: Part 2, immune checkpoint inhibitor antibodies. Eur. J. Cancer 2020, 128, 119–128. [Google Scholar] [CrossRef]
  311. Metropulos, A.E.; Munshi, H.G.; Principe, D.R. The difficulty in translating the preclinical success of combined TGFβ and immune checkpoint inhibition to clinical trial. EBioMedicine 2022, 86, 104380. [Google Scholar] [CrossRef]
  312. Tanaka, M.; Vécsei, L. From Lab to Life: Exploring Cutting-Edge Models for Neurological and Psychiatric Disorders. Biomedicines 2024, 12, 613. [Google Scholar] [CrossRef] [PubMed]
  313. Giacomini, P.S.; Bar-Or, A. Laquinimod in multiple sclerosis. Clin. Immunol. 2012, 142, 38–43. [Google Scholar] [CrossRef]
  314. Brück, W.; Zamvil, S.S. Laquinimod, a once-daily oral drug in development for the treatment of relapsing–remitting multiple sclerosis. Expert Rev. Clin. Pharmacol. 2012, 5, 245–256. [Google Scholar] [CrossRef]
  315. Haggiag, S.; Ruggieri, S.; Gasperini, C. Efficacy and safety of laquinimod in multiple sclerosis: Current status. Ther. Adv. Neurol. Disord. 2013, 6, 343–352. [Google Scholar] [CrossRef]
  316. Ahmad, S.R.; Zeyaullah, M.; Khan, M.S.; AlShahrani, A.M.; Altijani, A.A.G.; Ali, H.; Dawria, A.; Mohieldin, A.; Alam, M.S.; Mohamed, A.O.A. Pharmacogenomics for neurodegenerative disorders—A focused review. Front. Pharmacol. 2024, 15, 1478964. [Google Scholar] [CrossRef]
  317. Toader, C.; Tataru, C.P.; Munteanu, O.; Serban, M.; Covache-Busuioc, R.-A.; Ciurea, A.V.; Enyedi, M. Decoding Neurodegeneration: A Review of Molecular Mechanisms and Therapeutic Advances in Alzheimer’s, Parkinson’s, and ALS. Int. J. Mol. Sci. 2024, 25, 12613. [Google Scholar] [CrossRef]
  318. Hutson, P.H.; Barton, C.L. L-701,324, a glycine/NMDA receptor antagonist, blocks the increase of cortical dopamine metabolism by stress and DMCM. Eur. J. Pharmacol. 1997, 326, 127–132. [Google Scholar] [CrossRef] [PubMed]
  319. Zellinger, C.; Salvamoser, J.D.; Soerensen, J.; van Vliet, E.A.; Aronica, E.; Gorter, J.; Potschka, H. Pre-treatment with the NMDA receptor glycine-binding site antagonist L-701,324 improves pharmacosensitivity in a mouse kindling model. Epilepsy Res. 2014, 108, 634–643. [Google Scholar] [CrossRef] [PubMed]
  320. Harding, E.K.; Dedek, A.; Bonin, R.P.; Salter, M.W.; Snutch, T.P.; Hildebrand, M.E. The T-type calcium channel antagonist, Z944, reduces spinal excitability and pain hypersensitivity. Br. J. Pharmacol. 2021, 178, 3517–3532. [Google Scholar] [CrossRef]
  321. Obrenovitch, T.P.; Hardy, A.M.; Zilkha, E. Effects of L-701,324, a high-affinity antagonist at the N-methyl-D-aspartate (NMDA) receptor glycine site, on the rat electroencephalogram. Naunyn-Schmiedeberg’s Arch. Pharmacol. 1997, 355, 779–786. [Google Scholar] [CrossRef]
  322. Egunlusi, A.O.; Joubert, J. NMDA receptor antagonists: Emerging insights into molecular mechanisms and clinical applications in neurological disorders. Pharmaceuticals 2024, 17, 639. [Google Scholar] [CrossRef] [PubMed]
  323. Lotti, J.S.; Jones, J.; Farnsworth, J.C.; Yi, F.; Zhao, F.; Menniti, F.S.; Volkmann, R.A.; Clausen, R.P.; Hansen, K.B. Evaluation of allosteric N-methyl-d-aspartate receptor modulation by GluN2A-selective antagonists using pharmacological equilibrium modeling. Mol. Pharmacol. 2025, 107, 100004. [Google Scholar] [CrossRef]
  324. Ahmed, H.; Haider, A.; Ametamey, S.M. N-Methyl-D-Aspartate (NMDA) receptor modulators: A patent review (2015-present). Expert Opin. Ther. Pat. 2020, 30, 743–767. [Google Scholar] [CrossRef]
  325. Verma, S.; Ambatwar, R.; Datusalia, A.K.; Khatik, G.L. Convenient One-Pot Synthesis of Kynurenic Acid Ethyl Ester and Exploration to Direct Synthesis of Neuroprotective Kynurenic Acid and Amide Derivatives. J. Org. Chem. 2023, 88, 10494–10500. [Google Scholar] [CrossRef]
  326. Xiong, B.; Wang, Y.; Chen, Y.; Xing, S.; Liao, Q.; Chen, Y.; Li, Q.; Li, W.; Sun, H. Strategies for structural modification of small molecules to improve blood–brain barrier penetration: A recent perspective. J. Med. Chem. 2021, 64, 13152–13173. [Google Scholar] [CrossRef] [PubMed]
  327. Isaacs, J.T.; Antony, L.; Dalrymple, S.L.; Brennen, W.N.; Gerber, S.; Hammers, H.; Wissing, M.; Kachhap, S.; Luo, J.; Xing, L.; et al. Tasquinimod is an allosteric modulator of HDAC4 survival signaling within the compromised cancer microenvironment. Cancer Res. 2013, 73, 1386–1399. [Google Scholar] [CrossRef] [PubMed]
  328. Raymond, E.; Dalgleish, A.; Damber, J.-E.; Smith, M.; Pili, R. Mechanisms of action of tasquinimod on the tumour microenvironment. Cancer Chemother. Pharmacol. 2014, 73, 1–8. [Google Scholar] [CrossRef] [PubMed]
  329. Joseph, J.; Gonzalez-Lopez, M.; Galang, C.; Garcia, C.; Lemar, H.; Lu, J.; Vaidya, K.; Fischer, M.; Frey, C.; Alam, M. Small-molecule antagonists of the aryl hydrocarbon receptor (AhR) promote activation of human PBMCs in vitro and demonstrate significant impact on tumor growth and immune modulation in vivo. Cancer Res. 2018, 78, 4719. [Google Scholar] [CrossRef]
  330. Acharya, A.; Yadav, M.; Nagpure, M.; Kumaresan, S.; Guchhait, S.K. Molecular medicinal insights into scaffold hopping-based drug discovery success. Drug Discov. Today 2024, 29, 103845. [Google Scholar] [CrossRef]
  331. Zheng, H.; Dai, Q.; Yuan, Z.; Fan, T.; Zhang, C.; Liu, Z.; Chu, B.; Sun, Q.; Chen, Y.; Jiang, Y. Quinazoline-based hydroxamic acid derivatives as dual histone methylation and deacetylation inhibitors for potential anticancer agents. Bioorganic Med. Chem. 2022, 53, 116524. [Google Scholar] [CrossRef]
  332. Ma, C.; Taghour, M.S.; Belal, A.; Mehany, A.B.M.; Mostafa, N.; Nabeeh, A.; Eissa, I.H.; Al-Karmalawy, A.A. Design and Synthesis of New Quinoxaline Derivatives as Potential Histone Deacetylase Inhibitors Targeting Hepatocellular Carcinoma: In Silico, In Vitro, and SAR Studies. Front. Chem. 2021, 9, 725135. [Google Scholar] [CrossRef] [PubMed]
  333. Sforzini, L.; Nettis, M.A.; Mondelli, V.; Pariante, C.M. Inflammation in cancer and depression: A starring role for the kynurenine pathway. Psychopharmacology 2019, 236, 2997–3011. [Google Scholar] [CrossRef] [PubMed]
  334. Maddison, D.C.; Giorgini, F. The kynurenine pathway and neurodegenerative disease. Semin. Cell Dev. Biol. 2015, 40, 134–141. [Google Scholar] [CrossRef]
  335. Li, T.; Kung, H.J.; Mack, P.C.; Gandara, D.R. Genotyping and genomic profiling of non-small-cell lung cancer: Implications for current and future therapies. J. Clin. Oncol. 2013, 31, 1039–1049. [Google Scholar] [CrossRef]
  336. Correia, A.C.; Monteiro, A.R.; Silva, R.; Moreira, J.N.; Sousa Lobo, J.M.; Silva, A.C. Lipid nanoparticles strategies to modify pharmacokinetics of central nervous system targeting drugs: Crossing or circumventing the blood-brain barrier (BBB) to manage neurological disorders. Adv. Drug Deliv. Rev. 2022, 189, 114485. [Google Scholar] [CrossRef] [PubMed]
  337. Khare, P.; Edgecomb, S.X.; Hamadani, C.M.; Tanner, E.E.L.; Manickam, D.S. Lipid nanoparticle-mediated drug delivery to the brain. Adv. Drug Deliv. Rev. 2023, 197, 114861. [Google Scholar] [CrossRef]
  338. Teixeira, M.I.; Lopes, C.M.; Amaral, M.H.; Costa, P.C. Surface-modified lipid nanocarriers for crossing the blood-brain barrier (BBB): A current overview of active targeting in brain diseases. Colloids Surf. B Biointerfaces 2023, 221, 112999. [Google Scholar] [CrossRef]
  339. Rahimi, S.; Khoee, S.; Ghandi, M. Preparation and characterization of rod-like chitosan-quinoline nanoparticles as pH-responsive nanocarriers for quercetin delivery. Int. J. Biol. Macromol. 2019, 128, 279–289. [Google Scholar] [CrossRef]
  340. Ren, B.; Guo, C.; Liu, R.-Z.; Bian, Z.-Y.; Liu, R.-C.; Huang, L.-F.; Tang, J.-J. Imidazolylacetophenone oxime-based multifunctional neuroprotective agents: Discovery and structure-activity relationships. Eur. J. Med. Chem. 2022, 228, 114031. [Google Scholar] [CrossRef]
  341. Knyihar-Csillik, E.; Mihaly, A.; Krisztin-Peva, B.; Robotka, H.; Szatmari, I.; Fulop, F.; Toldi, J.; Csillik, B.; Vecsei, L. The kynurenate analog SZR-72 prevents the nitroglycerol-induced increase of c-fos immunoreactivity in the rat caudal trigeminal nucleus: Comparative studies of the effects of SZR-72 and kynurenic acid. Neurosci. Res. 2008, 61, 429–432. [Google Scholar] [CrossRef]
  342. Kassai, F.; Kedves, R.; Gyertyán, I.; Tuka, B.; Fülöp, F.; Toldi, J.; Lendvai, B.; Vécsei, L. Effect of a kynurenic acid analog on home-cage activity and body temperature in rats. Pharmacol. Rep. 2015, 67, 1188–1192. [Google Scholar] [CrossRef] [PubMed]
  343. Martos, D.; Lőrinczi, B.; Szatmári, I.; Vécsei, L.; Tanaka, M. The impact of C-3 side chain modifications on Kynurenic Acid: A behavioral analysis of its analogs in the Motor Domain. Int. J. Mol. Sci. 2024, 25, 3394. [Google Scholar] [CrossRef] [PubMed]
  344. Poles, M.Z.; Nászai, A.; Gulácsi, L.; Czakó, B.L.; Gál, K.G.; Glenz, R.J.; Dookhun, D.; Rutai, A.; Tallósy, S.P.; Szabó, A. Kynurenic acid and its synthetic derivatives protect against sepsis-associated neutrophil activation and brain mitochondrial dysfunction in rats. Front. Immunol. 2021, 12, 717157. [Google Scholar] [CrossRef]
  345. Mándi, Y.; Endrész, V.; Mosolygó, T.; Burián, K.; Lantos, I.; Fülöp, F.; Szatmári, I.; Lőrinczi, B.; Balog, A.; Vécsei, L. The opposite effects of kynurenic acid and different kynurenic acid analogs on tumor necrosis factor-α (TNF-α) production and tumor necrosis factor-stimulated gene-6 (TSG-6) expression. Front. Immunol. 2019, 10, 1406. [Google Scholar] [CrossRef]
  346. Knapp, L.; Szita, B.; Kocsis, K.; Vécsei, L.; Toldi, J. Nitroglycerin enhances the propagation of cortical spreading depression: Comparative studies with sumatriptan and novel kynurenic acid analogues. Drug Des. Dev. Ther. 2016, 11, 27–34. [Google Scholar] [CrossRef] [PubMed]
  347. Tanaka, M.; Szabó, Á.; Lőrinczi, B.; Szatmári, I.; Fülöp, F.; Vécsei, L. Antidepressant-like Effects of Kynurenic Acid Analogues. In Proceedings of the 1st International Electronic Conference on Biomedicine, Online, 1–26 March 2021; p. 26. [Google Scholar]
  348. Evelin, H.L.F. Kinurénsav-Analógok In Vitro Elektrofiziológiai Vizsgálata az Ischaemiás Stroke Szempontjából; Szegedi Tudomanyegyetem: Szeged, Hungary, 2022. [Google Scholar]
  349. Leeson, P.D.; Baker, R.; Carling, R.W.; Curtis, N.R.; Moore, K.W.; Williams, B.J.; Foster, A.C.; Donald, A.E.; Kemp, J.A.; Marshall, G.R. Kynurenic acid derivatives. Structure-activity relationships for excitatory amino acid antagonism and identification of potent and selective antagonists at the glycine site on the N-methyl-D-aspartate receptor. J. Med. Chem. 1991, 34, 1243–1252. [Google Scholar] [CrossRef]
  350. Vasilopoulos, A.; Krska, S.W.; Stahl, S.S. C(sp3)–H methylation enabled by peroxide photosensitization and Ni-mediated radical coupling. Science 2021, 372, 398–403. [Google Scholar] [CrossRef]
  351. Comeau, C.; Ries, B.; Stadelmann, T.; Tremblay, J.; Poulet, S.; Frohlich, U.; Côté, J.; Boudreault, P.-L.; Derbali, R.M.; Sarret, P. Modulation of the passive permeability of semipeptidic macrocycles: N-and C-methylations fine-tune conformation and properties. J. Med. Chem. 2021, 64, 5365–5383. [Google Scholar] [CrossRef]
  352. Jóźwiak, K.; Płazińska, A. Structural insights into ligand—Receptor interactions involved in biased agonism of G-protein coupled receptors. Molecules 2021, 26, 851. [Google Scholar] [CrossRef]
  353. Liu, L.; Ge, J.; Fu, H.; Wang, J.; Chen, Y.; Sun, Y.; Zhao, C.; Li, Y.; Li, Y. A fluorinated prodrug strategy enhances the therapeutic index of nanoparticle-delivered hydrophilic drugs. Colloids Surf. B Biointerfaces 2025, 249, 114539. [Google Scholar] [CrossRef]
  354. Chen, K.J.; Plaunt, A.J.; Leifer, F.G.; Kang, J.Y.; Cipolla, D. Recent advances in prodrug-based nanoparticle therapeutics. Eur. J. Pharm. Biopharm. 2021, 165, 219–243. [Google Scholar] [CrossRef] [PubMed]
  355. Tanaka, M.; Tuka, B.; Vécsei, L. Navigating the Neurobiology of Migraine: From pathways to potential therapies. Cells 2024, 13, 1098. [Google Scholar] [CrossRef] [PubMed]
  356. Juhász, L.; Spisák, K.; Szolnoki, B.G.; Nászai, A.; Szabó, Á.; Rutai, A.; Tallósy, S.P.; Szabó, A.; Toldi, J.; Tanaka, M.; et al. The Power Struggle: Kynurenine Pathway Enzyme Knockouts and Brain Mitochondrial Respiration. J. Neurochem. 2025; in press. [Google Scholar]
  357. Deora, G.S.; Kantham, S.; Chan, S.; Dighe, S.N.; Veliyath, S.K.; McColl, G.; Parat, M.O.; McGeary, R.P.; Ross, B.P. Multifunctional Analogs of Kynurenic Acid for the Treatment of Alzheimer’s Disease: Synthesis, Pharmacology, and Molecular Modeling Studies. ACS Chem. Neurosci. 2017, 8, 2667–2675. [Google Scholar] [CrossRef]
  358. Yoshida, Y.; Fujigaki, H.; Kato, K.; Yamazaki, K.; Fujigaki, S.; Kunisawa, K.; Yamamoto, Y.; Mouri, A.; Oda, A.; Nabeshima, T.; et al. Selective and competitive inhibition of kynurenine aminotransferase 2 by glycyrrhizic acid and its analogues. Sci. Rep. 2019, 9, 10243. [Google Scholar] [CrossRef] [PubMed]
  359. Agrafiotis, D.K.; Shemanarev, M.; Connolly, P.J.; Farnum, M.; Lobanov, V.S. SAR maps: A new SAR visualization technique for medicinal chemists. J. Med. Chem. 2007, 50, 5926–5937. [Google Scholar] [CrossRef]
  360. Lipinski, C.A. Overview of hit to lead: The medicinal chemist’s role from HTS retest to lead optimization hand off. In Lead-Seeking Approaches; Springer: Berlin/Heidelberg, Germany, 2010; pp. 1–24. [Google Scholar]
  361. Segall, M.D.; Beresford, A.P.; Gola, J.M.; Hawksley, D.; Tarbit, M.H. Focus on success: Using a probabilistic approach to achieve an optimal balance of compound properties in drug discovery. Expert Opin. Drug Metab. Toxicol. 2006, 2, 325–337. [Google Scholar] [CrossRef]
  362. Ward, K.W. Optimizing pharmacokinetic properties and attaining candidate selection. In Reducing Drug Attrition; Springer: Berlin/Heidelberg, Germany, 2014; pp. 73–95. [Google Scholar]
  363. Qin, D.; Lin, X.; Liu, Z.; Chen, Y.; Zhang, Z.; Wu, C.; Liu, L.; Pan, Y.; Laquerre, S.; Emery, J. Discovery of orally bioavailable ligand efficient quinazolindiones as potent and selective tankyrases inhibitors. ACS Med. Chem. Lett. 2021, 12, 1005–1010. [Google Scholar] [CrossRef]
  364. Morphy, R.; Kay, C.; Rankovic, Z. From magic bullets to designed multiple ligands. Drug Discov. Today 2004, 9, 641–651. [Google Scholar] [CrossRef]
  365. Mager, D.E. Quantitative structure–pharmacokinetic/pharmacodynamic relationships. Adv. Drug Deliv. Rev. 2006, 58, 1326–1356. [Google Scholar] [CrossRef]
  366. Guha, R. On exploring structure–activity relationships. In In Silico Models for Drug Discovery; Humana Press: Totowa, NJ, USA, 2013; pp. 81–94. [Google Scholar]
  367. Yan, G.; Rose, J.; Ellison, C.; Mudd, A.M.; Zhang, X.; Wu, S. Refine and strengthen SAR-based read-across by considering bioactivation and modes of action. Chem. Res. Toxicol. 2023, 36, 1532–1548. [Google Scholar] [CrossRef]
  368. Brogi, S.; Tabanelli, R.; Calderone, V. Combinatorial approaches for novel cardiovascular drug discovery: A review of the literature. Expert Opin. Drug Discov. 2022, 17, 1111–1129. [Google Scholar] [CrossRef] [PubMed]
  369. Zhang, C.; Chen, Y.-Y.; Chen, S.; Wang, Y.; Yuan, Y.; Yang, X.; Hu, W.; Chen, B.; Qi, Z.; Huse, J.T. Characterization of an Enhancer RNA Signature Reveals Treatment Strategies for Improving Immunotherapy Efficacy in Cancer. Cancer Res. 2025, 85, 1530–1543. [Google Scholar] [CrossRef]
  370. Günther, J.; Däbritz, J.; Wirthgen, E. Limitations and off-target effects of tryptophan-related IDO inhibitors in cancer treatment. Front. Immunol. 2019, 10, 1801. [Google Scholar] [CrossRef]
  371. Qian, S.; Zhang, M.; Chen, Q.; He, Y.; Wang, W.; Wang, Z. IDO as a drug target for cancer immunotherapy: Recent developments in IDO inhibitors discovery. RSC Adv. 2016, 6, 7575–7581. [Google Scholar] [CrossRef]
  372. Tanaka, M.; Bohar, Z.; Vecsei, L. Are kynurenines accomplices or principal villains in dementia? Maintenance of kynurenine metabolism. Molecules 2020, 25, 564. [Google Scholar] [CrossRef]
  373. Papathanasiou, T.; Strathe, A.; Hooker, A.C.; Lund, T.M.; Overgaard, R.V. Feasibility of exposure-response analyses for clinical dose-ranging studies of drug combinations. AAPS J. 2018, 20, 64. [Google Scholar] [CrossRef]
  374. Chakrabarti, S.; Michor, F. Pharmacokinetics and drug interactions determine optimum combination strategies in computational models of cancer evolution. Cancer Res. 2017, 77, 3908–3921. [Google Scholar] [CrossRef] [PubMed]
  375. Nowak-Sliwinska, P.; Weiss, A.; Ding, X.; Dyson, P.J.; Van Den Bergh, H.; Griffioen, A.W.; Ho, C.-M. Optimization of drug combinations using Feedback System Control. Nat. Protoc. 2016, 11, 302–315. [Google Scholar] [CrossRef]
  376. Tanaka, M.; Tóth, F.; Polyák, H.; Szabó, Á.; Mándi, Y.; Vécsei, L. Immune Influencers in Action: Metabolites and Enzymes of the Tryptophan-Kynurenine Metabolic Pathway. Biomedicines 2021, 9, 734. [Google Scholar] [CrossRef]
  377. Perez-Castro, L.; Garcia, R.; Venkateswaran, N.; Barnes, S.; Conacci-Sorrell, M. Tryptophan and its metabolites in normal physiology and cancer etiology. FEBS J. 2023, 290, 7–27. [Google Scholar] [CrossRef]
  378. Zuccarello, E.; Zhang, H.; Acquarone, E.; Pham, D.; Staniszewski, A.; Deng, S.X.; Landry, D.W.; Arancio, O.; Fiorito, J. Optimizing metabolic stability of phosphodiesterase 5 inhibitors: Discovery of a potent N-(pyridin-3-ylmethyl)quinoline derivative targeting synaptic plasticity. Bioorg. Med. Chem. Lett. 2023, 92, 129409. [Google Scholar] [CrossRef] [PubMed]
  379. de Lima, E.; Tanaka, M.; Lamas, C.; Quesada, K.; Detregiachi, C.; Araújo, A.; Guiguer, E.; Catharin, V.; de Castro, M.; Junior, E. Vascular Impairment, Muscle Atrophy, and Cognitive Decline: Critical Age-Related Conditions. Biomedicines 2024, 12, 2096. [Google Scholar] [CrossRef] [PubMed]
  380. Tanaka, M.; Battaglia, S.; Giménez-Llort, L.; Chen, C.; Hepsomali, P.; Avenanti, A.; Vécsei, L. Innovation at the Intersection: Emerging Translational Research in Neurology and Psychiatry. Cells 2024, 13, 790. [Google Scholar] [CrossRef]
  381. Jena, S.; Gonzalez, G.; Vítek, D.; Kvasnicová, M.; Štěpánková, Š.; Strnad, M.; Voller, J.; Chanda, K. Novel neuroprotective 5,6-dihydropyrido[2’,1’:2,3]imidazo[4,5-c]quinoline derivatives acting through cholinesterase inhibition and CB2 signaling modulation. Eur. J. Med. Chem. 2024, 276, 116592. [Google Scholar] [CrossRef]
  382. Behl, T.; Kaur, I.; Sehgal, A.; Singh, S.; Bhatia, S.; Al-Harrasi, A.; Zengin, G.; Bumbu, A.G.; Andronie-Cioara, F.L.; Nechifor, A.C.; et al. The Footprint of Kynurenine Pathway in Neurodegeneration: Janus-Faced Role in Parkinson’s Disorder and Therapeutic Implications. Int. J. Mol. Sci. 2021, 22, 6737. [Google Scholar] [CrossRef]
  383. Shen, H.; Xu, X.; Bai, Y.; Wang, X.; Wu, Y.; Zhong, J.; Wu, Q.; Luo, Y.; Shang, T.; Shen, R.; et al. Therapeutic potential of targeting kynurenine pathway in neurodegenerative diseases. Eur. J. Med. Chem. 2023, 251, 115258. [Google Scholar] [CrossRef] [PubMed]
  384. Pavlidis, N.; Kofinas, A.; Papanikolaou, M.G.; Miras, H.N.; Drouza, C.; Kalampounias, A.G.; Kabanos, T.A.; Konstandi, M.; Leondaritis, G. Synthesis, characterization and pharmacological evaluation of quinoline derivatives and their complexes with copper(ΙΙ) in in vitro cell models of Alzheimer’s disease. J. Inorg. Biochem. 2021, 217, 111393. [Google Scholar] [CrossRef]
  385. Barbalho, S.M.; Laurindo, L.F.; de Oliveira Zanuso, B.; da Silva, R.M.S.; Gallerani Caglioni, L.; Nunes Junqueira de Moraes, V.B.F.; Fornari Laurindo, L.; Dogani Rodrigues, V.; da Silva Camarinha Oliveira, J.; Beluce, M.E. AdipoRon’s Impact on Alzheimer’s Disease—A Systematic Review and Meta-Analysis. Int. J. Mol. Sci. 2025, 26, 484. [Google Scholar] [CrossRef]
  386. Adlard, P.A.; Cherny, R.A.; Finkelstein, D.I.; Gautier, E.; Robb, E.; Cortes, M.; Volitakis, I.; Liu, X.; Smith, J.P.; Perez, K.; et al. Rapid restoration of cognition in Alzheimer’s transgenic mice with 8-hydroxy quinoline analogs is associated with decreased interstitial Abeta. Neuron 2008, 59, 43–55. [Google Scholar] [CrossRef]
  387. Huang, W.; Liang, M.; Li, Q.; Zheng, X.; Zhang, C.; Wang, Q.; Tang, L.; Zhang, Z.; Wang, B.; Shen, Z. Development of the “hidden” multifunctional agents for Alzheimer’s disease. Eur. J. Med. Chem. 2019, 177, 247–258. [Google Scholar] [CrossRef]
  388. Tanaka, M.; Szabó, Á.; Körtési, T.; Szok, D.; Tajti, J.; Vécsei, L. From CGRP to PACAP, VIP, and Beyond: Unraveling the Next Chapters in Migraine Treatment. Cells 2023, 12, 2649. [Google Scholar] [CrossRef]
  389. Sarkar, S. Regulation of autophagy by mTOR-dependent and mTOR-independent pathways: Autophagy dysfunction in neurodegenerative diseases and therapeutic application of autophagy enhancers. Biochem. Soc. Trans. 2013, 41, 1103–1130. [Google Scholar] [CrossRef] [PubMed]
  390. Yin, S.; Liu, L.; Gan, W. The Roles of Post-Translational Modifications on mTOR Signaling. Int. J. Mol. Sci. 2021, 22, 1784. [Google Scholar] [CrossRef]
  391. Filomeni, G.; De Zio, D.; Cecconi, F. Oxidative stress and autophagy: The clash between damage and metabolic needs. Cell Death Differ. 2015, 22, 377–388. [Google Scholar] [CrossRef] [PubMed]
  392. Correia, A.S.; Vale, N. Tryptophan Metabolism in Depression: A Narrative Review with a Focus on Serotonin and Kynurenine Pathways. Int. J. Mol. Sci. 2022, 23, 8493. [Google Scholar] [CrossRef]
  393. Pathak, S.; Nadar, R.; Kim, S.; Liu, K.; Govindarajulu, M.; Cook, P.; Watts Alexander, C.S.; Dhanasekaran, M.; Moore, T. The Influence of Kynurenine Metabolites on Neurodegenerative Pathologies. Int. J. Mol. Sci. 2024, 25, 853. [Google Scholar] [CrossRef] [PubMed]
  394. Schwarcz, R.; Stone, T.W. The kynurenine pathway and the brain: Challenges, controversies and promises. Neuropharmacology 2017, 112, 237–247. [Google Scholar] [CrossRef]
  395. Sas, K.; Robotka, H.; Toldi, J.; Vécsei, L. Mitochondria, metabolic disturbances, oxidative stress and the kynurenine system, with focus on neurodegenerative disorders. J. Neurol. Sci. 2007, 257, 221–239. [Google Scholar] [CrossRef]
  396. Tanaka, M.; Vécsei, L. Revolutionizing our understanding of Parkinson’s disease: Dr. Heinz Reichmann’s pioneering research and future research direction. J. Neural Transm. 2024, 131, 1367–1387. [Google Scholar] [CrossRef]
  397. Battaglia, S.; Avenanti, A.; Vécsei, L.; Tanaka, M. Neurodegeneration in Cognitive Impairment and Mood Disorders for Experimental, Clinical and Translational Neuropsychiatry. Biomedicines 2024, 12, 574. [Google Scholar] [CrossRef]
  398. Battaglia, S.; Avenanti, A.; Vécsei, L.; Tanaka, M. Neural Correlates and Molecular Mechanisms of Memory and Learning. Int. J. Mol. Sci. 2024, 25, 2724. [Google Scholar] [CrossRef]
  399. Jászberényi, M.; Thurzó, B.; Bagosi, Z.; Vécsei, L.; Tanaka, M. The Orexin/Hypocretin System, the Peptidergic Regulator of Vigilance, Orchestrates Adaptation to Stress. Biomedicines 2024, 12, 448. [Google Scholar] [CrossRef] [PubMed]
  400. Manetti, F.; Maresca, L.; Crivaro, E.; Pepe, S.; Cini, E.; Singh, S.; Governa, P.; Maramai, S.; Giannini, G.; Stecca, B.; et al. Quinolines and Oxazino-quinoline Derivatives as Small Molecule GLI1 Inhibitors Identified by Virtual Screening. ACS Med. Chem. Lett. 2022, 13, 1329–1336. [Google Scholar] [CrossRef]
  401. Szabó, Á.; Galla, Z.; Spekker, E.; Szűcs, M.; Martos, D.; Takeda, K.; Ozaki, K.; Inoue, H.; Yamamoto, S.; Toldi, J.; et al. Oxidative and Excitatory Neurotoxic Stresses in CRISPR/Cas9-Induced Kynurenine Aminotransferase Knockout Mice: A Novel Model for Despair-Based Depression and Post-Traumatic Stress Disorder. Front. Biosci. 2025, 30, 25706. [Google Scholar] [CrossRef]
  402. Wang, S.W.; Gao, C.; Zheng, Y.M.; Yi, L.; Lu, J.C.; Huang, X.Y.; Cai, J.B.; Zhang, P.F.; Cui, Y.H.; Ke, A.W. Current applications and future perspective of CRISPR/Cas9 gene editing in cancer. Mol. Cancer 2022, 21, 57. [Google Scholar] [CrossRef] [PubMed]
  403. Li, X.; Ye, F.; Wang, Y.; Sun, X.; Chen, H.; Chen, T.; Gao, Y.; Chen, H. Synthesis, structure-activity relationship, and biological evaluation of quinolines for development of anticancer agents. Arch. Pharm. 2023, 356, e2200673. [Google Scholar] [CrossRef]
  404. Behan, F.M.; Iorio, F.; Picco, G.; Gonçalves, E.; Beaver, C.M.; Migliardi, G.; Santos, R.; Rao, Y.; Sassi, F.; Pinnelli, M.; et al. Prioritization of cancer therapeutic targets using CRISPR-Cas9 screens. Nature 2019, 568, 511–516. [Google Scholar] [CrossRef] [PubMed]
  405. Han, K.; Jeng, E.E.; Hess, G.T.; Morgens, D.W.; Li, A.; Bassik, M.C. Synergistic drug combinations for cancer identified in a CRISPR screen for pairwise genetic interactions. Nat. Biotechnol. 2017, 35, 463–474. [Google Scholar] [CrossRef]
  406. Ngai, C.; Sevian, H. Probing the Relevance of Chemical Identity Thinking in Biochemical Contexts. CBE Life Sci. Educ. 2018, 17, ar58. [Google Scholar] [CrossRef]
  407. Günther, D.; Boto, R.A.; Contreras-Garcia, J.; Piquemal, J.P.; Tierny, J. Characterizing Molecular Interactions in Chemical Systems. IEEE Trans. Vis. Comput. Graph. 2014, 20, 2476–2485. [Google Scholar] [CrossRef]
  408. Gallegos, L.C.; Luchini, G.; St John, P.C.; Kim, S.; Paton, R.S. Importance of Engineered and Learned Molecular Representations in Predicting Organic Reactivity, Selectivity, and Chemical Properties. Acc. Chem. Res. 2021, 54, 827–836. [Google Scholar] [CrossRef] [PubMed]
  409. Guan, K.; Wang, H.; Zhuang, L.; Sun, Y. Multi-granularity physicochemical-inspired molecular representation learning for property prediction. Expert Syst. Appl. 2025, 266, 126115. [Google Scholar] [CrossRef]
  410. Weyesa, A.; Mulugeta, E. Recent advances in the synthesis of biologically and pharmaceutically active quinoline and its analogues: A review. RSC Adv. 2020, 10, 20784–20793. [Google Scholar] [CrossRef]
  411. Palani, V.; Wendlandt, A.E. Strain-Inducing Positional Alkene Isomerization. J. Am. Chem. Soc. 2023, 145, 20053–20061. [Google Scholar] [CrossRef]
  412. Ishida, K.; Higashino, T.; Wada, Y.; Kaji, H.; Saeki, A.; Imahori, H. Thiophene-Fused Naphthodiphospholes: Modulation of the Structural and Electronic Properties of Polycyclic Aromatics by Precise Fusion of Heteroles. Chempluschem 2021, 86, 130–136. [Google Scholar] [CrossRef]
  413. Stone, T.W.; Williams, R.O. Modulation of T cells by tryptophan metabolites in the kynurenine pathway. Trends Pharmacol. Sci. 2023, 44, 442–456. [Google Scholar] [CrossRef]
  414. Mithaiwala, M.N.; Santana-Coelho, D.; Porter, G.A.; O’Connor, J.C. Neuroinflammation and the Kynurenine Pathway in CNS Disease: Molecular Mechanisms and Therapeutic Implications. Cells 2021, 10, 1548. [Google Scholar] [CrossRef] [PubMed]
  415. Erhardt, S.; Schwieler, L.; Imbeault, S.; Engberg, G. The kynurenine pathway in schizophrenia and bipolar disorder. Neuropharmacology 2017, 112, 297–306. [Google Scholar] [CrossRef]
  416. González Esquivel, D.; Ramírez-Ortega, D.; Pineda, B.; Castro, N.; Ríos, C.; Pérez de la Cruz, V. Kynurenine pathway metabolites and enzymes involved in redox reactions. Neuropharmacology 2017, 112 Pt B, 331–345. [Google Scholar] [CrossRef]
  417. Sutherland, J.J.; Yonchev, D.; Fekete, A.; Urban, L. A preclinical secondary pharmacology resource illuminates target-adverse drug reaction associations of marketed drugs. Nat. Commun. 2023, 14, 4323. [Google Scholar] [CrossRef]
  418. Casas, A.I.; Hassan, A.A.; Larsen, S.J.; Gomez-Rangel, V.; Elbatreek, M.; Kleikers, P.W.M.; Guney, E.; Egea, J.; López, M.G.; Baumbach, J.; et al. From single drug targets to synergistic network pharmacology in ischemic stroke. Proc. Natl. Acad. Sci. USA 2019, 116, 7129–7136. [Google Scholar] [CrossRef] [PubMed]
  419. Zhou, Y.; Zhang, Y.; Lian, X.; Li, F.; Wang, C.; Zhu, F.; Qiu, Y.; Chen, Y. Therapeutic target database update 2022: Facilitating drug discovery with enriched comparative data of targeted agents. Nucleic Acids Res. 2022, 50, D1398–D1407. [Google Scholar] [CrossRef]
  420. Ruiz, M.C.; Resasco, A.; Di Virgilio, A.L.; Ayala, M.; Cavaco, I.; Cabrera, S.; Aleman, J.; León, I.E. In vitro and in vivo anticancer effects of two quinoline-platinum(II) complexes on human osteosarcoma models. Cancer Chemother. Pharmacol. 2019, 83, 681–692. [Google Scholar] [CrossRef] [PubMed]
  421. Saxena, A.; Majee, S.; Ray, D.; Saha, B. Inhibition of cancer cells by Quinoline-Based compounds: A review with mechanistic insights. Bioorg. Med. Chem. 2024, 103, 117681. [Google Scholar] [CrossRef]
  422. Pich, O.; Bailey, C.; Watkins, T.B.K.; Zaccaria, S.; Jamal-Hanjani, M.; Swanton, C. The translational challenges of precision oncology. Cancer Cell 2022, 40, 458–478. [Google Scholar] [CrossRef] [PubMed]
  423. Zugazagoitia, J.; Guedes, C.; Ponce, S.; Ferrer, I.; Molina-Pinelo, S.; Paz-Ares, L. Current Challenges in Cancer Treatment. Clin. Ther. 2016, 38, 1551–1566. [Google Scholar] [CrossRef]
  424. Malone, E.R.; Oliva, M.; Sabatini, P.J.B.; Stockley, T.L.; Siu, L.L. Molecular profiling for precision cancer therapies. Genome Med. 2020, 12, 8. [Google Scholar] [CrossRef]
  425. Ioannidis, J.P.A.; Kim, B.Y.S.; Trounson, A. How to design preclinical studies in nanomedicine and cell therapy to maximize the prospects of clinical translation. Nat. Biomed. Eng. 2018, 2, 797–809. [Google Scholar] [CrossRef]
  426. Lin, J.; Sahakian, D.C.; de Morais, S.M.; Xu, J.J.; Polzer, R.J.; Winter, S.M. The role of absorption, distribution, metabolism, excretion and toxicity in drug discovery. Curr. Top. Med. Chem. 2003, 3, 1125–1154. [Google Scholar] [CrossRef]
  427. Liu, J.; Gui, Y.; Rao, J.; Sun, J.; Wang, G.; Ren, Q.; Qu, N.; Niu, B.; Chen, Z.; Sheng, X.; et al. In silico off-target profiling for enhanced drug safety assessment. Acta Pharm. Sin. B 2024, 14, 2927–2941. [Google Scholar] [CrossRef]
  428. David, P.O. Abstract A001: Navigating the future of therapeutic development: Innovations in safety and efficacy through advanced research methodologies and nanotechnology. Mol. Cancer Ther. 2024, 23, A001. [Google Scholar] [CrossRef]
  429. Son, A.; Park, J.; Kim, W.; Yoon, Y.; Lee, S.; Ji, J.; Kim, H. Recent Advances in Omics, Computational Models, and Advanced Screening Methods for Drug Safety and Efficacy. Toxics 2024, 12, 822. [Google Scholar] [CrossRef] [PubMed]
  430. Mor, A.; Tankiewicz-Kwedlo, A.; Ciwun, M.; Lewkowicz, J.; Pawlak, D. Kynurenines as a Novel Target for the Treatment of Inflammatory Disorders. Cells 2024, 13, 1259. [Google Scholar] [CrossRef] [PubMed]
  431. Tanaka, M. Beyond the boundaries: Transitioning from categorical to dimensional paradigms in mental health diagnostics. Adv. Clin. Exp. Med. 2024, 33, 1295–1301. [Google Scholar] [CrossRef]
  432. Tanaka, M.; Vécsei, L. A decade of dedication: Pioneering perspectives on neurological diseases and mental illnesses. Biomedicines 2024, 12, 1083. [Google Scholar] [CrossRef]
  433. Ryan, K.M.; Corrigan, M.; Murphy, T.M.; McLoughlin, D.M.; Harkin, A. Gene expression of kynurenine pathway enzymes in depression and following electroconvulsive therapy. Acta Neuropsychiatr. 2025, 37, e34. [Google Scholar] [CrossRef] [PubMed]
  434. Liu, M.; Wang, X.; Wang, L.; Ma, X.; Gong, Z.; Zhang, S.; Li, Y. Targeting the IDO1 pathway in cancer: From bench to bedside. J. Hematol. Oncol. 2018, 11, 100. [Google Scholar] [CrossRef] [PubMed]
  435. Tanaka, M. From Serendipity to Precision: Integrating AI, Multi-Omics, and Human-Specific Models for Personalized Neuropsychiatric Care. Biomedicines 2025, 13, 167. [Google Scholar] [CrossRef]
  436. Cadilla, R.; Deaton, D.N.; Do, Y.; Elkins, P.A.; Ennulat, D.; Guss, J.H.; Holt, J.; Jeune, M.R.; King, A.G.; Klapwijk, J.C.; et al. The exploration of aza-quinolines as hematopoietic prostaglandin D synthase (H-PGDS) inhibitors with low brain exposure. Bioorg. Med. Chem. 2020, 28, 115791. [Google Scholar] [CrossRef]
  437. Boros, F.A.; Bohár, Z.; Vécsei, L. Genetic alterations affecting the genes encoding the enzymes of the kynurenine pathway and their association with human diseases. Mutat. Res. Rev. Mutat. Res. 2018, 776, 32–45. [Google Scholar] [CrossRef]
  438. Tamimou, R.; Montout, C.; Mura, T.; Conejero, I.; Evrard, A.; Courtet, P.; Bonilla-Escribano, P.; Riaza, C.; Vaquero-Lorenzo, C.; Baca-Garcia, E.; et al. Genetic association of the kynurenine pathway to suicidal behavior. Brain Behav. Immun. Health 2024, 42, 100903. [Google Scholar] [CrossRef] [PubMed]
  439. Nemkov, T.; Stephenson, D.; Erickson, C.; Dzieciatkowska, M.; Key, A.; Moore, A.; Earley, E.J.; Page, G.P.; Lacroix, I.S.; Stone, M.; et al. Regulation of kynurenine metabolism by blood donor genetics and biology impacts red cell hemolysis in vitro and in vivo. Blood 2024, 143, 456–472. [Google Scholar] [CrossRef] [PubMed]
  440. Martin, J.H.; Galettis, P.; Flynn, A.; Schneider, J. Phenotype versus genotype to optimize cancer dosing in the clinical setting-focus on 5-fluorouracil and tyrosine kinase inhibitors. Pharmacol. Res. Perspect. 2024, 12, e1182. [Google Scholar] [CrossRef] [PubMed]
  441. Swenberg, J.A.; Fryar-Tita, E.; Jeong, Y.C.; Boysen, G.; Starr, T.; Walker, V.E.; Albertini, R.J. Biomarkers in toxicology and risk assessment: Informing critical dose-response relationships. Chem. Res. Toxicol. 2008, 21, 253–265. [Google Scholar] [CrossRef]
  442. Cores, Á.; Carmona-Zafra, N.; Clerigué, J.; Villacampa, M.; Menéndez, J.C. Quinones as Neuroprotective Agents. Antioxidants 2023, 12, 1464. [Google Scholar] [CrossRef]
  443. Wang, Z.; Hu, J.; Yang, X.; Feng, X.; Li, X.; Huang, L.; Chan, A.S.C. Design, Synthesis, and Evaluation of Orally Bioavailable Quinoline-Indole Derivatives as Innovative Multitarget-Directed Ligands: Promotion of Cell Proliferation in the Adult Murine Hippocampus for the Treatment of Alzheimer’s Disease. J. Med. Chem. 2018, 61, 1871–1894. [Google Scholar] [CrossRef]
  444. Pietta, P.G. Flavonoids as antioxidants. J. Nat. Prod. 2000, 63, 1035–1042. [Google Scholar] [CrossRef]
  445. Nunes, Y.C.; Mendes, N.M.; Pereira de Lima, E.; Chehadi, A.C.; Lamas, C.B.; Haber, J.F.; dos Santos Bueno, M.; Araújo, A.C.; Catharin, V.C.S.; Detregiachi, C.R.P. Curcumin: A golden approach to healthy aging: A systematic review of the evidence. Nutrients 2024, 16, 2721. [Google Scholar] [CrossRef]
  446. Galleano, M.; Verstraeten, S.V.; Oteiza, P.I.; Fraga, C.G. Antioxidant actions of flavonoids: Thermodynamic and kinetic analysis. Arch. Biochem. Biophys. 2010, 501, 23–30. [Google Scholar] [CrossRef]
  447. Venkanna, A.; Cho, K.H.; Dhorma, L.P.; Kumar, D.N.; Hah, J.M.; Park, H.G.; Kim, S.Y.; Kim, M.H. Chemistry-oriented synthesis (ChOS) and target deconvolution on neuroprotective effect of a novel scaffold, oxaza spiroquinone. Eur. J. Med. Chem. 2019, 163, 453–480. [Google Scholar] [CrossRef]
  448. Karale, U.B.; Shinde, A.; Gaikwad, V.R.; Kalari, S.; Gourishetti, K.; Radhakrishnan, M.; Poornachandra, Y.; Amanchy, R.; Chakravarty, S.; Andugulapati, S.B.; et al. Iron mediated reductive cyclization/oxidation for the generation of chemically diverse scaffolds: An approach in drug discovery. Bioorg. Chem. 2023, 139, 106698. [Google Scholar] [CrossRef] [PubMed]
  449. Kim, S.; Kim, D.; Hong, S.Y.; Chang, S. Tuning Orbital Symmetry of Iridium Nitrenoid Enables Catalytic Diastereo- and Enantioselective Alkene Difunctionalizations. J. Am. Chem. Soc. 2021, 143, 3993–4004. [Google Scholar] [CrossRef] [PubMed]
  450. Cubillos-Ruiz, A.; Guo, T.; Sokolovska, A.; Miller, P.F.; Collins, J.J.; Lu, T.K.; Lora, J.M. Engineering living therapeutics with synthetic biology. Nat. Rev. Drug Discov. 2021, 20, 941–960. [Google Scholar] [CrossRef]
  451. Liu, Z.; Cao, S.; Liu, M.; Kang, W.; Xia, J. Self-Assembled Multienzyme Nanostructures on Synthetic Protein Scaffolds. ACS Nano 2019, 13, 11343–11352. [Google Scholar] [CrossRef] [PubMed]
  452. Siefen, T.; Lokhnauth, J.; Liang, A.; Larsen, C.C.; Lamprecht, A. An ex-vivo model for transsynovial drug permeation of intraarticular injectables in naive and arthritic synovium. J. Control. Release 2021, 332, 581–591. [Google Scholar] [CrossRef]
  453. Lavallie, E.R.; Dorner, A.J.; Burczynski, M.E. Use of ex vivo systems for biomarker discovery. Curr. Opin. Pharmacol. 2008, 8, 647–653. [Google Scholar] [CrossRef]
  454. Stefaniak, J.; Huber, K.V.M. Importance of Quantifying Drug-Target Engagement in Cells. ACS Med. Chem. Lett. 2020, 11, 403–406. [Google Scholar] [CrossRef]
  455. Wu, K.; Kwon, S.H.; Zhou, X.; Fuller, C.; Wang, X.; Vadgama, J.; Wu, Y. Overcoming Challenges in Small-Molecule Drug Bioavailability: A Review of Key Factors and Approaches. Int. J. Mol. Sci. 2024, 25, 13121. [Google Scholar] [CrossRef]
  456. Stefanov, B.A.; Fussenegger, M. Biomarker-driven feedback control of synthetic biology systems for next-generation personalized medicine. Front. Bioeng. Biotechnol. 2022, 10, 986210. [Google Scholar] [CrossRef]
  457. Hassanzadeh, P.; Atyabi, F.; Dinarvand, R. Technical and engineering considerations for designing therapeutics and delivery systems. J. Control. Release 2023, 353, 411–422. [Google Scholar] [CrossRef]
Figure 1. The structure of quinoline. The quinoline scaffold, featuring a fused benzene and pyridine ring, forms the structural backbone of many biologically active compounds.
Figure 1. The structure of quinoline. The quinoline scaffold, featuring a fused benzene and pyridine ring, forms the structural backbone of many biologically active compounds.
Pharmaceuticals 18 00607 g001
Table 1. Biochemical properties of key kynurenine (KYN) pathway metabolites and nicotinic acid (NA). This table presents the primary physiological roles and biochemical features of these compounds, underscoring their relevance to neurotransmission, immunological regulation, and metabolic pathways.
Table 1. Biochemical properties of key kynurenine (KYN) pathway metabolites and nicotinic acid (NA). This table presents the primary physiological roles and biochemical features of these compounds, underscoring their relevance to neurotransmission, immunological regulation, and metabolic pathways.
CompoundsMain CharacteristicsRef.

Pharmaceuticals 18 00607 i001

Kynurenic Acid (KYNA)
Neuroprotective—Blocks NMDA and α7nAChR to reduce excitotoxicity
KYN pathway metabolite—Modulates neurotransmission and immune response
Poor BBB permeability—Limited CNS access, driving prodrug development
[30,51,52]

[53,54,55]

[56,57,58]

Pharmaceuticals 18 00607 i002

Nicotinic Acid (NA)
Essential vitamin (B3)—Crucial for NAD+/NADP+ synthesis, supporting cellular metabolism
Lipid-lowering effects—Reduces LDL and triglycerides while increasing HDL cholesterol
Modulation of nicotinic acid-induced flushing—GPR109A and Langerhans cells drive flushing; DP1 antagonists suppress it
[59,60,61]

[62,63,64]

[65,66,67]

Pharmaceuticals 18 00607 i003

Quinolinic Acid (QUIN)
Excitotoxic agent—Acts as a potent NMDA receptor agonist, leading to neuronal overexcitation and potential cell death
Oxidative stress contributor—Promotes free radical generation, exacerbating cellular damage
Neurodegenerative implications—Linked to the progression of neurological disorders through its detrimental effects on brain tissue
[68,69,70]

[71,72,73]

[69,74,75]

Pharmaceuticals 18 00607 i004

Picolinic Acid
Metal ion chelator—Binds zinc, iron, and other metals, influencing cellular metabolism
Immune modulator—Enhances macrophage activity and antimicrobial defense
Neuroactive compound—Involved in neurotransmission and potential neuroprotective effects
[76,77,78]

[79,80,81]

[82,83,84]

Pharmaceuticals 18 00607 i005

Xanthurenic Acid (XA)
Neuromodulatory effects: Influences neuronal signaling and may modulate receptor activity
Putative neuroactive role—Modulates glutamate signaling and may influence neurotransmission
Metal chelating properties—Binds with zinc and other metal ions, potentially affecting oxidative stress
[48,85,86]

[48,85,86]

[87,88,89]

Pharmaceuticals 18 00607 i006

Quinaldic Acid
Quinoline derivative—Structurally related to other KYN pathway metabolites
Antimicrobial properties—Exhibits activity against certain bacteria and fungi
Potential neuroactive role—May influence neurotransmission, though its biological significance remains underexplored
[90,91,92]

[93,94]

[92,95]
α7nAChR, α7 nicotinic acetylcholine receptor; BBB, blood–brain barrier; CNS, central nervous system; DP1, prostaglandin D2 receptor 1; GPR109A, G protein-coupled receptor 109A; HDL, high-density lipoprotein; LDL, low-density lipoprotein; NAD+/NADP+, reduced nicotinamide adenine dinucleotide/reduced nicotinamide adenine dinucleotide phosphate; NMDA, N-methyl-D-aspartate.
Table 2. Structural and pharmacological overview of quinoline, quinolone derivatives, and pyridinecarboxylic acids. Key compounds within these chemical classes are categorized alongside their main therapeutic actions, biological targets, and significant applications. This table traces the progression from classic antimalarials to contemporary antibiotics, emphasizing how structural changes shape efficacy, safety profiles, and clinical relevance.
Table 2. Structural and pharmacological overview of quinoline, quinolone derivatives, and pyridinecarboxylic acids. Key compounds within these chemical classes are categorized alongside their main therapeutic actions, biological targets, and significant applications. This table traces the progression from classic antimalarials to contemporary antibiotics, emphasizing how structural changes shape efficacy, safety profiles, and clinical relevance.
CompoundsMain CharacteristicsRef.
Quinoline and quinolone derivatives

Pharmaceuticals 18 00607 i007

Quinine

Pharmaceuticals 18 00607 i008

Quinidine
Quinine:
Antimalarial agent—Inhibits Plasmodium pp. by interfering with heme detoxification
Historically used to reduce fever and pain
Muscle relaxant—Helps alleviate nocturnal leg cramps by modulating ion channels

Quinidine:
Class I antiarrhythmic—Blocks sodium channels, stabilizing cardiac rhythm
Chiral isomer of quinine—Shares structural similarities but has distinct pharmacological effects
Proarrhythmic risk—Can prolong QT interval 1, requiring careful clinical use
[142,143,144]

[145,146,147]

[148,149]


[150,151,152]

[153,154,155]

[156,157,158]

Pharmaceuticals 18 00607 i009
Cinchoninic Acid
Quinoline derivative—Structurally related to quinine and other cinchona alkaloids
Metal chelation—Binds with metal ions, potentially influencing enzymatic activity
Pharmacological potential—Investigated for antimicrobial and neuroactive properties
[149,159,160]

[161,162]

[163,164,165]

Pharmaceuticals 18 00607 i010

2-Quinolinone

Pharmaceuticals 18 00607 i011

4-Quinolinone
Core scaffold for quinolone antibiotics—Forms the backbone of fluoroquinolones, targeting bacterial DNA gyrase and topoisomerase IV

Broad-spectrum antimicrobial activity—Effective against Gram-positive and Gram-negative bacteria

Synthetic versatility—Modifiable structure allows for improved potency, pharmacokinetics, and resistance mitigation
[166,167,168]



[169,170,171]


[170,172,173]


Pharmaceuticals 18 00607 i012

Nalidixic Acid
First-generation quinolone antibiotic—Inhibits DNA gyrase, primarily effective against Gram-negative bacteria
Limited spectrum and rapid resistance—Narrow activity and high bacterial resistance limit its clinical use
Urinary tract infection treatment—Historically used for urinary tract infections, though largely replaced by newer fluoroquinolones
[174,175,176]

[177,178]

[179,180,181]

Pharmaceuticals 18 00607 i013

Oxolinic Acid
Early quinolone antibiotic—Inhibits DNA gyrase, effective against Gram-negative bacteria
Used in veterinary medicine—Primarily employed for treating bacterial infections in animals
Limited clinical use—Replaced by newer fluoroquinolones due to resistance and pharmacokinetic limitations
[182,183,184]

[185,186,187]

[188]
Pyridinecarboxylic Acids

Pharmaceuticals 18 00607 i014

Isonicotinic Acid (INA)
Pyridine carboxylic acid derivative—Structurally related to NA (vitamin B3)
Key precursor for isoniazid—Used in the synthesis of isoniazid, a frontline anti-tuberculosis drug
Pharmacological potential—Investigated for antimicrobial and metabolic regulatory properties
[189,190]

[191,192]

[191,193,194]

Pharmaceuticals 18 00607 i015

Dipicolinic Acid (DPA)
Metal ion chelator—Strongly binds calcium and other metal ions, playing a role in metal homeostasis
Bacterial spore component—Essential for bacterial endospore resistance and heat stability
Potential neuroprotective role—Explored for its effects on metal-related oxidative stress in neurodegeneration
[195,196]

[197]

[198,199,200]
Hydroxy-substituted derivatives

Pharmaceuticals 18 00607 i016

8-Hydroxyquinoline
Metal chelator—Strongly binds iron, copper, and zinc, influencing redox balance and enzymatic activity
Antimicrobial and antifungal agent—Exhibits broad-spectrum activity against bacteria and fungi
Potential neuroprotective role—Investigated for treating neurodegenerative diseases by regulating metal toxicity
[201,202,203]

[204,205,206]

[207,208,209]
1 QT interval, time between onset of Q wave and end of T wave on electrocardiogram (ECG), reflecting ventricular depolarization and repolarization. NA, nicotinic acid.
Table 3. Synthetic exogenous quinoline-based compounds: structures and pharmacological profiles. Synthetic and exogenous quinoline derivatives are outlined here, with an emphasis on their primary mechanisms of action, therapeutic potential, and development hurdles. Each compound is linked to its intended molecular target—from NMDA receptor antagonism to immunomodulatory and anticancer functions—and is accompanied by references for in-depth exploration.
Table 3. Synthetic exogenous quinoline-based compounds: structures and pharmacological profiles. Synthetic and exogenous quinoline derivatives are outlined here, with an emphasis on their primary mechanisms of action, therapeutic potential, and development hurdles. Each compound is linked to its intended molecular target—from NMDA receptor antagonism to immunomodulatory and anticancer functions—and is accompanied by references for in-depth exploration.
CompoundsMain CharacteristicsRef.

Pharmaceuticals 18 00607 i017

7-Chlorokynurenic Acid (7-CKA)
NMDA receptor antagonist—Blocks the glycine-binding site, reducing excitotoxicity

Preclinical behavioral actions—Elicits antidepressant-like effects, blocks NMDA-induced convulsions, and attenuates ischemia-induced learning deficits

Phase II clinical trials have demonstrated that 4-chlorokynurenine, a prodrug of 7-CKA, lacks significant antidepressant efficacy in individuals with treatment-resistant depression. It is currently undergoing Phase III evaluation for hypertension and Phase I trials for neurological disorders and neuropathic pain. Additional clinical studies are planned to investigate its potential utility in epilepsy, Huntington’s disease, Parkinson’s disease, various psychiatric conditions, and suicidal ideation.
[253,254]



[255,256,257]




[125,258]


Pharmaceuticals 18 00607 i018

5,7-Dichlorokynurenic Acid
NMDA receptor antagonist—Blocks the glycine site to reduce excitotoxicity

Enhanced potency—More effective than KYNA at inhibiting NMDA receptor activity

Preclinical behavioral actions—Shows anxiolytic effects and enhances short-term memory and recognition

[259,260,261]

[246,259,261,262]

[263,264,265]


Pharmaceuticals 18 00607 i019

L-701,324
NMDA receptor antagonist—Blocks the glycine site, reducing excitotoxicity

Preclinical behavioral evidence—Demonstrates antidepressant-like activity, reduces anxiety-like behavior

Potential clinical application–Epilepsy, schizophrenia, and chronic pain
[266,267]

[265,268]

[269,270,271]

Pharmaceuticals 18 00607 i020

Gavestinel
Glycine site NMDA receptor antagonist—Blocks NMDA receptor activity to reduce excitotoxicity
Preclinical behavioral findings—Potential in reducing ischemic damage and modulating certain NMDA-receptor-mediated schizophrenia-like behaviors
Stroke neuroprotection candidate—Investigated for acute stroke, primary intracerebral hemorrhage, and acute ischemic stroke, but failed in clinical trials
[272,273]

[272,274]

[272,275,276]

Pharmaceuticals 18 00607 i021

Laquinimod
Immunomodulatory agent—Reduces pro-inflammatory cytokines and modulates immune cell activity

Preclinical findings—Improves motor function in a Huntington’s disease model, activates the AhR in the EAE Model of MS
Clinical trials—Modestly reduces relapse rates and disability progression and significantly reduces brain volume change atrophy in relapsing–remitting MS, and shosw limited efficacy in active non-infectious intermediate, posterior, or panuveitis (NCT02720102)
[277,278,279]



[280,281]

[282,283,284]

Pharmaceuticals 18 00607 i022


Tasquinimod
Anti-cancer properties—Inhibits tumor angiogenesis, suppresses myeloid-derived suppressor cells, downregulates immune suppressive pathways and inflammatory cytokine signaling
S100A9 modulation—Suppresses inflammatory factor expression, potentially inhibist the upregulation of S100A9 in AD, and inhibits MDSC recruitment
HDAC4 modulation—potentially influences epigenetic regulation in neurodegenerative or cognitive disorders and potentially controls neuronal memory, plasticity, and learning
[285,286,287]

[288,289,290]

[291,292]
AD, Alzheimer’s disease; HDAC, histone deacetylase; AhR, aryl hydrocarbon receptor; EAE, experimental autoimmune encephalomyelitis; MS, multiple sclerosis; MDSCs, myeloid-derived duppressor cells.
Table 4. Proposed strategies for consolidating and cross-referencing structure–activity relationship (SAR) data.
Table 4. Proposed strategies for consolidating and cross-referencing structure–activity relationship (SAR) data.
RecommendationApproachIllustrative Examples
Create a Unified SAR OverviewGather scattered substituent data (chlorine, fluorine, sulfonamides, etc.) in a dedicated section or table.Summarize each motif (e.g., Cl, F, sulfonamide) and its known effects on receptor affinity or metabolism.
Cross-Reference Key Examples and MotifsExplicitly reference primary compounds (7-CKA, Gavestinel) to illustrate general rules about halogenation or sulfonamide additions.For halogens (Cl, F): provide examples showing increased receptor blockade or enhanced lipophilicity (7-CKA, 5,7-DCKA).
SAR, structure–activity relationship; 7-CKA, 7-chlorokynurenic acid; 5,7-DCKA, 5,7-dichlorokynurenic acid.
Table 5. Kynurenic acid (KYNA) analogues from the SZR series: structural variations and pharmacological insights. The primary SZR-series analogues derived from KYNA are summarized here, highlighting their structural modifications and core pharmacological properties. The data emphasize their therapeutic relevance to excitotoxicity, neurodegenerative diseases, and related conditions, with references provided for deeper investigation.
Table 5. Kynurenic acid (KYNA) analogues from the SZR series: structural variations and pharmacological insights. The primary SZR-series analogues derived from KYNA are summarized here, highlighting their structural modifications and core pharmacological properties. The data emphasize their therapeutic relevance to excitotoxicity, neurodegenerative diseases, and related conditions, with references provided for deeper investigation.
CompoundsMain CharacteristicsRef.

Pharmaceuticals 18 00607 i023

SZR-72
BBB penetration—effectively crosses the blood–brain barrier, enhancing its therapeutic potential in the CNS
Neuroprotective effect—Offers enhanced neuroprotection, reduces brainstem c-fos, and induces therapeutic hypothermia
Behavioral effects—Alters motor and exploratory behaviors, reducing vertical activity and affecting curiosity-linked emotional and motor responses
[342,343]

[341,342]

[343]


Pharmaceuticals 18 00607 i024

SZR-73
Mitochondrial function enhancement—Enhances mitochondrial respiration and ATP production, improving complex I- and II-linked OXPHOS in a rodent sepsis model
Systemic inflammatory activation reduction—Reduces systemic inflammation in sepsis, lowering ET-1, IL-6, and NT levels and XOR activity
Microcirculatory effects—Improves mitochondrial function but does not restore microcirculation, unlike KYNA, which improves microvascular perfusion in sepsis
[229]


[229]


[229]

Pharmaceuticals 18 00607 i025

SZR-81
Antidepressant-like effects—Reduces immobility and increasing swimming in FST via serotonin 5-HT system involvement
BBB permeability—Poor BBB penetration or metabolic alteration before reaching the CNS
Neuroprotective potential–To be explored for use in stroke, neurodegenerative diseases, and inflammatory conditions
[347]



[347]


[348]

Pharmaceuticals 18 00607 i026


SZR-104
High permeability through the blood—Shows high BBB permeability, surpassing KYNA and analogues
Neuroprotective effects in sepsis—Demonstrates neuroprotection in septic rodents, reducing BBB disruption and CNS mitochondrial dysfunction
Influence on motor behavior—Increases horizontal exploration, indicating BBB penetration and retention of KYNA-like effects on motor-related curiosity and emotional behavior
[56]


[344]


[343]

Pharmaceuticals 18 00607 i027

SZR-105
High BBB penetrability—Significantly better BBB permeability than KYNA and earlier analogues due to its dual cationic side chains
Potent anti-inflammatory effects—Suppresses TNF-α production and strongly induces TSG-6
Neuroprotective activity in CNS models—Reduces cortical spreading depression propagation in migraine models
[56]

[345]

[346]

Pharmaceuticals 18 00607 i028

SZR-109
Enhanced BBB penetration—penetrates the BBB more effectively than KYNA
Neuroprotective effects—Suppresses pro-inflammatory TNF-α production and upregulates TSG-6
Anti-convulsant activity—prevents or diminishes seizure-like activity in the brain
[56]

[345]

[346]

Pharmaceuticals 18 00607 i029

SZRG-21
BBB penetration—Demonstrated efficacy in reducing cytokine levels in colitis models
Impact on motor activity—Alters motor activity and exploratory behavior

Emotional and behavioral modulation—Affects emotional functions, such as motor-associated curiosity and emotions
[343]


[343]


[343]
ATP, adenosine triphosphate; BBB, blood–brain barrier; CNS, central nervous system; ET-1; endothelin-1; FST, forced swim test; 5-HT, 5-hydroxytryptamine; IL-6, interleukin 6; KYNA, kynurenic acid; NT, N-terminal peptide; OXPHOS, oxidative phosphorylation; TNF-α, tumor necrosis factor-alpha; TSG-6, tumor necrosis factor-stimulated gene-6; XOR, xanthine oxidoreductase.
Table 6. Four pillars for advancing kynurenic acid (KYNA)-based drug design.
Table 6. Four pillars for advancing kynurenic acid (KYNA)-based drug design.
PillarPrimary ObjectivesIllustrative Approaches
Pinpoint Disease PathwaysLink structural motifs to specific therapeutic outcomes.Combat excitotoxicity in Alzheimer’s
Refine SAR StrategiesOptimize structure–activity to boost efficacy and reduce risks.Use computational docking
Validate in Context-Specific ModelsConfirm mechanistic relevance in robust disease models.Test in transgenic mice (e.g., Parkinson’s)
Incorporate Biomarker-Based SelectionUse metabolic/genomic markers for targeted therapy.Stratify patients by IDO/KMO polymorphisms
IDO, indoleamine 2,3-dioxygenase; KMO, kynurenine 3-monooxygenase.
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

Tanaka, M.; Szatmári, I.; Vécsei, L. Quinoline Quest: Kynurenic Acid Strategies for Next-Generation Therapeutics via Rational Drug Design. Pharmaceuticals 2025, 18, 607. https://doi.org/10.3390/ph18050607

AMA Style

Tanaka M, Szatmári I, Vécsei L. Quinoline Quest: Kynurenic Acid Strategies for Next-Generation Therapeutics via Rational Drug Design. Pharmaceuticals. 2025; 18(5):607. https://doi.org/10.3390/ph18050607

Chicago/Turabian Style

Tanaka, Masaru, István Szatmári, and László Vécsei. 2025. "Quinoline Quest: Kynurenic Acid Strategies for Next-Generation Therapeutics via Rational Drug Design" Pharmaceuticals 18, no. 5: 607. https://doi.org/10.3390/ph18050607

APA Style

Tanaka, M., Szatmári, I., & Vécsei, L. (2025). Quinoline Quest: Kynurenic Acid Strategies for Next-Generation Therapeutics via Rational Drug Design. Pharmaceuticals, 18(5), 607. https://doi.org/10.3390/ph18050607

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