Next Article in Journal
Insight into Rheological Properties and Structure of Native Waxy Starches: Cluster Analysis Grouping
Next Article in Special Issue
Formation and Reactions of Brønsted and Lewis Acid Adducts with Electron-Rich Heteroaromatic Compounds
Previous Article in Journal
Monitoring Ammonium Polyphosphate (APP) Biodegradation by Acinetobacter nosocomialis D-3 Using DAPI
Previous Article in Special Issue
Palladium-Catalyzed Arylations towards 3,6-Diaryl-1,3a,6a-triazapentalenes and Evaluation of Their Fluorescence Properties
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Imidazopyridine Family: Versatile and Promising Heterocyclic Skeletons for Different Applications

Department of Chemistry, University of Turin, Via Pietro Giuria 7, 10125 Torino, Italy
*
Author to whom correspondence should be addressed.
Molecules 2024, 29(11), 2668; https://doi.org/10.3390/molecules29112668
Submission received: 3 May 2024 / Revised: 30 May 2024 / Accepted: 31 May 2024 / Published: 5 June 2024
(This article belongs to the Special Issue Heterocyclic Chemistry in Europe)

Abstract

:
In recent years, there has been increasing attention focused on various products belonging to the imidazopyridine family; this class of heterocyclic compounds shows unique chemical structure, versatile optical properties, and diverse biological attributes. The broad family of imidazopyridines encompasses different heterocycles, each with its own specific properties and distinct characteristics, making all of them promising for various application fields. In general, this useful category of aromatic heterocycles holds significant promise across various research domains, spanning from material science to pharmaceuticals. The various cores belonging to the imidazopyridine family exhibit unique properties, such as serving as emitters in imaging, ligands for transition metals, showing reversible electrochemical properties, and demonstrating biological activity. Recently, numerous noteworthy advancements have emerged in different technological fields, including optoelectronic devices, sensors, energy conversion, medical applications, and shining emitters for imaging and microscopy. This review intends to provide a state-of-the-art overview of this framework from 1955 to the present day, unveiling different aspects of various applications. This extensive literature survey may guide chemists and researchers in the quest for novel imidazopyridine compounds with enhanced properties and efficiency in different uses.

1. Introduction

The imidazopyridines are heterocyclic compounds formed by the fusion of an imidazole ring with a pyridine ring. Figure 1 illustrates the various possible combinations of the imidazole and pyridine rings, resulting in different nuclei, all identified and documented in the literature as imidazopyridines. These compounds are primarily distinguished from each other by their geometric arrangements and the bonding between the two aromatic heterocycles.
According to current nomenclature, imidazopyridines are generally categorized into various groups based on the position of the nitrogen atoms and the distinct connections between the imidazole and pyridine rings (refer to Figure 1). Despite potential variations in composition or nitrogen atom count across individual structures (e.g., imidazo[1,2-a]pyridine and imidazo[4,5-c]pyridine), they all fall under the classification of imidazopyridines [1,2,3,4]. Each imidazopyridine nucleus differs in its peculiar biological, physical, and chemical properties, and these diverse behaviors imply various fields of application [5,6,7].
Furthermore, imidazopyridines can be modified by the insertion of functional groups to alter their properties; the accessibility of each structural position varies depending on the specific family being considered.
Among the different families, the recent scientific literature highlights three main application areas for these promising aromatic heterocycles:
  • Imidazo[1,2-a]pyridines are principally studied and applied in pharmacology and imaging, as modulators of the central nervous system, and as a biologically active core for the development of various commercial drugs [8,9,10,11,12].
  • Imidazo[1,5-a]pyridines are investigated and utilized for their emissive properties (serving as fluorophores) [13,14,15] or chelating ligands for metallic ions, contributing to the development of on–off-type sensors [16,17,18,19,20,21,22].
  • Imidazo[4,5-b]pyridines are principally studied for the development of emissive products for OLEDs (organic light-emitting diodes) and sensor applications due to their excellent emissive properties [23,24,25,26,27,28].
  • Imidazo[4,5-c]pyridines display a bioisosteric resemblance to the purine nucleus, manifesting similar structural and electronic properties. This structural affinity facilitates imidazopyridine’s facile interaction with macromolecules such as DNA, RNA, and specific proteins. Moreover, imidazo[4,5-c]pyridines exhibit notable cytotoxic activity by selectively inhibiting kinase activity [29,30,31].
Moreover, imidazoquinolines correspond to various structures (such as imidazo[5,1-a]quinolines and imidazo[2,1-a]isoquinolines) in which a third aromatic ring connects to the imidazopyridine nucleus. These derivatives represent the extension of the aromatic system of the classic imidazopyridines; the extended delocalized system alters their optical, structural, and solubility properties, sometimes presenting a promising structural alternative to the common heterocyclic ring such as phenanthroline, carbazole, benzoquinoline, acridine, and many others [32,33,34,35,36,37,38,39,40,41,42].
Numerous recent publications have underscored new synthetic strategies leading to promising optical properties, compelling many researchers to delve deeper into exploring this class of emissive compounds. Thanks to innovative methodologies, they have introduced various chemical groups aimed at conjugation and modulation of optical properties [5,43,44,45,46].
In recent years, many works focused on imidazopyridine derivatives have been published, describing different synthetic cyclization strategies or structural modifications. Generally, the imidazopyridine skeleton has been designed to obtain structural derivatives such as luminescent probes and ligands for transition metal ions or to increase pharmaceutical and biological activities. In particular, some works were focused on the connection of imidazopyridine with other fluorophores (such as boron-dipyrromethene, BODIPY or boron-dipyrromethene, and rhodamine) or on their activation as the fluorogenic center itself [8,47,48,49].
In general, the distinctive emission of the imidazopyridine derivatives is commonly found in the range of 430–520 nm, with a broad Stokes shift (up to 80 nm) and high values of quantum yield (ranging from 5% to 60%). Due to their structural flexibility and tunability of optical properties, imidazopyridine fluorophores are excellent candidates for various technological applications such as downshifting and optoelectronic devices [50,51,52]. G. Albrecht et al. recently investigated the electroluminescence of imidazopyridine derivatives utilizing layered OLEDs constructed with these emitting molecules [39,40,53,54]. Until now, only copper complexes derived from imidazopyridine ligands have proven effective in light-emitting electrochemical cells [55,56,57].
Moreover, imidazopyridine-based emitters offer a compelling alternative for attaining robust emission in fluorescence-based microscopy and confocal microscopy. Typically, imidazopyridine derivatives demonstrate favorable solubility, intense emission, high quantum yield, pH sensitivity, and strong biocompatibility. In recent years, imidazopyridine fluorophores have been successfully tested for obtaining detailed images through fluorescence microscopy and confocal microscopy [5,58,59,60,61,62,63,64].
Moreover, imidazopyridines present other areas of interest beyond their use as fluorophores. Indeed, among aromatic heterocycles, imidazopyridines have experienced a significant surge in interest in the literature due to their wide variety of applications in pharmaceutical chemistry. Although structurally different, imidazopyridines exhibit pharmacological properties quite similar to benzodiazepines [37,65,66,67]. Compounds containing the imidazopyridine moiety exhibit a diverse array of captivating biological characteristics, including antifungal, antipyretic, antiviral, analgesic, antibacterial, anticancer, antiprotozoal, anti-inflammatory, anticonvulsant, anthelmintic, antitubercular, antiulcer, antiepileptic, hypnotic, and anxiolytic properties [18,68,69,70,71,72]. They have also been the subject of investigation as receptor agonists. The imidazopyridine nucleus, in particular the imidazo[1,2-a]pyridines, is also present in several commercially available drugs, as reported in Figure 2, such as zolpidem, alpidem, saripidem, necopidem, olprinone, miroprofene, zolimidine, and many others [28,66,73,74,75,76,77,78,79,80,81].
Therefore, it is not surprising that the number of scientific articles related to the synthesis and application of this family of aromatic heterocycles has exponentially increased in the last decade. In recent years, notable advancements have been made in the heterocyclization and structural alteration of imidazopyridine derivatives through diverse methodologies, including, among others, multicomponent reactions, tandem sequences, transition metal-catalyzed C-H functionalizations, and one-pot syntheses [5,7,12,46,82,83]. These methods offer easy access to imidazopyridine products and their functionalizations starting from simple and readily available precursors.
The objective of this article is to compile and present the documented applications of numerous compounds belonging to the imidazopyridine family. Instead of focusing solely on a specific heterocyclic nucleus, the aim is to offer a comprehensive overview of this promising heterocyclic family, which holds potential for diverse biological and technological applications [6,45,84,85]. The target audience includes researchers already engaged in this field, as well as newcomers seeking insights.

2. Studies and Applications of Imidazopyridine Derivatives

2.1. Imidazopyridines in Medicine

A detailed survey of the literature has revealed that various classes of imidazopyridines can act as potent modulators toward several pathologies associated with central nervous system dysfunction, including Parkinson’s and Alzheimer’s diseases, depression, schizophrenia, or sleep disorders. The recent literature described in detail the interaction of imidazopyridine derivatives with different enzymes (e.g., leucine-rich repeat kinase, β-secretase, fatty acid amide hydrolase, γ-secretase), receptors (e.g., adenosine A2A, GABA-A (γ-Aminobutyric acid), histamine, serotonin H3, 5-HT35-HT4, 5-HT6, and dopamine D4), and their therapeutic role [16]. In the field of central nervous system (CNS) modulators, the era of imidazopyridines began in the late 1980s and further developed starting in 1992 with the synthesis and approval of the drug zolpidem based on the imidazopyridine nucleus [66,86]. In the subsequent decades, numerous other imidazopyridine-based drugs have been introduced to the market (see Figure 2). Recent research has concentrated on molecular targets within the central nervous system (CNS), specifically linked to diverse neurodegenerative conditions and psychiatric disorders, where imidazopyridines demonstrate intriguing modulation potential [16,65,87,88,89].
Certainly, up to now, the most investigated and utilized imidazopyridines are associated with the imidazo[1,2-a]pyridine nucleus, particularly for pharmaceutical and medical applications [84,85,90]. These derivatives exhibit a wide range of biological activities, such as cardiotonic, antifungal, anti-inflammatory, antipyretic, analgesic, antitumor, antiapoptotic, hypnotic, antiviral, antibacterial, antiprotozoal, and anxiolytic agents. Moreover, they act as inhibitors of β-amyloid formation, agonists of GABA, and benzodiazepine receptors [1,37,66,91,92,93]. Several drugs based on imidazo[1,2-a]pyridine derivatives are available in the market, such as zolpidem (used in the treatment of insomnia), alpidem (an anxiolytic agent), olprinone (for the treatment of acute heart failure), zolimidine (used for the treatment of peptic ulcer), necopidem, and saripidem (both acting as anxiolytic agents), all containing the imidazo[1,2-a]pyridine unit. Additionally, GSK812397 is a drug for HIV infection treatment, and the antibiotic rifaximin also contains this peculiar heterocyclic skeleton [7,43,45,94].
Different imidazo[1,2-a]pyridine derivatives exhibit excellent brain permeability and ideal metabolic stability. These data strongly suggest that imidazo[1,2-a]pyridine derivatives labeled with iodine-123 can be efficiently employed as potential imaging agents for single-photon emission computed tomography (SPECT) to detect amyloid pathology in the brains of patients with Alzheimer’s disease [17,95]. Recent studies have demonstrated that these compounds exhibit peculiar anticancer activity and could be valuable in developing more effective drugs with antiproliferative activity against lung, breast, and cervical cancer cells [96,97,98,99,100].
Among the pharmacologically relevant imidazopyridines, the group of imidazo[1,5-a]pyridines, despite being mostly employed for their luminescent features, has yielded interesting results. Currently, there are no commercial drugs containing this unit, but many studies have demonstrated the potential applicability of imidazo[1,5-a]pyridines in pharmacology. In Figure 3, diverse biologically active derivatives of imidazo[1,5-a]pyridine are depicted, including agonists of cannabinoid receptor type 2 (CB2R) (compound 1), serotonin 5-hydroxytryptamine (5-HT4) antagonists (compound 2), inhibitors of hypoxia-inducible factor 1α (HIF-1α) (compound 3), inhibitors of indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) (compound 4), cribrostatin-6 (compound 5), phosphodiesterase 10A inhibitors (compound 6), tubulin polymerization inhibitors (compound 7), neurokinin antagonists, kinase inhibitors, and various chemotherapeutic agents that have been evaluated [65,75,76,87,92,101,102,103,104,105,106,107]. In addition, in the past, this nucleus has been investigated as a selective, nonsteroidal inhibitor of aromatase for the treatment of estrogen-dependent disease [108].
Other important examples of imidazopyridines with significant pharmacological implications are as follows:
  • Imidazo[1,2-a]quinoline was investigated for its antitumor activity, DNA binding, and antitumor evaluation [109,110,111].
  • Imidazo[4,5-b]pyridine described in a review focused on the synthesis and pharmacological use of this particular imidazopyridine [30], in particular as an inhibitor of the B-Raf kinase [112].
  • Imidazo[4,5-c]pyridines were investigated for their antitumor activity [31] and notable cytotoxic, antiviral, and antimicrobial activity, which are inhibitors of Bruton’s tyrosine kinase enzyme activity [113,114,115].
  • Imidazo[1,2-b]pyridazines represent an important class of heterocyclic nuclei that provide various bioactive molecules. Among these, the successful kinase inhibitor ponatinib has sparked renewed interest in exploring new imidazo[1,2-b]pyridazine derivatives for their potential therapeutic applications in medicine, including anticancer agents, diagnostic tools for neuropathic diseases, thymic enhancers, antiparasitic, antibacterial, and antiviral agents, anti-inflammatory agents, and treatments for circadian rhythm sleep disorders [116,117,118,119].

2.2. Imidazopyridines as Ligands toward Different Metal Ions

Imidazopyridines are versatile and unique molecules in terms of their coordination toward metal ions. They are easily functionalizable by introducing substituents in different positions and can act as ligands toward different metal ions showing varying geometries and bonding angles. The optoelectronic properties of these imidazopyridine derivatives are noteworthy, as compounds derived from this nucleus display distinctive photophysical traits and robust stability under various experimental conditions. Consequently, this class of heterocyclic derivatives is garnering interest from researchers across diverse fields of study worldwide.
While imidazo[1,2-a]pyridines have found significant applications as drugs or in the treatment of various pathologies, imidazo[1,5-a]pyridines have attracted more interest concerning the formation of organometallic complexes and the study of the optical properties of these products and their applications. In general, extensive literature on transition metal complexes with imidazole or pyridine nuclei is available. Similarly, the imidazopyridinic skeleton can coordinate a wide variety of transition metal ions, yielding numerous metal complexes with diverse coordination motifs.
The metal–ligand chemistry associated with these heterocycles has been extensively studied, involving the formation of metal complexes featuring bidentate or tridentate structures. Various imidazopyridines have been effectively incorporated at numerous positions along a central aromatic ring, a critical structural characteristic ensuring geometrical equivalency with the more extensively researched multi-pyridinic or multi-bipyridinic ligands in coordination chemistry [120]. Multi-imidazopyridines have already been employed as ditopic ligands in binuclear complexes [48,121,122,123,124,125,126,127,128].
The imidazo[1,5-a]pyridinic skeleton, serving as a ligand, introduces a novel polyazine framework featuring diverse and adjustable coordination motifs adaptable for mono-, di-, and tritopic coordination sites. These molecules exhibit potential as polydentate ligands or precursors for supramolecular designs. Furthermore, the phenylimidazole unit has been observed to function as a cyclometalating ligand, akin to the conventional 2-phenylpyridine. In this scenario, a pendant phenyl group is appended at position 1 on the imidazolic nucleus, yielding a suitable C-N ligand, successfully employed with various metals (see Figure 4 for complete numeration of the imidazopyridine nuclei).
To date, there are no reported instances of utilizing the imidazo[1,5-a]pyridinic skeleton as a C-N ligand, despite its structural resemblance to phenylimidazole or the more traditional 2-phenylpyridine. Typically, the nitrogen of the imidazole ring can act as a monodentate ligand, like common pyridine, albeit with only limited binding capacity and stability. It is indeed the substituted imidazopyridines at positions 1 or 3 with suitable substituents that represent the best ligands obtained from this class of compounds.
Similarly, the imidazo[1,5-a]pyridinic nucleus, substituted at position 1 with a pendant pyridinic group, has been utilized for coordinating transition metal ions as a chelating N-N ligand (see Figure 5) [125,129,130,131]. Specifically, the inclusion of a pendant pyridinic group at position 1 on the imidazo[1,5-a]pyridinic skeleton (as opposed to introducing, for instance, a pendant phenyl group) enhances the quantum yield values [132]. Moreover, the notable presence of the pendant pyridinic group ensures the characteristic bidentate N-N ligand ability [126,133,134,135,136,137]. This motif is widely recognized for facilitating stable complexation with diverse metals.
In a different scenario, the imidazo[1,5-a]pyridinic nucleus can be employed as an N-O ligand if an appropriate substituent is previously introduced at position 1 on the imidazo[1,5-a]pyridinic unit (see Figure 5); this type of ligand has been coordinated to Ni, Co, and Pd, with the purpose to prepare emissive products for optoelectronic devices [134,135,138].
Furthermore, significant effort has been directed towards the advancement of luminescent boron complexes, such as BODIPY, the most renowned luminescent organo-boron compound. However, only a limited number of boron complexes based on the N-O chelating imidazopyridine ligand have been documented [8,20,122,137,139].
Zinc complexes have been successfully synthesized to produce fluorescent materials for optoelectronic devices. In each instance, a comparison of the optical properties between free imidazopyridinic ligands and their corresponding Zn(II) complexes reveals a notable enhancement in quantum yield and an intriguing hypsochromic shift. This shift arises from structural alterations in the conformation of imidazo[1,5-a]pyridinic ligands during zinc coordination and subsequent complexation reactions [36,125,136,138,140,141,142,143].
In a prior study focusing on the catalytic dimerization of ethylene, nickel complexes based on imidazo[1,5-a]pyridine derivatives were synthesized and characterized [144,145,146].
Copper complexes have undergone extensive investigation in OLED and LEC (Light-emitting Electrochemical Cell) devices, showcasing notable yellow electrochemical emission in cells employing heteroleptic Cu(I) complexes emitting in blue [55]. Despite this, the development of new Cu(I) complexes for blue LEC design remains largely unexplored. In this context, imidazopyridinic derivatives emerge as a promising alternative to complexes based on the more widely recognized bipyridine and phenanthroline [39,40,53,54,55,57,147,148].
Iron and cobalt complexes based on imidazopyridine have been synthesized and structurally characterized [149,150,151,152,153]; initial findings suggest the spontaneous resolution of enantiomers during crystallization, indicative of a rare p-donor/p-acceptor chiral recognition mechanism [154].
The design and self-assembly of chiral complexes have garnered significant interest due to their relevance in various enantioselective processes such as asymmetric catalysis, chemical sensing, and selective guest inclusion. Although spontaneous resolution is considered a more economical and convenient method, it is relatively rare and less predictable because the underlying conglomeration processes are not fully understood.
Imidazo[1,5-a]pyridinic chelating ligands have been effectively utilized to form complexes with iridium and rhenium, resulting in emissive products [155,156,157,158]. Computational calculations in these instances reveal emission primarily centered on the ligand, displaying minimal charge transfer character or dual emission. The employment of DFT (Density Functional Theory) and TDDFT (Time-dependent Density Functional Theory) calculations has played a pivotal role in elucidating the distinctions among these imidazo[1,5-a]pyridine-based complexes, their structures, and excited states. These investigations underscore the potential of Re and Ir complexes for optical applications, as the excited states of their metal complexes can be readily manipulated by altering substituents on the imidazo[1,5-a]pyridinic ligand nucleus [10,155,157,159]. Finally, cadmium, osmium, and ruthenium have been successfully complexed with imidazo[1,5-a]pyridinic-based ligands, yielding interesting products suitable for supramolecular chemistry [133,160,161,162,163].
Currently, no examples of complexation of other imidazopyridine derivatives have been reported, except for imidazo[1,2-a]pyridines, for which some references are available concerning ruthenium complexes used as catalysts [164] and highly emissive zinc complexes [165,166,167].

2.3. Imidazopyridines Applications in On–Off-Type Sensors

Detecting and monitoring hazardous contaminants like pesticides, metals, and anions within acceptable environmental and cellular limits is crucial for assessing environmental quality (air, water, soil) and understanding their harmful effects on living organisms. The health of organisms is directly or indirectly impacted by these contaminants. Consequently, developing sensors capable of detecting such contaminants is vital for protecting ecosystems and living organisms. To this purpose, among the various sensing techniques employed, optical methods based on fluorescence properties represent an excellent approach. Fluorescent probes have gained attention due to their ability to monitor molecular interactions in real time with high sensitivity, selectivity, direct visualization, and short response time, and without interfering with biological samples. A wide array of fluorescent probes, including naphthalimide, fluorescein, BODIPY, coumarin, rhodamine, cyanine, and, more recently, imidazopyridine-related examples, are available for this purpose [8,48,168,169].
Thennarasu and his group reported a chemosensor based on the imidazo[1,2-a]pyridinic nucleus for the sequential detection of Cu2+ and CN ions using fluorimetry [26]. The studied compound exhibits a characteristic absorption band at 322 nm. Upon the addition of various metal ions, a slight shift in the fluorescence spectrum was observed, whereas the Cu2+ ion nearly completely quenched the fluorescence. Subsequently, among the various anions tested, only CN caused the turn-on fluorescence of the imidazo[1,2-a]pyridinic-Cu2+ complex produced. Competition experiments between metal cations and anions showed a complete lack of potential interference in the detection and quantification of Cu2+ and CN ions. It has also been demonstrated through in vivo experiments that this chemosensor is suitable for fluorescence detection of Cu2+ and CN ions in human urine and blood [32,170,171]. Following this initial example, several others on similar compounds have been reported and are documented in the review dedicated to chemosensors based on imidazo[1,2-a]pyridines [5].
Similarly, derivatives of imidazo[1,5-a]pyridines, known for their intense luminescence, have been widely reported as a basis for new chemosensors. Hou et al. reported a robust ratiometric fluorescent probe for glutathione detection based on imidazo[1,5-a]pyridine fluorophore [172]. R. Mayilmurugan et al. reported an interesting study regarding the application of imidazo[1,5-a]pyridine derivatives as sensors for cysteine [173]. Generally, cysteine overexpression is well known in a wide variety of different kinds of tumors. In their research, the authors developed and prepared novel Cu(II) complexes utilizing imidazo[1,5-a]pyridine derivatives as optical chemosensors, employing a “turn-on” luminescent mechanism for cysteine detection. The investigated probe demonstrated distinct and intense emission upon interaction with cysteine, enabling imaging of tumor cells through the in situ reduction of copper (from Cu(II) to Cu(I)).
Another intriguing application of chemosensors derived from imidazo[1,5-a]pyridines involves the detection of hydrogen polysulfides (H2Sn), offering the potential for biomedical applications [174]. In general, hydrogen polysulfides play a pivotal role in various critical biological functions and processes associated with hydrogen sulfide. Despite their significance, developing probes capable of rapidly, selectively, and sensitively detecting hydrogen polysulfides poses a considerable challenge. S. Wang et al. have addressed this challenge by designing, synthesizing, and effectively applying a novel probe based on the imidazo[1,5-a]pyridinic structure. This probe offers a wide Stokes shift, a low detection limit, and minimal cytotoxicity. Furthermore, this newly developed lysosome-targeted probe has proven successful in detecting endogenous hydrogen polysulfides in cellular imaging [62,175,176,177,178,179].
Moreover, many studies have been conducted regarding the application of chemosensors based on imidazo[1,5-a]pyridines for the quantification of hydrogen sulfide [62] and hydrogen polysulfides [175], sulfur dioxide [177,180], hypochlorous acid [181,182,183], and metal ions such as Cd2+ and Zn2+ [184], Hg2+ [47,185], and Cu2+ [170,186].
It is well known that hydrogen ions exert a crucial influence on various aspects of cellular function, including enzymatic and metabolic activity, proliferation, and tumor growth. Therefore, monitoring hydrogen ions through chemosensors holds particular significance. Compounds derived from imidazo[1,5-a]pyridines have shown promise as effective ratiometric pH sensors, capitalizing on dual emission for accurate detection [187,188]. Many detection strategies have been tested and employed to assess the exact value of intracellular pH in different cellular compartments and organelles. Probes based on imidazo[1,5-a]pyridines have many advantages, such as good selectivity, sensitivity and solubility, and sometimes real-time monitoring. Probes based on imidazo[1,5-a]pyridines have been previously reported as fluorescent probes for pH in living cells [59,168].
In addition to the internal charge transfer (ICT) process employed by a single emitter to achieve ratiometric variations, Förster resonance energy transfer (FRET) stands out as one of the most extensively investigated and utilized mechanisms for ratiometric luminescent probes. FRET operates through a non-radiative mechanism where an excited emitter transfers energy to a chromophore acceptor in its ground state. Emitters such as rhodamine, fluorescein, quinine, BODIPY, xanthene, and coumarin have been utilized in the design and synthesis of FRET systems. Typically, the emission spectrum of the donor must substantially overlap with the absorption spectrum of the acceptor, a condition that often poses serious constrains to the development of FRET-based coupled systems. To address this limitation, several research groups have recently developed and synthesized novel imidazopyridinic probes, showcasing their potential for practical FRET-based applications in biological systems [47,177,181,189].

2.4. Imidazopyridines as Luminescent Probes in Energy Conversion Technology

For potential applications in optoelectronic devices or down-shifting technologies, luminescent molecules require specific characteristics, notably a broad Stokes shift to prevent reabsorption and a high quantum yield. Generally, absorbed photons should be re-emitted rather than dissipated as heat. A broad Stokes shift minimizes reabsorption, while a high quantum yield ensures that almost every absorbed photon is re-emitted and effectively utilized.
Compact luminescent molecules with a broad Stokes shift are increasingly appealing due to their potential as eco-friendly and cost-effective down-shifting luminescent materials in lighting and photovoltaic applications. Derivatives of imidazopyridines, with their inherent properties, are especially well suited for employment in these technological fields. These compounds have been studied for a large variety of optical applications, including optoelectronic devices [56,57,147], non-linear optical (NLO) studies [14,190,191], and down shifting [192,193].
G. Albrecht et al. recently conducted innovative investigations into the electroluminescence of imidazopyridines [194,195]. They employed layered OLEDs utilizing this fluorogenic core as an emission system [39,40,53,54]. So far, only copper complexes have been successfully applied in light-emitting electrochemical cells. However, several silver, iridium, and zinc complexes have been reported for potential applications in light-emitting electrochemical cells, showing good optical behavior, as well as high quantum efficiency and blue emission [133,135,136,138]. Salassa et al. were the first to report on the electrochemical characterization of both free imidazopyridinic ligands and their corresponding complexes. Their investigations demonstrated that the imidazopyridinic core generally undergoes reversible oxidation at low potentials, a conclusion subsequently corroborated by other studies [155,156,158]. In this context, metal complexes based on imidazopyridine ligands have been studied both as photosensitizers and as redox couples for dye-sensitized solar cells (DSSCs) [196,197,198].
Metal-based complexes offer the opportunity to finely adjust the redox potential of resultant devices through the modification of both metal centers and ligands. Within this framework, the modifiability of imidazopyridine ligands, along with their corresponding electronic levels, presents an intriguing prospect for applications in DSSCs as redox mediators or dyes [199].

2.5. Imidazopyridines in Fluorescent and Confocal Microscopy

In recent years, numerous new luminescent probes have emerged for utilization in fluorescence microscopy and confocal microscopy. These investigations encompass a variety of aims, including selective penetration into distinct cellular compartments, visualization of regions with varying pH levels through pH-sensitive fluorescent compounds, and examination of tissues with diverse and specific fluorophores exhibiting distinct optical and chemical characteristics. The research in this domain is concentrated on developing luminescent molecules with intense emission, such as the renowned BODIPY and rhodamine. Conversely, owing to their inherent structural adaptability and remarkable optical attributes, imidazopyridinic derivatives emerge as promising contenders for these applications. The characteristic emission of the imidazopyridinic unit typically falls within the 430–520 nm range, accompanied by a broad Stokes shift extending up to 80 nm [14,59,63,200].
Imidazopyridinic derivatives represent an interesting alternative to achieve intense emission with fluorescence-based microscopy and induced selectivity by the possibility of introducing functional groups or bioconjugation at various points of the heterocycle. Furthermore, imidazopyridinic products exhibit good solubility, intense emission, strong pH dependence, high quantum yield, and remarkable biocompatibility. In recent years, imidazopyridinic emitters have been successfully tested for cell imaging using confocal microscopy. Additionally, imidazopyridinic fluorophores have been tested for multichannel or multicolor bioimaging. To this purpose, cells or tissues are usually treated with different dyes and excited at various wavelengths; consequently, it is necessary to merge various fluorescent images collected under each suitable excitation wavelength for each fluorophore. In this context, imidazopyridinic derivatives have demonstrated the ability to achieve multicolor bioimaging using a single excitation wavelength. This methodology allows for the visualization and detection of reaction kinetics and biomolecular interactions concurrently. As a result, imidazopyridine emitters have been effectively employed in cell imaging utilizing confocal microscopy [59,168,172,174,179].
The comprehensive findings highlight that imidazopyridinic compounds possess sufficient permeability across plant plasma membranes and cell walls, exhibit high adsorption, and distribute effectively within plant internal tissues. Notably, they maintain robust fluorescence in aqueous environments, a crucial attribute for fluorophores designed for in vivo probing. Furthermore, imidazopyridinic compounds have demonstrated effectiveness in distinctly delineating the cytoplasm of mouse fibroblast cells in vitro, suggesting their potential suitability as counterstains to enhance contrast in multicolor fluorescence microscopy [168,187,188,201,202].

3. Imidazopyridines: Different Synthetic Approaches

While sharing the generic name of imidazopyridines, each nucleus described above is obtained through different synthetic approaches, ensuring the formation of these heterocycles, which are distinguished by the varying position of nitrogen atoms on the two aromatic rings of imidazole and pyridine. To achieve these structural modifications, it is of primary importance to be aware of the various synthetic approaches available to introduce functional groups, spacers, or additional conjugated systems.
Several reviews have recently been published regarding the synthesis of imidazo[1,5-a]pyridines [6,12,46] and imidazo[1,2-a]pyridines [43,82,83], the two most studied imidazopyridinic nuclei; relatively few reviews have been published on other imidazopyridine derivatives, but various synthetic approaches have been reported to obtain differently substituted products. Imidazo[1,2-a]pyridines are primarily obtained through cyclocondensation reactions, oxidative cyclization, rearrangement reactions, oxidative coupling and multicomponent reactions (see Figure 6) [45].
In the last ten years, significant developments have been made in the synthesis of imidazo[1,2-a]pyridines [7,43,45,73,82,203]; however, most of these methods still use a single base reagent, namely 2-aminopyridine, which is employed as a coupling partner in most cases.
Coupling reactions involve various other reagents such as ketones, aldehydes, substituted alkenes, and alkynes (see Figure 6 and Figure 7). Tandem reactions utilize 2-aminopyridine and nitroalkenes as reactants, while multicomponent reactions employ 2-aminopyridine with aldehydes and nitroalkanes or isonitriles or alkynes. Also of interest is the direct reaction between alcohols and 2-aminopyridine, which involves amination followed by cyclization and isomerization [43,45]. Oxidative coupling–cyclization reactions involve the use of copper, silver, or palladium-based catalysts, an oxidizing agent (often atmospheric oxygen), during the reaction of 2-aminopyridine, and other reagents bearing nitrogen-containing functional groups such as nitriles, nitroalkenes, and others [204,205].
Regarding imidazo[1,5-a]pyridines, the earliest references involve Vilsmeier-type cyclizations [206]. Subsequently, numerous derivatives of imidazo[1,5-a]pyridines (with one or two electron-attracting substituents at positions 1, 2, or 3), were obtained from substituted 2-aminomethylpyridines and phosgene (or ethyl chloroformate) by G. Palazzo and G. Picconi [207].
In 1986, A.P. Krapcho and J.R. Powell employed the same synthetic method as an efficient synthetic approach to collect 1,3-disubstituted imidazo[1,5-a]pyridines by treating ketimines derived from commercial 2,2′-dipyridylketone or benzophenone [9]. Interestingly, these ketones have been previously employed to obtain many other ligands with structures different from imidazopyridines [157,208,209,210,211].
More recently, imidazo[1,5-a]pyridines are mainly synthesized through condensation reactions (see Figure 8 and Figure 9). Over the years, various catalytic and non-catalytic methods have been developed.
The main condensations involve:
  • Substituted pyridyl-2-ketones with aromatic aldehydes and ammonium acetate (see Figure 9) [103,126,193,212,213,214];
  • Substituted picolylamines (2-(Aminomethyl)pyridin) with carboxylic acids in strongly acidic and dehydrating conditions [9];
  • Other couplings involve cyano-pyridines with aromatic aldehydes, picolylamines with thioesters, and cyclizations of 2-picolylamides [2,46,215,216].
Other synthesis mechanisms involve a cycloaddition approach using benzyl isonitriles as reagents or oxidative cycloadditions using pyridyl-2-ketones and amines (including natural amino acids) or 2-pyridine aldehydes and amines catalyzed by copper oxide and oxygen as oxidant [42,217,218,219].
Several alternative synthesis mechanisms include oxidative cyclization, rearrangement, photochemical reactions, and electrosynthesis. Presently, more than twenty different synthetic strategies have been delineated to achieve this cyclization process. Each unique synthetic approach offers distinct advantages and complementary features for synthesizing imidazo[1,5-a]pyridine derivatives.
In recent years, continuous efforts have been dedicated to this fascinating heterocyclization, since the pioneering work of Vilsmeier-type cyclization [206]. However, the substrates employed appear to be limited to specific ketones, nitriles, amides, aldehydes, and substituted N-heteroaryl pyridotriazoles. Additionally, for these transformations, transition metal-based catalysts and additional oxidants were generally required, inevitably leading to the generation of toxic waste and undesired reactions.
Many of the developed synthetic approaches encountered certain limitations, often associated with the use of various catalysts such as Se(IV) [38,220,221], Pd(II) [222,223], Fe(II) and Fe(III) [224,225], Cu(II) ion [226,227,228,229], CuCl2 [230,231,232], copper/iodine [233], Cu-MOF structures [234,235], boron trifluoride etherate [236], CBr4 [237] or MnO2 [53]. In addition, some catalysts require further reagents or co-catalysts such as selenium dioxide [238], dehydrating agents (e.g., molecular sieves [239] or propane phosphoric acid anhydride [213]), Lewis acids (BF3), or oxidant agents (e.g., oxygen, iodine [240,241], elemental sulfur [242], or tert-butylperoxybenzoate) [243].
Other important examples of innovative synthetic approaches for particular imidazopyridines have been recently reported, focusing on the following:

4. Summary and Outlook

Over the past forty years, the family of imidazopyridines has significantly expanded its membership, including extended heterocycles such as imidazo[1,2-a]quinolines and imidazo[1,5-a]quinolones and corresponding isoquinoline. The application of imidazopyridine products ranges from sensing to pharmaceuticals and from materials science to solar energy conversion, optoelectronics, and imaging. Each group of imidazopyridines seems to have taken on a specific role in different application fields; imidazo[1,2-a]pyridines have certainly sparked considerable interest in pharmaceuticals, with several commercial products based on this particular heterocycle. Countless studies demonstrate the activity of imidazo[1,2-a]pyridines as cardiotonic, antifungal, anti-inflammatory, antitumor, antiviral, antibacterial, antiprotozoal, antipyretic, analgesic, antiapoptotic, and hypnoselective agents.
Conversely, imidazo[1,5-a]pyridines owe their interest to their intense and tunable fluorescence emission. The ease with which these heterocycles can be modified and functionalized ensures the ability to modulate absorption and emission wavelength, quantum yield, and lifetime, all crucial parameters for applications in fluorescence microscopy, confocal microscopy, optoelectronics, and solar energy conversion technologies.
Sensor technology can be considered the latest application field of this interesting family; imidazo[1,2-a]pyridines and imidazo[1,5-a]pyridines have been studied to develop molecular sensors based on on–off fluorescence emission. These fluorophores can act as sensors for pH or metallic ions (such as copper, mercury, or cadmium) or small molecules or ions (sulfides, hypochlorite, cysteine) depending on the functions introduced on these particular aromatic heterocycles.
All imidazopyridines can act as ligands for metallic ions, either as monodentate ligands or, following appropriate functionalizations, as chelating or polydentate ligands. Metallic complexes based on imidazopyridines have been synthesized with all first- and second-row transition metals. Their use ranges from catalysis to electrochemiluminescence; some complexes have been used in OLED technology, confocal microscopy, and solar energy conversion technology. Imidazopyridines certainly represent an excellent basis for the development of fluorescent probes for imaging. Their ease of functionalization, low synthetic costs, along with good biocompatibility, stability, and solubility, make them excellent fluorophores for use in fluorescence microscopy, confocal microscopy, sensing, and quantitative method development.
The future prospects of this versatile compounds’ family certainly involve further exploration of less studied imidazopyridines such as imidazo[4,5-c]pyridines, imidazo[4,5-b]pyridines, imidazo[5,1-a]quinolines, and imidazo[2,1-a]isoquinolines, all recently synthesized heterocycles with fewer studies regarding their properties and interactions with other compounds.
Regarding synthesis, methods have recently been developed that exclude the use of expensive or toxic/rare metal-based catalysts. Synthetic strategies have been proposed in water and solvent-free conditions, starting from economical and readily available compounds such as natural amino acids, organic acids, and even aldehydes and ketones of natural origin (such as cuminaldehyde, vanillin, and papaverine). Commercial utilization of these products must involve low-cost, low-impact, highly sustainable synthesis, and recent publications seem to support this possibility.
Imidazopyridines thus describe an extensive family of heterocyclic compounds with very diverse properties, applicable in various technological fields due to their unique biological and photophysical properties. They represent an incredibly interesting group of promising compounds whose study and development could bring significant benefits in many application fields from pharmaceuticals to sensing and from imaging to light energy conversion.
A multidisciplinary approach is necessary to appreciate the versatility of this broad family, which is not found in nature and obtained through various synthesis approaches, from condensation to photocatalytic or microwave-mediated cyclization, and many other methods. Different skills are needed to correctly design the synthesis of imidazopyridine probes, which can be used as sensors, drugs, biological modulators, metallic ion ligands, fluorophores, and many other possible and future application fields. The versatility of these particular molecules is certainly guaranteed by their ease of functionalization, conjugation, and structural modification; however, it is the generic imidazopyridine core that ensures the versatile properties that make this family unique and extremely interesting.

Author Contributions

Conceptualization, G.V.; methodology, G.V. and R.R.; software, G.V.; investigation, G.V.; resources, R.R. and E.L.; writing—original draft preparation, G.V.; writing—review and editing, R.R. and E.L.; visualization, G.V.; supervision, G.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Project CH4.0 under the MUR program “Dipartimenti di Eccellenza 2023–2027” (CUP: D13C22003520001).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available within the article.

Acknowledgments

G. Volpi, R. Rabezzana and E. Laurenti acknowledge support from Project CH4.0 under the MUR program “Dipartimenti di Eccellenza 2023–2027” (CUP: D13C22003520001).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sanapalli, B.K.R.; Ashames, A.; Sigalapalli, D.K.; Shaik, A.B.; Bhandare, R.R.; Yele, V. Synthetic Imidazopyridine-Based Derivatives as Potential Inhibitors against Multi-Drug Resistant Bacterial Infections: A Review. Antibiotics 2022, 11, 1680. [Google Scholar] [CrossRef] [PubMed]
  2. Liu, Z.; Song, R.; Zhang, D.; Wu, R.; Liu, T.; Wu, Z.; Zhang, J.; Hu, D. Synthesis, Insecticidal Activity, and Mode of Action of Novel Imidazopyridine Mesoionic Derivatives Containing an Amido Group. Pest Manag. Sci. 2022, 78, 4983–4993. [Google Scholar] [CrossRef] [PubMed]
  3. Ghosh, D.; Ghosh, S.; Hajra, A. Electrochemical Functionalization of Imidazopyridine and Indazole: An Overview. Adv. Synth. Catal. 2021, 363, 5047–5071. [Google Scholar] [CrossRef]
  4. Camats, M.; Favier, I.; Mallet-Ladeira, S.; Pla, D.; Gómez, M. Understanding Cu(II)-Based Systems for C(Sp3)–H Bond Functionalization: Insights into the Synthesis of Aza-Heterocycles. Org. Biomol. Chem. 2022, 20, 219–227. [Google Scholar] [CrossRef] [PubMed]
  5. Chaudhran, P.A.; Sharma, A. Progress in the Development of Imidazopyridine-Based Fluorescent Probes for Diverse Applications. Crit. Rev. Anal. Chem. 2022, 1–18. [Google Scholar] [CrossRef] [PubMed]
  6. Volpi, G.; Rabezzana, R. Imidazo[1,5-a]Pyridine Derivatives: Useful, Luminescent and Versatile Scaffolds for Different Applications. New J. Chem. 2021, 45, 5737–5743. [Google Scholar] [CrossRef]
  7. Pericherla, K.; Kaswan, P.; Pandey, K.; Kumar, A. Recent Developments in the Synthesis of Imidazo[1,2-a]Pyridines. Synthesis 2015, 47, 887–912. [Google Scholar] [CrossRef]
  8. Hu, J.; Li, Y.; Wu, Y.; Liu, W.; Wang, Y.; Li, Y. Syntheses and Optical Properties of BODIPY Derivatives Based on Imidazo[1,5-a]Pyridine. Chem. Lett. 2015, 44, 645–647. [Google Scholar] [CrossRef]
  9. Krapcho, A.P.; Powell, J.R. Syntheses of 1,3-Disubstituted Imidazo[1,5-a]Pyridines. Tetrahedron Lett. 1986, 27, 3713–3714. [Google Scholar] [CrossRef]
  10. Murai, T.; Nagaya, E.; Shibahara, F.; Maruyama, T.; Nakazawa, H. Rhodium(I) and Iridium(I) Imidazo[1,5-a]Pyridine-1-Ylalkylalkoxy Complexes: Synthesis, Characterization and Application as Catalysts for Hydrosilylation of Alkynes. J. Organomet. Chem. 2015, 794, 76–80. [Google Scholar] [CrossRef]
  11. Volpi, G.; Garino, C.; Fresta, E.; Casamassa, E.; Giordano, M.; Barolo, C.; Viscardi, G. Strategies to Increase the Quantum Yield: Luminescent Methoxylated Imidazo[1,5-a]Pyridines. Dye. Pigment. 2021, 192, 109455. [Google Scholar] [CrossRef]
  12. Volpi, G. Luminescent Imidazo[1,5-a]Pyridine Scaffold: Synthetic Heterocyclization Strategies-Overview and Promising Applications. Asian J. Org. Chem. 2022, 11, e202200171. [Google Scholar] [CrossRef]
  13. Colombo, G.; Attilio Ardizzoia, G.; Brenna, S. Imidazo[1,5-a]Pyridine-Based Derivatives as Highly Fluorescent Dyes. Inorganica Chim. Acta 2022, 535, 120849. [Google Scholar] [CrossRef]
  14. Mohbiya, D.R.; Sekar, N. Tuning ‘Stokes Shift’ and ICT Character by Varying the Donor Group in Imidazo[1,5 a]Pyridines: A Combined Optical, DFT, TD-DFT and NLO Approach. ChemistrySelect 2018, 3, 1635–1644. [Google Scholar] [CrossRef]
  15. Volpi, G.; Garino, C.; Priola, E.; Magistris, C.; Chierotti, M.R.; Barolo, C. Halogenated Imidazo[1,5-a]Pyridines: Chemical Structure and Optical Properties of a Promising Luminescent Scaffold. Dye. Pigment. 2019, 171, 107713. [Google Scholar] [CrossRef]
  16. Vanda, D.; Zajdel, P.; Soural, M. Imidazopyridine-Based Selective and Multifunctional Ligands of Biological Targets Associated with Psychiatric and Neurodegenerative Diseases. Eur. J. Med. Chem. 2019, 181, 111569. [Google Scholar] [CrossRef] [PubMed]
  17. Esfandiari, R.; Moghimi-Rad, P.; Bule, M.H.; Souri, E.; Nadri, H.; Mahdavi, M.; Ghobadian, R.; Amini, M. New Imidazo[1,2-a]Pyridin-2-Yl Derivatives as AChE, BChE, and LOX Inhibitors; Design, Synthesis, and Biological Evaluation. Lett. Drug Des. Discov. 2023, 20, 1784–1798. [Google Scholar] [CrossRef]
  18. Samanta, S.; Kumar, S.; Aratikatla, K.E.; Ghorpade, S.R.; Singh, V. Recent Developments of Imidazo[1,2-a]Pyridine Analogues as Antituberculosis Agents. RSC Med. Chem. 2023, 14, 644–657. [Google Scholar] [CrossRef] [PubMed]
  19. Yamaguchi, E.; Shibahara, F.; Murai, T. 1-Alkynyl- and 1-Alkenyl-3-Arylimidazo[1,5-a]Pyridines: Synthesis, Photophysical Properties, and Observation of a Linear Correlation between the Fluorescent Wavelength and Hammett Substituent Constants. J. Org. Chem. 2011, 76, 6146–6158. [Google Scholar] [CrossRef]
  20. Murai, T.; Nagaya, E.; Miyahara, K.; Shibahara, F.; Maruyama, T. Synthesis and Characterization of Boron Complexes of Imidazo[1,5-a]Pyridylalkyl Alcohols. Chem. Lett. 2013, 42, 828–830. [Google Scholar] [CrossRef]
  21. Shibahara, F.; Kitagawa, A.; Yamaguchi, E.; Murai, T. Synthesis of 2-Azaindolizines by Using an Iodine-Mediated Oxidative Desulfurization Promoted Cyclization of N-2-Pyridylmethyl Thioamides and an Investigation of Their Photophysical Properties. Org. Lett. 2006, 8, 5621–5624. [Google Scholar] [CrossRef] [PubMed]
  22. Shibahara, F.; Mizuno, T.; Shibata, Y.; Murai, T. Transfer Semihydrogenation of Alkynes Catalyzed by Imidazo[1,5-a]Pyrid-3-Ylidene–Pd Complexes: Positive Effects of Electronic and Steric Features on N-Heterocyclic Carbene Ligands. Bull. Chem. Soc. Jpn. 2020, 93, 332–337. [Google Scholar] [CrossRef]
  23. Pavlinac, I.B.; Zlatić, K.; Persoons, L.; Daelemans, D.; Banjanac, M.; Radovanović, V.; Butković, K.; Kralj, M.; Hranjec, M. Biological Activity of Amidino-Substituted Imidazo [4,5-b]Pyridines. Molecules 2023, 28, 34. [Google Scholar] [CrossRef] [PubMed]
  24. Boček, I.; Hranjec, M.; Vianello, R. Imidazo[4,5-b]Pyridine Derived Iminocoumarins as Potential pH Probes: Synthesis, Spectroscopic and Computational Studies of Their Protonation Equilibria. J. Mol. Liq. 2022, 355, 118982. [Google Scholar] [CrossRef]
  25. Boček, I.; Hok, L.; Persoons, L.; Daelemans, D.; Vianello, R.; Hranjec, M. Imidazo[4,5-b]Pyridine Derived Tubulin Polymerization Inhibitors: Design, Synthesis, Biological Activity in Vitro and Computational Analysis. Bioorg. Chem. 2022, 127, 106032. [Google Scholar] [CrossRef] [PubMed]
  26. Zhang, B.; Suo, Q.; Li, Q.; Hu, J.; Zhu, Y.; Gao, Y.; Wang, Y. Multiresponsive Chemosensors Based on Ferrocenylimidazo[4,5-b]Pyridines: Solvent-Dependent Selective Dual Sensing of Hg2+ and Pb2+. Tetrahedron 2022, 120, 132878. [Google Scholar] [CrossRef]
  27. Zhang, B.; Suo, Q.; Li, Q.; Zhu, Y.; Gao, X.; Lv, L.; Gao, Y.; Jia, H.; Wang, Y. New Sulfur-Containing Ferrocenylimidazo[4,5-b]Pyridines: Multiresponsive Hg2+ Ion Sensing and Structure-Sensing Correlation. ChemistrySelect 2022, 7, e202103565. [Google Scholar] [CrossRef]
  28. Boček, I.; Starčević, K.; Novak Jovanović, I.; Vianello, R.; Hranjec, M. Novel Imidazo[4,5-b]Pyridine Derived Acrylonitriles: A Combined Experimental and Computational Study of Their Antioxidative Potential. J. Mol. Liq. 2021, 342, 117527. [Google Scholar] [CrossRef]
  29. Jarmoni, K.; Misbahi, K.; Ferrières, V. Imidazo[4,5-b]Pyridines: From Kinase Inhibitors to More Diversified Biological Properties. Curr. Med. Chem. 2024, 31, 515–528. [Google Scholar] [CrossRef]
  30. Krause, M.; Foks, H.; Gobis, K. Pharmacological Potential and Synthetic Approaches of Imidazo[4,5-b]Pyridine and Imidazo[4,5-c]Pyridine Derivatives. Molecules 2017, 22, 399. [Google Scholar] [CrossRef]
  31. Rejinthala, S.; Endoori, S.; Thumma, V.; Mondal, T. New Imidazo[4,5-c]Pyridine-Piperidine Hybrids as Potential Anti-Cancer Agents and In-Silico Studies. ChemistrySelect 2024, 9, e202303299. [Google Scholar] [CrossRef]
  32. Mahata, S.; Dey, S.; Mandal, B.B.; Manivannan, V. 3-(2-Hydroxyphenyl)Imidazo[5, 1-a]Isoquinoline as Cu(II) Sensor, Its Cu(II) Complex for Selective Detection of CN− Ion and Biological Compatibility. J. Photochem. Photobiol. A Chem. 2022, 427, 113795. [Google Scholar] [CrossRef]
  33. Khalili, G. A Diastereoselective Synthesis of (Z)-3-[(Aryl)(Hydroxyimino)Methyl]-2-Cyclohexyl-1-(Cyclohexylamino)Imidazo[5,1-a]Isoquinolinium Chlorides from Isoquinoline, Chlorooximes, and Cyclohexyl Isocyanide. Monatshefte Chem. 2016, 147, 429–433. [Google Scholar] [CrossRef]
  34. Jafarpour, F.; Ashtiani, P.T. A Novel One-Step Synthesis of Imidazo[5,1-a]Isoquinolines via a Tandem Pd-Catalyzed Alkylation−Direct Arylation Sequence. J. Org. Chem. 2009, 74, 1364–1366. [Google Scholar] [CrossRef] [PubMed]
  35. Chen, X.-H.; Cui, H.-L. Asymmetric Synthesis of Imidazo[2,1-a]Isoquinolin-3-Ones with Dihydroisoquinolines and N-Substituted Amino Acids. Asian J. Org. Chem. 2022, 11, e202100761. [Google Scholar] [CrossRef]
  36. Meena, N.; Dhiman, S.; Rangan, K.; Kumar, A. Cobalt-Catalyzed Tandem One-Pot Synthesis of Polysubstituted Imidazo[1,5-a]Pyridines and Imidazo[1,5-a]Isoquinolines. Org. Biomol. Chem. 2022, 20, 4215–4223. [Google Scholar] [CrossRef]
  37. Cappelli, A.; Anzini, M.; Castriconi, F.; Grisci, G.; Paolino, M.; Braile, C.; Valenti, S.; Giuliani, G.; Vomero, S.; Di Capua, A.; et al. Design, Synthesis, and Biological Evaluation of Imidazo[1,5-a]Quinoline as Highly Potent Ligands of Central Benzodiazepine Receptors. J. Med. Chem. 2016, 59, 3353–3372. [Google Scholar] [CrossRef] [PubMed]
  38. Zhang, J.; Wang, H.; Chen, Y.; Xie, H.; Ding, C.; Tan, J.; Xu, K. Electrochemical Synthesis of Selenocyanated Imidazo[1,5-a]Quinolines under Metal Catalyst- and Chemical Oxidant-Free Conditions. Chin. Chem. Lett. 2020, 31, 1576–1579. [Google Scholar] [CrossRef]
  39. Albrecht, G.; Geis, C.; Herr, J.M.; Ruhl, J.; Göttlich, R.; Schlettwein, D. Electroluminescence and Contact Formation of 1-(Pyridin-2-Yl)-3-(Quinolin-2-Yl)Imidazo[1,5-a]Quinoline Thin Films. Org. Electron. 2019, 65, 321–326. [Google Scholar] [CrossRef]
  40. Albrecht, G.; Rössiger, C.; Herr, J.M.; Locke, H.; Yanagi, H.; Göttlich, R.; Schlettwein, D. Optimization of the Substitution Pattern of 1,3-Disubstituted Imidazo[1,5-a]Pyridines and -Quinolines for Electro-Optical Applications. Phys. Status Solidi B 2020, 257, 1900677. [Google Scholar] [CrossRef]
  41. Reddy, V.P.; Iwasaki, T.; Kambe, N. Synthesis of Imidazo and Benzimidazo[2,1-a]Isoquinolines by Rhodium-Catalyzed Intramolecular Double C–H Bond Activation. Org. Biomol. Chem. 2013, 11, 2249–2253. [Google Scholar] [CrossRef] [PubMed]
  42. Giordano, M.; Volpi, G.; Garino, C.; Cardano, F.; Barolo, C.; Viscardi, G.; Fin, A. New Fluorescent Derivatives from Papaverine: Two Mechanisms to Increase the Quantum Yield. Dye. Pigment. 2023, 218, 111482. [Google Scholar] [CrossRef]
  43. Panda, J.; Raiguru, B.P.; Mishra, M.; Mohapatra, S.; Nayak, S. Recent Advances in the Synthesis of Imidazo[1,2-a]Pyridines: A Brief Review. ChemistrySelect 2022, 7, e202103987. [Google Scholar] [CrossRef]
  44. Yan, Z.; Wan, C.; Yang, Y.; Zha, Z.; Wang, Z. The Synthesis of Imidazo[1,5-a]Quinolines via a Decarboxylative Cyclization under Metal-Free Conditions. RSC Adv. 2018, 8, 23058–23065. [Google Scholar] [CrossRef]
  45. Kumar Bagdi, A.; Santra, S.; Monir, K.; Hajra, A. Synthesis of Imidazo[1,2-a]Pyridines: A Decade Update. Chem. Commun. 2015, 51, 1555–1575. [Google Scholar] [CrossRef]
  46. Ramana Reddy, M.; Darapaneni, C.M.; Patil, R.D.; Kumari, H. Recent Synthetic Methodologies for Imidazo[1,5-a]Pyridines and Related Heterocycles. Org. Biomol. Chem. 2022, 20, 3440–3468. [Google Scholar] [CrossRef]
  47. Ge, Y.; Xing, X.; Liu, A.; Ji, R.; Shen, S.; Cao, X. A Novel Imidazo[1,5-a]Pyridine-Rhodamine FRET System as an Efficient Ratiometric Fluorescent Probe for Hg2+ in Living Cells. Dye. Pigment. 2017, 146, 136–142. [Google Scholar] [CrossRef]
  48. Zhang, S.; Yuan, W.; Qin, Y.; Zhang, J.; Lu, N.; Liu, W.; Li, H.; Wang, Y.; Li, Y. Bidentate BODIPY-Appended 2-Pyridylimidazo[1,2-a]Pyridine Ligand and Fabrication of Luminescent Transition Metal Complexes. Polyhedron 2018, 148, 22–31. [Google Scholar] [CrossRef]
  49. Wu, W.H.; Zhao, J.Z.; Guo, H.M.; Sun, J.F.; Ji, S.M.; Wang, Z.L. Long-Lived Room-Temperature Near-IR Phosphorescence of BODIPY in a Visible-Light-Harvesting N C N PtII-Acetylide Complex with a Directly Metalated BODIPY Chromophore. Chem.-Eur. J. 2012, 18, 1961–1968. [Google Scholar] [CrossRef]
  50. Anupriya; Thomas, K.R.J.; Nagar, M.R.; Shahnawaz; Jou, J.-H. Imidazo[1,2-a]Pyridine Based Deep-Blue Emitter: Effect of Donor on the Optoelectronic Properties. J. Mater. Sci. Mater. Electron. 2021, 32, 26838–26850. [Google Scholar] [CrossRef]
  51. Zheng, X.-H.; Zhao, J.-W.; Chen, X.; Cai, R.; Yang, G.-X.; Zhu, J.-J.; Tang, S.-S.; Lin, Z.-H.; Tao, S.-L.; Tong, Q.-X. Imidazo[1,2-a]Pyridine as an Electron Acceptor to Construct High-Performance Deep-Blue Organic Light-Emitting Diodes with Negligible Efficiency Roll-Off. Chem.—Eur. J. 2020, 26, 8588–8596. [Google Scholar] [CrossRef]
  52. Girase, J.D.; Kajjam, A.B.; Dubey, D.K.; Kesavan, K.K.; Jou, J.-H.; Vaidyanathan, S. Unipolar 1-Phenylimidazo[1,5-a]Pyridine: A New Class of Ultra-Bright Sky-Blue Emitters for Solution-Processed Organic Light Emitting Diodes. New J. Chem. 2022, 46, 16717–16729. [Google Scholar] [CrossRef]
  53. Herr, J.M.; Rössiger, C.; Albrecht, G.; Yanagi, H.; Göttlich, R. Solvent-Free Microwave-Assisted Synthesis of Imidazo[1,5-a]Pyridine and –Quinoline Derivatives. Synth. Commun. 2019, 49, 2931–2940. [Google Scholar] [CrossRef]
  54. Albrecht, G.; Herr, J.M.; Steinbach, M.; Yanagi, H.; Göttlich, R.; Schlettwein, D. Synthesis, Optical Characterization and Thin Film Preparation of 1-(Pyridin-2-Yl)-3-(Quinolin-2-Yl)Imidazo[1,5-a]Quinoline. Dye. Pigment. 2018, 158, 334–341. [Google Scholar] [CrossRef]
  55. Weber, M.D.; Garino, C.; Volpi, G.; Casamassa, E.; Milanesio, M.; Barolo, C.; Costa, R.D. Origin of a Counterintuitive Yellow Light-Emitting Electrochemical Cell Based on a Blue-Emitting Heteroleptic Copper(I) Complex. Dalton Trans. 2016, 45, 8984–8993. [Google Scholar] [CrossRef]
  56. Fresta, E.; Volpi, G.; Garino, C.; Barolo, C.; Costa, R.D. Contextualizing Yellow Light-Emitting Electrochemical Cells Based on a Blue-Emitting Imidazo-Pyridine Emitter. Polyhedron 2018, 140, 129–137. [Google Scholar] [CrossRef]
  57. Fresta, E.; Volpi, G.; Milanesio, M.; Garino, C.; Barolo, C.; Costa, R.D. Novel Ligand and Device Designs for Stable Light-Emitting Electrochemical Cells Based on Heteroleptic Copper(I) Complexes. Inorg. Chem. 2018, 57, 10469–10479. [Google Scholar] [CrossRef]
  58. Yagishita, F.; Nii, C.; Tezuka, Y.; Tabata, A.; Nagamune, H.; Uemura, N.; Yoshida, Y.; Mino, T.; Sakamoto, M.; Kawamura, Y. Fluorescent N-Heteroarenes Having Large Stokes Shift and Water Solubility Suitable for Bioimaging. Asian J. Org. Chem. 2018, 7, 1614–1619. [Google Scholar] [CrossRef]
  59. Wang, L.; Han, X.; Qu, G.; Su, L.; Zhao, B.; Miao, J. A pH Probe Inhibits Senescence in Mesenchymal Stem Cells. Stem Cell Res. Ther. 2018, 9, 343. [Google Scholar] [CrossRef]
  60. Fukaminato, T. Single-Molecule Fluorescence Photoswitching: Design and Synthesis of Photoswitchable Fluorescent Molecules. J. Photochem. Photobiol. C-Photochem. Rev. 2011, 12, 177–208. [Google Scholar] [CrossRef]
  61. Sedgwick, A.C.; Wu, L.; Han, H.-H.; Bull, S.D.; He, X.-P.; James, T.D.; Sessler, J.L.; Tang, B.Z.; Tian, H.; Yoon, J. Excited-State Intramolecular Proton-Transfer (ESIPT) Based Fluorescence Sensors and Imaging Agents. Chem. Soc. Rev. 2018, 47, 8842–8880. [Google Scholar] [CrossRef]
  62. Ren, Y.; Zhang, L.; Zhou, Z.; Wang, S.; Xu, Y.; Gu, Y.; Zha, X. A New Fluorescent Probe for Quick and Highly Selective Detection of Hydrogen Sulfide and Its Application in Living Cells. New J. Chem. 2018, 42, 13884–13888. [Google Scholar] [CrossRef]
  63. Volpi, G.; Lace, B.; Garino, C.; Priola, E.; Artuso, E.; Cerreia Vioglio, P.; Barolo, C.; Fin, A.; Genre, A.; Prandi, C. New Substituted Imidazo[1,5-a]Pyridine and Imidazo[5,1-a]Isoquinoline Derivatives and Their Application in Fluorescence Cell Imaging. Dye. Pigment. 2018, 157, 298–304. [Google Scholar] [CrossRef]
  64. Kumari, N.; Singh, S.; Baral, M.; Kanungo, B.K. Schiff Bases: A Versatile Fluorescence Probe in Sensing Cations. J. Fluoresc. 2023, 33, 859–893. [Google Scholar] [CrossRef]
  65. Nirogi, R.; Mohammed, A.R.; Shinde, A.K.; Bogaraju, N.; Gagginapalli, S.R.; Ravella, S.R.; Kota, L.; Bhyrapuneni, G.; Muddana, N.R.; Benade, V.; et al. Synthesis and SAR of Imidazo[1,5-a]Pyridine Derivatives as 5-HT4 Receptor Partial Agonists for the Treatment of Cognitive Disorders Associated with Alzheimer’s Disease. Eur. J. Med. Chem. 2015, 103, 289–301. [Google Scholar] [CrossRef]
  66. Nicholson, A.N.; Pascoe, P.A. Hypnotic Activity of an Imidazo-Pyridine (Zolpidem). Br. J. Clin. Pharmacol. 1986, 21, 205–211. [Google Scholar] [CrossRef]
  67. Thakare, P.P.; Dakhane, S.; Shikh, A.N.; Modak, M.; Patil, A.; Bobade, V.D.; Mhaske, P.C. Design, Synthesis, Antimicrobial and Ergosterol Inhibition Activity of New 4-(Imidazo[1,2-a]Pyridin-2-Yl)Quinoline Derivatives. Polycycl. Aromat. Compd. 2022, 42, 5282–5299. [Google Scholar] [CrossRef]
  68. Zhou, S.; Chen, G.; Huang, G. Design, Synthesis and Biological Evaluation of Imidazo[1,2-a]Pyridine Analogues or Derivatives as Anti-helmintic Drug. Chem. Biol. Drug Des. 2019, 93, 503–510. [Google Scholar] [CrossRef]
  69. Roby, O.; Kadiri, F.Z.; Loukhmi, Z.; Moutaouakil, M.; Tighadouini, S.; Saddik, R.; Aboulmouhajir, A. Synthesis of new set of imidazo[1,2-a]pyridine-schiff bases derivatives as potential antimicrobial agents: Experimental and theoretical approaches. J. Mol. Struct. 2023, 1292, 136186. [Google Scholar] [CrossRef]
  70. Chakravarty, H.; Ojha, D.; Konreddy, A.K.; Bal, C.; Chandra, N.S.; Sharon, A.; Chattopadhyay, D. Synthesis of Multi Ring-Fused Imidazo [1,2-a]Isoquinoline-Based Fluorescent Scaffold as Anti-Herpetic Agent. Antivir. Chem. Chemother. 2015, 24, 127–135. [Google Scholar] [CrossRef]
  71. Mannem, G.R.; Navudu, R.; Dubasi, N.; Mohammed, M.A.; Bollikolla, H.B. Design, and Synthesis of Aryl Derivatives of Imidazopyridine-Thiadiazoles as Possible Anticancer Agents. ChemistrySelect 2022, 7, e202200455. [Google Scholar] [CrossRef]
  72. Ge, Y.Q.; Li, F.R.; Zhang, Y.J.; Bi, Y.S.; Cao, X.Q.; Duan, G.Y.; Wang, J.W.; Liu, Z.L. Synthesis, Crystal Structure, Optical Properties and Antibacterial Evaluation of Novel Imidazo [1, 5-a] Pyridine Derivatives Bearing a Hydrazone Moiety. Luminescence 2014, 29, 293–300. [Google Scholar] [CrossRef]
  73. Guo, L.; Ding, Y.; Wang, H.; Liu, Y.; Qiang, Q.; Luo, Q.; Song, F.; Li, C. Imidazo[1,2-a]Pyridine Derivatives Synthesis from Lignin β-O-4 Segments via a One-Pot Multicomponent Reaction. iScience 2023, 26, 106834. [Google Scholar] [CrossRef]
  74. Chitrakar, R.; Rawat, D.; Sistla, R.; Vadithe, L.N.; Subbarayappa, A. Design, Synthesis and Anticancer Activity of Sulfenylated Imidazo-Fused Heterocycles. Bioorg. Med. Chem. Lett. 2021, 49, 128307. [Google Scholar] [CrossRef]
  75. Fauber, B.P.; Gobbi, A.; Robarge, K.; Zhou, A.; Barnard, A.; Cao, J.; Deng, Y.; Eidenschenk, C.; Everett, C.; Ganguli, A.; et al. Discovery of Imidazo[1,5-a]Pyridines and -Pyrimidines as Potent and Selective RORc Inverse Agonists. Bioorg. Med. Chem. Lett. 2015, 25, 2907–2912. [Google Scholar] [CrossRef]
  76. Roy, M.; Chakravarthi, B.V.S.K.; Jayabaskaran, C.; Karande, A.A.; Chakravarty, A.R. Impact of Metal Binding on the Antitumor Activity and Cellular Imaging of a Metal Chelator Cationic Imidazopyridine Derivative. Dalton Trans. 2011, 40, 4855–4864. [Google Scholar] [CrossRef]
  77. Spallarossa, A.; Rapetti, F.; Signorello, M.G.; Rosano, C.; Iervasi, E.; Ponassi, M.; Brullo, C. New Insights on Pharmacological Activity of Imidazo-Pyrazole Scaffold. ChemMedChem 2023, 18, e202300252. [Google Scholar] [CrossRef]
  78. Lončar, B.; Perin, N.; Mioč, M.; Boček, I.; Grgić, L.; Kralj, M.; Tomić, S.; Stojković, M.R.; Hranjec, M. Novel Amino Substituted Tetracyclic Imidazo[4,5-b]Pyridine Derivatives: Design, Synthesis, Antiproliferative Activity and DNA/RNA Binding Study. Eur. J. Med. Chem. 2021, 217, 113342. [Google Scholar] [CrossRef]
  79. Hranjec, M.; Lučić, B.; Ratkaj, I.; Pavelić, S.K.; Piantanida, I.; Pavelić, K.; Karminski-Zamola, G. Novel Imidazo[4,5-b]Pyridine and Triaza-Benzo[c]Fluorene Derivatives: Synthesis, Antiproliferative Activity and DNA Binding Studies. Eur. J. Med. Chem. 2011, 46, 2748–2758. [Google Scholar] [CrossRef]
  80. Taha, M.; Ismail, N.H.; Imran, S.; Rashwan, H.; Jamil, W.; Ali, S.; Kashif, S.M.; Rahim, F.; Salar, U.; Khan, K.M. Synthesis of 6-Chloro-2-Aryl-1H-Imidazo[4,5-b]Pyridine Derivatives: Antidiabetic, Antioxidant, β-Glucuronidase Inhibiton and Their Molecular Docking Studies. Bioorg. Chem. 2016, 65, 48–56. [Google Scholar] [CrossRef]
  81. Sedic, M.; Poznic, M.; Gehrig, P.; Scott, M.; Schlapbach, R.; Hranjec, M.; Karminski-Zamola, G.; Pavelic, K.; Pavelic, S.K. Differential Antiproliferative Mechanisms of Novel Derivative of Benzimidazo[1,2- α ]Quinoline in Colon Cancer Cells Depending on Their P53 Status. Mol. Cancer Ther. 2008, 7, 2121–2132. [Google Scholar] [CrossRef] [PubMed]
  82. Kurteva, V. Recent Progress in Metal-Free Direct Synthesis of Imidazo[1,2-a]Pyridines. ACS Omega 2021, 6, 35173–35185. [Google Scholar] [CrossRef] [PubMed]
  83. Tali, J.A.; Kumar, G.; Sharma, B.K.; Rasool, Y.; Sharma, Y.; Shankar, R. Synthesis and Site Selective C–H Functionalization of Imidazo-[1,2-a]Pyridines. Org. Biomol. Chem. 2023, 21, 7267–7289. [Google Scholar] [CrossRef] [PubMed]
  84. Deep, A.; Kaur Bhatia, R.; Kaur, R.; Kumar, S.; Kumar Jain, U.; Singh, H.; Batra, S.; Kaushik, D.; Kishore Deb, P. Imidazo[1,2-a]Pyridine Scaffold as Prospective Therapeutic Agents. Curr. Top. Med. Chem. 2017, 17, 238–250. [Google Scholar] [CrossRef] [PubMed]
  85. Enguehard-Gueiffier, C.; Gueiffier, A. Recent Progress in the Pharmacology of Imidazo[1,2-a]Pyridines. Mini Rev. Med. Chem. 2007, 7, 888–899. [Google Scholar] [CrossRef]
  86. Kobayashi, Y.; Kumadaki, I.; Kobayashi, E. 1,3-Dipolar Cycloaddition Reaction of Trifluoroacetonitrile with Heterocyclic Ylides. Heterocycles 1981, 15, 1223. [Google Scholar] [CrossRef]
  87. Trotter, B.W.; Nanda, K.K.; Burgey, C.S.; Potteiger, C.M.; Deng, J.Z.; Green, A.I.; Hartnett, J.C.; Kett, N.R.; Wu, Z.; Henze, D.A.; et al. Imidazopyridine CB2 Agonists: Optimization of CB2/CB1 Selectivity and Implications for in Vivo Analgesic Efficacy. Bioorg. Med. Chem. Lett. 2011, 21, 2354–2358. [Google Scholar] [CrossRef]
  88. Hatano, K.; Sekimata, K.; Yamada, T.; Abe, J.; Ito, K.; Ogawa, M.; Magata, Y.; Toyohara, J.; Ishiwata, K.; Biggio, G.; et al. Radiosynthesis and in Vivo Evaluation of Two Imidazopyridineacetamides, [11C]CB184 and [11C]CB190, as a PET Tracer for 18 kDa Translocator Protein: Direct Comparison with [11C](R)-PK11195. Ann. Nucl. Med. 2015, 29, 325–335. [Google Scholar] [CrossRef]
  89. Chaudhari, V.; Bagwe-Parab, S.; Buttar, H.S.; Gupta, S.; Vora, A.; Kaur, G. Challenges and Opportunities of Metal Chelation Therapy in Trace Metals Overload-Induced Alzheimer’s Disease. Neurotox. Res. 2023, 41, 270–287. [Google Scholar] [CrossRef]
  90. El-Sayed, W.M.; Hussin, W.A.; Al-Faiyz, Y.S.; Ismail, M.A. The Position of Imidazopyridine and Metabolic Activation Are Pivotal Factors in the Antimutagenic Activity of Novel Imidazo[1,2-a]Pyridine Derivatives. Eur. J. Pharmacol. 2013, 715, 212–218. [Google Scholar] [CrossRef]
  91. Rival, Y.; Grassy, G.; Michel, G. Synthesis and Antibacterial Activity of Some Imidazo(1,2-a)Pyrimidine Derivatives. Chem. Pharm. Bull. 1992, 40, 1170–1176. [Google Scholar] [CrossRef] [PubMed]
  92. Ingersoll, M.A.; Lyons, A.S.; Muniyan, S.; D’Cunha, N.; Robinson, T.; Hoelting, K.; Dwyer, J.G.; Bu, X.R.; Batra, S.K.; Lin, M.-F. Novel Imidazopyridine Derivatives Possess Anti-Tumor Effect on Human Castration-Resistant Prostate Cancer Cells. PLoS ONE 2015, 10, e0131811. [Google Scholar] [CrossRef]
  93. Ahmadi, N.; Khoramjouy, M.; Azami Movahed, M.; Amidi, S.; Faizi, M.; Zarghi, A. Design, Synthesis, In Vitro and In Vivo Evaluation of New Imidazo[1,2-a]Pyridine Derivatives as Cyclooxygenase-2 Inhibitors. Anti-Cancer Agents Med. Chem. (Former. Curr. Med. Chem.-Anti-Cancer Agents) 2024, 24, 504–513. [Google Scholar] [CrossRef]
  94. Goel, R.; Luxami, V.; Paul, K. Imidazo[1,2-a]Pyridines: Promising Drug Candidate for Antitumor Therapy. Curr. Top. Med. Chem. 2016, 16, 3590–3616. [Google Scholar] [CrossRef] [PubMed]
  95. Chen, C.-J.; Bando, K.; Ashino, H.; Taguchi, K.; Shiraishi, H.; Fujimoto, O.; Kitamura, C.; Matsushima, S.; Fujinaga, M.; Zhang, M.-R.; et al. Synthesis and Biological Evaluation of Novel Radioiodinated Imidazopyridine Derivatives for Amyloid-β Imaging in Alzheimer’s Disease. Bioorg. Med. Chem. 2014, 22, 4189–4197. [Google Scholar] [CrossRef] [PubMed]
  96. Pan, M.; Solozobova, V.; Kuznik, N.C.; Jung, N.; Gräßle, S.; Gourain, V.; Heneka, Y.M.; Cramer von Clausbruch, C.A.; Fuhr, O.; Munuganti, R.S.N.; et al. Identification of an Imidazopyridine-Based Compound as an Oral Selective Estrogen Receptor Degrader for Breast Cancer Therapy. Cancer Res. Commun. 2023, 3, 1378–1396. [Google Scholar] [CrossRef]
  97. Sucu, B.O. Biological Evaluation of Imidazopyridine Derivatives as Potential Anticancer Agents against Breast Cancer Cells. Med. Chem. Res. 2022, 31, 2231–2242. [Google Scholar] [CrossRef]
  98. Wang, J.; Wu, H.; Song, G.; Yang, D.; Huang, J.; Yao, X.; Qin, H.; Chen, Z.; Xu, Z.; Xu, C. A Novel Imidazopyridine Derivative Exerts Anticancer Activity by Inducing Mitochondrial Pathway-Mediated Apoptosis. BioMed Res. Int. 2020, 2020, e4929053. [Google Scholar] [CrossRef]
  99. Peytam, F.; Emamgholipour, Z.; Mousavi, A.; Moradi, M.; Foroumadi, R.; Firoozpour, L.; Divsalar, F.; Safavi, M.; Foroumadi, A. Imidazopyridine-Based Kinase Inhibitors as Potential Anticancer Agents: A Review. Bioorg. Chem. 2023, 140, 106831. [Google Scholar] [CrossRef]
  100. Soumyashree, D.K.; Reddy, D.S.; Nagarajaiah, H.; Naik, L.; Savanur, H.M.; Shilpa, C.D.; Sunitha Kumari, M.; Shanavaz, H.; Padmashali, B. Imidazopyridine Chalcones as Potent Anticancer Agents: Synthesis, Single-Crystal X-ray, Docking, DFT and SAR Studies. Arch. Pharm. 2023, 356, 2300106. [Google Scholar] [CrossRef]
  101. Zeng, K.; Ye, J.; Meng, X.; Dechert, S.; Simon, M.; Gong, S.; Mata, R.A.; Zhang, K. Anomeric Stereoauxiliary Cleavage of the C-N Bond of D-glucosamine for the Preparation of Imidazo[1,5-α]Pyridines. Chem.—A Eur. J. 2022, 28, e202200648. [Google Scholar] [CrossRef] [PubMed]
  102. Davey, D.; Erhardt, P.W.; Lumma, W.C.; Wiggins, J.; Sullivan, M.; Pang, D.; Cantor, E. Cardiotonic Agents. 1. Novel 8-Aryl-Substituted Imidazo[1,2-a] and [1,5-a]Pyridines and Imidazo[1,5-a]Pyridinones as Potential Positive Inotropic Agents. J. Med. Chem. 1987, 30, 1337–1342. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, J.; Mason, R.; VanDerveer, D.; Feng, K.; Bu, X.R. Convenient Preparation of a Novel Class of Imidazo[1,5-a]Pyridines:  Decisive Role by Ammonium Acetate in Chemoselectivity. J. Org. Chem. 2003, 68, 5415–5418. [Google Scholar] [CrossRef] [PubMed]
  104. Ford, N.F.; Browne, L.J.; Campbell, T.; Gemenden, C.; Goldstein, R.; Gude, C.; Wasley, J.W.F. Imidazo[1,5-a]Pyridines—A New Class of Thromboxane-A2 Synthetase Inhibitors. J. Med. Chem. 1985, 28, 164–170. [Google Scholar] [CrossRef]
  105. Renno, G.; Cardano, F.; Volpi, G.; Barolo, C.; Viscardi, G.; Fin, A. Imidazo[1,5-a]Pyridine-Based Fluorescent Probes: A Photophysical Investigation in Liposome Models. Molecules 2022, 27, 3856. [Google Scholar] [CrossRef] [PubMed]
  106. Yagishita, F.; Tanigawa, J.; Nii, C.; Tabata, A.; Nagamune, H.; Takanari, H.; Imada, Y.; Kawamura, Y. Fluorescent Imidazo[1,5-a]Pyridinium Salt for a Potential Cancer Therapy Agent. ACS Med. Chem. Lett. 2019, 10, 1110–1114. [Google Scholar] [CrossRef] [PubMed]
  107. Fuse, S.; Ohuchi, T.; Asawa, Y.; Sato, S.; Nakamura, H. Development of 1-Aryl-3-Furanyl/Thienyl-Imidazopyridine Templates for Inhibitors against Hypoxia Inducible Factor (HIF)-1 Transcriptional Activity. Bioorg. Med. Chem. Lett. 2016, 26, 5887–5890. [Google Scholar] [CrossRef] [PubMed]
  108. Browne, L.J.; Gude, C.; Rodriguez, H.; Steele, R.E.; Bhatnager, A. Fadrozole Hydrochloride—A Potent, Selective, Nonsteroidal Inhibitor of Aromatase for the Treatment of Estrogen-Dependent Disease. J. Med. Chem. 1991, 34, 725–736. [Google Scholar] [CrossRef] [PubMed]
  109. Perin, N.; Martin-Kleiner, I.; Nhili, R.; Laine, W.; David-Cordonnier, M.-H.; Vugrek, O.; Karminski-Zamola, G.; Kralj, M.; Hranjec, M. Biological Activity and DNA Binding Studies of 2-Substituted Benzimidazo[1,2-a]Quinolines Bearing Different Amino Side Chains. Med. Chem. Commun. 2013, 4, 1537. [Google Scholar] [CrossRef]
  110. Hranjec, M.; Kralj, M.; Piantanida, I.; Sedić, M.; Šuman, L.; Pavelić, K.; Karminski-Zamola, G. Novel Cyano- and Amidino-Substituted Derivatives of Styryl-2-Benzimidazoles and Benzimidazo[1,2-a]Quinolines. Synthesis, Photochemical Synthesis, DNA Binding, and Antitumor Evaluation, Part 3. J. Med. Chem. 2007, 50, 5696–5711. [Google Scholar] [CrossRef]
  111. Pässler, U.; Knölker, H.-J. The Pyrrolo[2,1-a]Isoquinoline Alkaloids. In The Alkaloids: Chemistry and Biology; Elsevier: Amsterdam, The Netherlands, 2011; Volume 70, pp. 79–151. ISBN 978-0-12-391426-2. [Google Scholar]
  112. Newhouse, B.J.; Wenglowsky, S.; Grina, J.; Laird, E.R.; Voegtli, W.C.; Ren, L.; Ahrendt, K.; Buckmelter, A.; Gloor, S.L.; Klopfenstein, N.; et al. Imidazo[4,5-b]Pyridine Inhibitors of B-Raf Kinase. Bioorg. Med. Chem. Lett. 2013, 23, 5896–5899. [Google Scholar] [CrossRef] [PubMed]
  113. Krajčovičová, S.; Jorda, R.; Vanda, D.; Soural, M.; Kryštof, V. 1,4,6-Trisubstituted Imidazo[4,5-c]Pyridines as Inhibitors of Bruton’s Tyrosine Kinase. Eur. J. Med. Chem. 2021, 211, 113094. [Google Scholar] [CrossRef] [PubMed]
  114. Puerstinger, G.; Paeshuyse, J.; De Clercq, E.; Neyts, J. Antiviral 2,5-Disubstituted Imidazo[4,5-c]Pyridines: From Anti-Pestivirus to Anti-Hepatitis C Virus Activity. Bioorg. Med. Chem. Lett. 2007, 17, 390–393. [Google Scholar] [CrossRef] [PubMed]
  115. Jose, G.; Suresha Kumara, T.H.; Nagendrappa, G.; Sowmya, H.B.V.; Jasinski, J.P.; Millikan, S.P.; Chandrika, N.; More, S.S.; Harish, B.G. New Polyfunctional Imidazo[4,5-C]Pyridine Motifs: Synthesis, Crystal Studies, Docking Studies and Antimicrobial Evaluation. Eur. J. Med. Chem. 2014, 77, 288–297. [Google Scholar] [CrossRef] [PubMed]
  116. Garrido, A.; Vera, G.; Delaye, P.-O.; Enguehard-Gueiffier, C. Imidazo[1,2-b]Pyridazine as Privileged Scaffold in Medicinal Chemistry: An Extensive Review. Eur. J. Med. Chem. 2021, 226, 113867. [Google Scholar] [CrossRef] [PubMed]
  117. Henderson, S.H.; Sorrell, F.J.; Bennett, J.M.; Fedorov, O.; Hanley, M.T.; Godoi, P.H.; Ruela de Sousa, R.; Robinson, S.; Navratilova, I.H.; Elkins, J.M.; et al. Imidazo[1,2-b]Pyridazines as Inhibitors of DYRK Kinases. Eur. J. Med. Chem. 2024, 269, 116292. [Google Scholar] [CrossRef] [PubMed]
  118. Akwata, D.; Kempen, A.L.; Lamptey, J.; Dayal, N.; Brauer, N.R.; Sintim, H.O. Discovery of Imidazo[1,2-b]Pyridazine-Containing TAK1 Kinase Inhibitors with Excellent Activities against Multiple Myeloma. RSC Med. Chem. 2024, 15, 178–192. [Google Scholar] [CrossRef] [PubMed]
  119. Xiao, X.; Xu, Y.; Yu, X.; Chen, Y.; Zhao, W.; Xie, Z.; Zhu, X.; Xu, H.; Yang, Y.; Zhang, P. Discovery of Imidazo[1,2-b]Pyridazine Macrocyclic Derivatives as Novel ALK Inhibitors Capable of Combating Multiple Resistant Mutants. Bioorg. Med. Chem. Lett. 2023, 89, 129309. [Google Scholar] [CrossRef] [PubMed]
  120. Yagishita, F.; Kozai, N.; Nii, C.; Tezuka, Y.; Uemura, N.; Yoshida, Y.; Mino, T.; Sakamoto, M.; Kawamura, Y. Synthesis of Dimeric Imidazo[1, 5-a]Pyridines and Their Photophysical Properties. ChemistrySelect 2017, 2, 10694–10698. [Google Scholar] [CrossRef]
  121. Roseblade, S.J.; Ros, A.; Monge, D.; Alcarazo, M.; Alvarez, E.; Lassaletta, J.M.; Fernandez, R. Imidazo[1,5-a]Pyridin-3-Ylidene/Thioether Mixed C/S Ligands and Complexes Thereof. Organometallics 2007, 26, 2570–2578. [Google Scholar] [CrossRef]
  122. Yagishita, F.; Hoshi, K.; Mukai, S.; Kinouchi, T.; Katayama, T.; Yoshida, Y.; Minagawa, K.; Furube, A.; Imada, Y. Effect of Phenolic Substituent Position in Boron Complexes of Imidazo[1,5-a]Pyridine. Asian J. Org. Chem. 2022, 11, e202200040. [Google Scholar] [CrossRef]
  123. Volpi, G.; Garino, C.; Conterosito, E.; Barolo, C.; Gobetto, R.; Viscardi, G. Facile Synthesis of Novel Blue Light and Large Stoke Shift Emitting Tetradentate Polyazines Based on Imidazo[1,5-a]Pyridine. Dye. Pigment. 2016, 128, 96–100. [Google Scholar] [CrossRef]
  124. Volpi, G.; Garino, C.; Priola, E.; Diana, E.; Gobetto, R.; Buscaino, R.; Viscardi, G.; Barolo, C. Facile Synthesis of Novel Blue Light and Large Stoke Shift Emitting Tetradentate Polyazines Based on Imidazo[1,5-a]Pyridine—Part 2. Dye. Pigment. 2017, 143, 284–290. [Google Scholar] [CrossRef]
  125. Cerrato, V.; Volpi, G.; Priola, E.; Giordana, A.; Garino, C.; Rabezzana, R.; Diana, E. Mono-, Bis-, and Tris-Chelate Zn(II) Complexes with Imidazo[1,5-a]Pyridine: Luminescence and Structural Dependence. Molecules 2023, 28, 3703. [Google Scholar] [CrossRef] [PubMed]
  126. Wang, J.; Dyers, L.; Mason, R.; Amoyaw, P.; Bu, X.R. Highly Efficient and Direct Heterocyclization of Dipyridyl Ketone to N,N-Bidentate Ligands. J. Org. Chem. 2005, 70, 2353–2356. [Google Scholar] [CrossRef] [PubMed]
  127. Herr, J.M.; Rössiger, C.; Locke, H.; Wilhelm, M.; Becker, J.; Heimbrodt, W.; Schlettwein, D.; Göttlich, R. Synthesis, Optical and Theoretical Characterization of Heteroleptic Iridium(III) Imidazo[1,5-a]Pyridine and -Quinoline Complexes. Dye. Pigment. 2020, 180, 108512. [Google Scholar] [CrossRef]
  128. Wei, Q.; Wei, M.-J.; Ou, Y.-J.; Zhang, J.-Y.; Huang, X.; Cai, Y.-P.; Si, L.-P. Formation and Conversion of Six Temperature-Dependent Fluorescent ZnII-Complexes Containing Two in Situ Formed N-Rich Heterocyclic Ligands. RSC Adv. 2017, 7, 6994–7002. [Google Scholar] [CrossRef]
  129. Priola, E.; Volpi, G.; Rabezzana, R.; Borfecchia, E.; Garino, C.; Benzi, P.; Martini, A.; Operti, L.; Diana, E. Bridging Solution and Solid-State Chemistry of Dicyanoaurate: The Case Study of Zn–Au Nucleation Units. Inorg. Chem. 2020, 59, 203–213. [Google Scholar] [CrossRef] [PubMed]
  130. Volpi, G.; Zago, S.; Rabezzana, R.; Diana, E.; Priola, E.; Garino, C.; Gobetto, R. N-Based Polydentate Ligands and Corresponding Zn(II) Complexes: A Structural and Spectroscopic Study. Inorganics 2023, 11, 435. [Google Scholar] [CrossRef]
  131. Priola, E.; Conterosito, E.; Giordana, A.; Volpi, G.; Garino, C.; Andreo, L.; Diana, E.; Barolo, C.; Milanesio, M. Polymorphism and Solid State Peculiarities in Imidazo[1,5-a]Pyridine Core Deriving Compounds: An Analysis of Energetic and Structural Driving Forces. J. Mol. Struct. 2022, 1253, 132175. [Google Scholar] [CrossRef]
  132. Álvarez, C.M.; Álvarez-Miguel, L.; García-Rodríguez, R.; Martín-Álvarez, J.M.; Miguel, D. 3-(Pyridin-2-Yl)Imidazo[1,5-a]Pyridine (Pyridylindolizine) as Ligand in Complexes of Transition and Main-Group Metals: N,N-Chelating Ligands. Eur. J. Inorg. Chem. 2015, 2015, 4921–4934. [Google Scholar] [CrossRef]
  133. Ardizzoia, G.A.; Brenna, S.; Durini, S.; Therrien, B. Synthesis and Characterization of Luminescent Zinc(II) Complexes with a N,N-Bidentate 1-Pyridylimidazo[1,5-a]Pyridine Ligand. Polyhedron 2015, 90, 214–220. [Google Scholar] [CrossRef]
  134. Ardizzoia, G.A.; Ghiotti, D.; Therrien, B.; Brenna, S. Homoleptic Complexes of Divalent Metals Bearing N,O-Bidentate Imidazo[1,5-a]Pyridine Ligands: Synthesis, X-Ray Characterization and Catalytic Activity in the Heck Reaction. Inorganica Chim. Acta 2018, 471, 384–390. [Google Scholar] [CrossRef]
  135. Durini, S.; Ardizzoia, G.A.; Therrien, B.; Brenna, S. Tuning the Fluorescence Emission in Mononuclear Heteroleptic Trigonal Silver(I) Complexes. New J. Chem. 2017, 41, 3006–3014. [Google Scholar] [CrossRef]
  136. Ardizzoia, G.A.; Brenna, S.; Durini, S.; Therrien, B.; Veronelli, M. Synthesis, Structure, and Photophysical Properties of Blue-Emitting Zinc(II) Complexes with 3-Aryl-Substituted 1-Pyridylimidazo[1,5-a]Pyridine Ligands: Blue-Emitting Zinc(II) Complexes. Eur. J. Inorg. Chem. 2014, 2014, 4310–4319. [Google Scholar] [CrossRef]
  137. Colombo, G.; Romeo, A.; Ardizzoia, G.A.; Furrer, J.; Therrien, B.; Brenna, S. Boron Difluoride Functionalized (Tetrahydroimidazo[1,5-a]Pyridin-3-Yl)Phenols: Highly Fluorescent Blue Emissive Materials. Dye. Pigment. 2020, 182, 108636. [Google Scholar] [CrossRef]
  138. Ardizzoia, G.A.; Colombo, G.; Therrien, B.; Brenna, S. Tuning the Fluorescence Emission and HOMO-LUMO Band Gap in Homoleptic Zinc(II) Complexes with N,O-Bidentate (Imidazo[1,5-a]Pyrid-3-Yl)Phenols. Eur. J. Inorg. Chem. 2019, 2019, 1825–1831. [Google Scholar] [CrossRef]
  139. Colombo, G.; Attilio Ardizzoia, G.; Furrer, J.; Therrien, B.; Brenna, S. Driving the Emission Towards Blue by Controlling the HOMO-LUMO Energy Gap in BF2-Functionalized 2-(Imidazo[1,5-a]Pyridin-3-yl)Phenols. Chem. Eur. J. 2021, 27, 12380–12387. [Google Scholar] [CrossRef]
  140. Evans, R.C.; Douglas, P.; Winscom, C.J. Coordination Complexes Exhibiting Room-Temperature Phosphorescence: Evaluation of Their Suitability as Triplet Emitters in Organic Light Emitting Diodes. Coord. Chem. Rev. 2006, 250, 2093–2126. [Google Scholar] [CrossRef]
  141. Volpi, G.; Priola, E.; Garino, C.; Daolio, A.; Rabezzana, R.; Benzi, P.; Giordana, A.; Diana, E.; Gobetto, R. Blue Fluorescent Zinc(II) Complexes Based on Tunable Imidazo[1,5-a]Pyridines. Inorganica Chim. Acta 2020, 509, 119662. [Google Scholar] [CrossRef]
  142. Kundu, N.; Abtab, S.M.T.; Kundu, S.; Endo, A.; Teat, S.J.; Chaudhury, M. Triple-Stranded Helicates of Zinc(II) and Cadmium(II) Involving a New Redox-Active Multiring Nitrogenous Heterocyclic Ligand: Synthesis, Structure, and Electrochemical and Photophysical Properties. Inorg. Chem. 2012, 51, 2652–2661. [Google Scholar] [CrossRef] [PubMed]
  143. Sozzi, M.; Chierotti, M.R.; Gobetto, R.; Gomila, R.M.; Marzaroli, V.; Priola, E.; Volpi, G.; Zago, S.; Frontera, A.; Garino, C. One-Dimensional and Two-Dimensional Zn(II) Coordination Polymers with Ditopic Imidazo[1,5-a]Pyridine: A Structural and Computational Study. Molecules 2024, 29, 653. [Google Scholar] [CrossRef] [PubMed]
  144. Zamora, R.; Hidalgo, F.J. 2-Amino-1-Methyl-6-Phenylimidazo[4,5-b]Pyridine (PhIP) Formation and Fate: An Example of the Coordinate Contribution of Lipid Oxidation and Maillard Reaction to the Production and Elimination of Processing-Related Food Toxicants. RSC Adv. 2015, 5, 9709–9721. [Google Scholar] [CrossRef]
  145. Kim, J.; Hahm, H.; Ryu, J.Y.; Byun, S.; Park, D.-A.; Lee, S.H.; Lim, H.; Lee, J.; Hong, S. Pyridine-Chelated Imidazo[1,5-a]Pyridine N-Heterocyclic Carbene Nickel(II) Complexes for Acrylate Synthesis from Ethylene and CO2. Catalysts 2020, 10, 758. [Google Scholar] [CrossRef]
  146. Swamy, C.A.P.; Varenikov, A.; Ruiter, G. de Chiral Imidazo[1,5-a]Pyridine–Oxazolines: A Versatile Family of NHC Ligands for the Highly Enantioselective Hydrosilylation of Ketones. Organometallics 2020, 39, 247–257. [Google Scholar] [CrossRef]
  147. Fresta, E.; Costa, R.D. Beyond Traditional Light-Emitting Electrochemical Cells—A Review of New Device Designs and Emitters. J. Mater. Chem. C 2017, 5, 5643–5675. [Google Scholar] [CrossRef]
  148. Cavinato, L.M.; Volpi, G.; Fresta, E.; Garino, C.; Fin, A.; Barolo, C. Microwave-Assisted Synthesis, Optical and Theoretical Characterization of Novel 2-(Imidazo[1,5-a]Pyridine-1-Yl)Pyridinium Salts. Chemistry 2021, 3, 714–727. [Google Scholar] [CrossRef]
  149. Zhang, X.; He, C.; Yang, X.; Zhang, Q.; Li, Y.; Yao, J. Fe II, Co II and Ni II Complexes Based on 1-Chloro-3-(Pyridin-2-Yl)Imidazo[1,5-a]Pyridine: Synthesis, Structures, Single-Molecule Magnetic and Electrocatalytic Properties. New J. Chem. 2022, 46, 21780–21787. [Google Scholar] [CrossRef]
  150. Gao, Q.; Chen, Y.; Liu, Y.; Li, C.; Gao, D.; Wu, B.; Li, H.; Li, Y.; Liu, W.; Li, W. A Series of ZnII and CoII Complexes Based on 2-(Imidazo[1,5-a]Pyridin-3-Yl)Phenol: Syntheses, Structures, and Luminescent and Magnetic Properties. J. Coord. Chem. 2014, 67, 1673–1692. [Google Scholar] [CrossRef]
  151. Mukherjee, A.; Dhar, S.; Nethaji, M.; Chakravarty, R.A. Ternary Iron(II) Complex with an Emissive Imidazopyridine Arm from Schiff Base Cyclizations and Its Oxidative DNA Cleavage Activity. Dalton Trans. 2005, 2, 349–353. [Google Scholar] [CrossRef]
  152. Santra, S.; Bagdi, A.K.; Majee, A.; Hajra, A. Iron(III)-Catalyzed Cascade Reaction between Nitroolefins and 2-Aminopyridines: Synthesis of Imidazo[1,2-a]Pyridines and Easy Access towards Zolimidine. Adv. Synth. Catal. 2013, 355, 1065–1070. [Google Scholar] [CrossRef]
  153. Prakasham, A.P.; Gangwar, M.K.; Ghosh, P. β-Enaminone Synthesis from 1,3-Dicarbonyl Compounds and Aliphatic and Aromatic Amines Catalyzed by Iron Complexes of Fused Bicyclic Imidazo[1,5-a]Pyridine Derived N-Heterocyclic Carbenes. Eur. J. Inorg. Chem. 2019, 2019, 295–313. [Google Scholar] [CrossRef]
  154. Wu, J.-J.; Cao, M.-L.; Ye, B.-H. Spontaneous Chiral Resolution of Mer-[CoII(N,N,O-L3)2] Enantiomers Mediated by π–π Interactions. Chem. Commun. 2010, 46, 3687–3689. [Google Scholar] [CrossRef] [PubMed]
  155. Volpi, G.; Garino, C.; Salassa, L.; Fiedler, J.; Hardcastle, K.I.; Gobetto, R.; Nervi, C. Cationic Heteroleptic Cyclometalated Iridium Complexes with 1-Pyridylimidazo[1,5-Alpha]Pyridine Ligands: Exploitation of an Efficient Intersystem Crossing. Chem.-Eur. J. 2009, 15, 6415–6427. [Google Scholar] [CrossRef] [PubMed]
  156. Salassa, L.; Garino, C.; Albertino, A.; Volpi, G.; Nervi, C.; Gobetto, R.; Hardcastle, K.I. Computational and Spectroscopic Studies of New Rhenium(I) Complexes Containing Pyridylimidazo[1,5-a]Pyridine Ligands: Charge Transfer and Dual Emission by Fine-Tuning of Excited States. Organometallics 2008, 27, 1427–1435. [Google Scholar] [CrossRef]
  157. Volpi, G.; Garino, C.; Gobetto, R.; Nervi, C. Dipyridylmethane Ethers as Ligands for Luminescent Ir Complexes. Molecules 2021, 26, 7161. [Google Scholar] [CrossRef] [PubMed]
  158. Garino, C.; Ruiu, T.; Salassa, L.; Albertino, A.; Volpi, G.; Nervi, C.; Gobetto, R.; Hardcastle, K.I. Spectroscopic and Computational Study on New Blue Emitting ReL(CO)(3)Cl Complexes Containing Pyridylimidazo[1,5-a]Pyridine Ligands. Eur. J. Inorg. Chem. 2008, 23, 3587–3591. [Google Scholar] [CrossRef]
  159. Takizawa, S.; Nishida, J.; Tsuzuki, T.; Tokito, S.; Yamashita, Y. Phosphorescent Iridium Complexes Based on 2-Phenylimidazo[1,2-a]Pyridine Ligands: Tuning of Emission Color toward the Blue Region and Application to Polymer Light-Emitting Devices. Inorg. Chem. 2007, 46, 4308–4319. [Google Scholar] [CrossRef]
  160. Blanco-Rodríguez, A.M.; Kvapilova, H.; Sykora, J.; Towrie, M.; Nervi, C.; Volpi, G.; Zalis, S.; Vlcek, A. Photophysics of Singlet and Triplet Intraligand Excited States in [ReCl(CO)3(1-(2-Pyridyl)-Imidazo[1,5-α]Pyridine)] Complexes. J. Am. Chem. Soc. 2014, 136, 5963–5973. [Google Scholar] [CrossRef]
  161. Guckian, A.L.; Doering, M.; Ciesielski, M.; Walter, O.; Hjelm, J.; O’Boyle, N.M.; Henry, W.; Browne, W.R.; McGarvey, J.J.; Vos, J.G. Assessment of Intercomponent Interaction in Phenylene Bridged Dinuclear Ruthenium(II) and Osmium(II) Polypyridyl Complexes. Dalton Trans. 2004, 23, 3943–3949. [Google Scholar] [CrossRef]
  162. Sandroni, M.; Volpi, G.; Fiedler, J.; Buscaino, R.; Viscardi, G.; Milone, L.; Gobetto, R.; Nervi, C. Iridium and Ruthenium Complexes Covalently Bonded to Carbon Surfaces by Means of Electrochemical Oxidation of Aromatic Amines. Catal. Today 2010, 158, 22–28. [Google Scholar] [CrossRef]
  163. Chen, Y.; Li, L.; Chen, Z.; Liu, Y.; Hu, H.; Chen, W.; Liu, W.; Li, Y.; Lei, T.; Cao, Y.; et al. Metal-Mediated Controllable Creation of Secondary, Tertiary, and Quaternary Carbon Centers: A Powerful Strategy for the Synthesis of Iron, Cobalt, and Copper Complexes with in Situ Generated Substituted 1-Pyridineimidazo[1,5-a]Pyridine Ligands. Inorg. Chem. 2012, 51, 9705–9713. [Google Scholar] [CrossRef] [PubMed]
  164. Li, K.; Niu, J.-L.; Yang, M.-Z.; Li, Z.; Wu, L.-Y.; Hao, X.-Q.; Song, M.-P. New Type of 2,6-Bis(Imidazo[1,2-a]Pyridin-2-Yl)Pyridine-Based Ruthenium Complexes: Active Catalysts for Transfer Hydrogenation of Ketones. Organometallics 2015, 34, 1170–1176. [Google Scholar] [CrossRef]
  165. Giuso, V.; Jouaiti, E.; Cebrián, C.; Parant-Aury, S.; Kyritsakas, N.; Gourlaouen, C.; Mauro, M. Symmetry-Broken Charge-Transfer Excited State in Homoleptic Zinc(II) Imidazo[1,2-a]Pyridine Complexes. ChemPhotoChem 2023, 7, e202300092. [Google Scholar] [CrossRef]
  166. Divya, D.; Mala, R.; Nandhagopal, M.; Narayanasamy, M.; Thennarasu, S. Coordination of Distal Carboxylate Anion Alters Metal Ion Specific Binding in Imidazo[1,2-a]Pyridine Congeners. J. Fluoresc. 2023, 33, 1397–1412. [Google Scholar] [CrossRef] [PubMed]
  167. Yurdakul, Ş.; Badoğlu, S. FT-IR Spectra, Vibrational Assignments, and Density Functional Calculations of Imidazo[1,2-a]Pyridine Molecule and Its Zn(II) Halide Complexes. Struct. Chem. 2009, 20, 423–434. [Google Scholar] [CrossRef]
  168. Song, G.-J.; Bai, S.-Y.; Dai, X.; Cao, X.-Q.; Zhao, B.-X. A Ratiometric Lysosomal pH Probe Based on the Imidazo[1,5-a]Pyridine-Rhodamine FRET and ICT System. RSC Adv. 2016, 6, 41317–41322. [Google Scholar] [CrossRef]
  169. Alharbi, K.H. A Review on Organic Colorimetric and Fluorescent Chemosensors for the Detection of Zn(II) Ions. Crit. Rev. Anal. Chem. 2022, 53, 1472–1488. [Google Scholar] [CrossRef]
  170. Ge, Y.; Ji, R.; Shen, S.; Cao, X.; Li, F. A Ratiometric Fluorescent Probe for Sensing Cu2+ Based on New Imidazo[1,5-a]Pyridine Fluorescent Dye. Sens. Actuators B Chem. 2017, 245, 875–881. [Google Scholar] [CrossRef]
  171. Bej, S.; Das, R.; Mondal, A.; Saha, R.; Sarkar, K.; Banerjee, P. Knoevenagel Condensation Triggered Synthesis of Dual-Channel Oxene Based Chemosensor: Discriminative Spectrophotometric Recognition of F, CN and HSO4− with Breast Cancer Cell Imaging, Real Sample Analysis and Molecular Keypad Lock Applications. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2022, 273, 120989. [Google Scholar] [CrossRef]
  172. Hou, P.; Sun, J.; Wang, H.; Liu, L.; Zou, L.; Chen, S. TCF-Imidazo[1,5-α]Pyridine: A Potential Robust Ratiometric Fluorescent Probe for Glutathione Detection with High Selectivity. Sens. Actuators B Chem. 2020, 304, 127244. [Google Scholar] [CrossRef]
  173. Priyanga, S.; Khamrang, T.; Velusamy, M.; Karthi, S.; Ashokkumar, B.; Mayilmurugan, R. Coordination Geometry-Induced Optical Imaging of L-Cysteine in Cancer Cells Using Imidazopyridine-Based Copper(II) Complexes. Dalton Trans. 2019, 48, 1489–1503. [Google Scholar] [CrossRef]
  174. Chen, S.; Li, H.; Hou, P. A Novel Imidazo[1,5-α]Pyridine-Based Fluorescent Probe with a Large Stokes Shift for Imaging Hydrogen Sulfide. Sens. Actuators B Chem. 2018, 256, 1086–1092. [Google Scholar] [CrossRef]
  175. Ren, Y.; Zhang, L.; Zhou, Z.; Luo, Y.; Wang, S.; Yuan, S.; Gu, Y.; Xu, Y.; Zha, X. A New Lysosome-Targetable Fluorescent Probe with a Large Stokes Shift for Detection of Endogenous Hydrogen Polysulfides in Living Cells. Anal. Chim. Acta 2019, 1056, 117–124. [Google Scholar] [CrossRef] [PubMed]
  176. Yuan, Q.; Chen, L.-L.; Zhu, X.-H.; Yuan, Z.-H.; Duan, Y.-T.; Yang, Y.-S.; Wang, B.-Z.; Wang, X.-M.; Zhu, H.-L. An Imidazo[1,5-α]Pyridine-Derivated Fluorescence Sensor for Rapid and Selective Detection of Sulfite. Talanta 2020, 217, 121087. [Google Scholar] [CrossRef] [PubMed]
  177. Chen, F.; Liu, A.; Ji, R.; Xu, Z.; Dong, J.; Ge, Y. A FRET-Based Probe for Detection of the Endogenous SO2 in Cells. Dye. Pigment. 2019, 165, 212–216. [Google Scholar] [CrossRef]
  178. Strianese, M.; Brenna, S.; Ardizzoia, G.A.; Guarnieri, D.; Lamberti, M.; D’Auria, I.; Pellecchia, C. Imidazo-Pyridine-Based Zinc(II) Complexes as Fluorescent Hydrogen Sulfide Probes. Dalton Trans. 2021, 50, 17075–17085. [Google Scholar] [CrossRef]
  179. Chen, S.; Hou, P.; Sun, J.; Wang, H.; Liu, L. Imidazo[1,5-α]Pyridine-Based Fluorescent Probe with a Large Stokes Shift for Specific Recognition of Sulfite. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2020, 225, 117508. [Google Scholar] [CrossRef] [PubMed]
  180. Cui, R.; Liu, C.; Zhang, P.; Qin, K.; Ge, Y. An Imidazo[1,5-a]Pyridine Benzopyrylium-Based NIR Fluorescent Probe with Ultra-Large Stokes Shifts for Monitoring SO2. Molecules 2023, 28, 515. [Google Scholar] [CrossRef]
  181. Shen, S.-L.; Huang, X.-Q.; Zhang, Y.-Y.; Zhu, Y.; Hou, C.; Ge, Y.-Q.; Cao, X.-Q. Ratiometric Fluorescent Probe for the Detection of HOCl in Lysosomes Based on FRET Strategy. Sens. Actuators B Chem. 2018, 263, 252–257. [Google Scholar] [CrossRef]
  182. Song, G.-J.; Ma, H.-L.; Luo, J.; Cao, X.-Q.; Zhao, B.-X. A New Ratiometric Fluorescent Probe for Sensing HOC1 Based on TBET in Real Time. Dye. Pigment. 2018, 148, 206–211. [Google Scholar] [CrossRef]
  183. Shen, S.-L.; Zhang, X.-F.; Ge, Y.-Q.; Zhu, Y.; Cao, X.-Q. A Novel Ratiometric Fluorescent Probe for the Detection of HOCl Based on FRET Strategy. Sens. Actuator B Chem. 2018, 254, 736–741. [Google Scholar] [CrossRef]
  184. Ciupa, A.; Mahon, F.M.; Bank, P.A.D.; Caggiano, L. Simple Pyrazoline and Pyrazole “Turn on” Fluorescent Sensors Selective for Cd2+ and Zn2+ in MeCN. Org. Biomol. Chem. 2012, 10, 8753–8757. [Google Scholar] [CrossRef] [PubMed]
  185. Srivastava, S.; Thakur, N.; Singh, A.; Shukla, P.; Kumar Maikhuri, V.; Garg, N.; Prasad, A.; Pandey, R. Development of a Fused Imidazo[1,2-a]Pyridine Based Fluorescent Probe for Fe3+ and Hg2+ in Aqueous Media and HeLa Cells. RSC Adv. 2019, 9, 29856–29863. [Google Scholar] [CrossRef] [PubMed]
  186. Zhang, X.; Huang, Y.; Zhang, Q.; Li, D.; Li, Y. A One-Dimensional Cadmium Coordination Polymer: Synthesis, Structure, and Application as Luminescent Sensor for Cu2+ and CrO42−/Cr2O72− Ions. Eur. J. Inorg. Chem. 2021, 2021, 1349–1357. [Google Scholar] [CrossRef]
  187. Hutt, J.T.; Jo, J.; Olasz, A.; Chen, C.-H.; Lee, D.; Aron, Z.D. Fluorescence Switching of Imidazo[1,5-a]Pyridinium Ions: pH-Sensors with Dual Emission Pathways. Org. Lett. 2012, 14, 3162–3165. [Google Scholar] [CrossRef] [PubMed]
  188. Zhang, X.; Song, G.-J.; Cao, X.-J.; Liu, J.-T.; Chen, M.-Y.; Cao, X.-Q.; Zhao, B.-X. A New Fluorescent pH Probe for Acidic Conditions. RSC Adv. 2015, 5, 89827–89832. [Google Scholar] [CrossRef]
  189. Shen, S.-L.; Zhang, X.-F.; Ge, Y.-Q.; Zhu, Y.; Cao, X.-Q. A Mitochondria-Targeting Ratiometric Fluorescent Probe for the Detection of Hypochlorite Based on the FRET Strategy. RSC Adv. 2017, 7, 55296–55300. [Google Scholar] [CrossRef]
  190. Jadhav, S.D.; Alswaidan, I.A.; Rhyman, L.; Ramasami, P.; Sekar, N. Effect of Methoxy Group on NLOphoric Properties of Fluorescent 7-Arylstyryl-2-Methoxyphenylimidazo[1,2-a]Pyridine—Solvatochromic and Computational Method. J. Mol. Struct. 2018, 1173, 349–365. [Google Scholar] [CrossRef]
  191. Jadhav, S.D.; Ramasami, P.; Sekar, N. Substituent Effects on Linear and Nonlinear Optical Properties of Fluorescent (E)-2-(4-Halophenyl)-7-Arlstyrylimidazo[1,2-A] Pyridine: Spectroscopic and Computational Methods. Physical Sciences Reviews 2019, 4, 20180032. [Google Scholar] [CrossRef]
  192. Volpi, G.; Galliano, S.; Buscaino, R.; Viscardi, G.; Barolo, C. Fluorescent Trifluoromethylated Imidazo[1,5-a]Pyridines and Their Application in Luminescent down-Shifting Conversion. J. Lumin. 2022, 242, 118529. [Google Scholar] [CrossRef]
  193. Volpi, G.; Magnano, G.; Benesperi, I.; Saccone, D.; Priola, E.; Gianotti, V.; Milanesio, M.; Conterosito, E.; Barolo, C.; Viscardi, G. One Pot Synthesis of Low Cost Emitters with Large Stokes’ Shift. Dye. Pigment. 2017, 137, 152–164. [Google Scholar] [CrossRef]
  194. Rössiger, C.; Oel, T.; Schweitzer, P.; Vasylets, O.; Kirchner, M.; Abdullahu, A.; Schlettwein, D.; Göttlich, R. Synthesis and Optical and Theoretical Characterization of Imidazo[5,1-a]Isoquinolines and Imidazo[1,5-a]Quinolines. Eur. J. Org. Chem. 2024, e202400298. [Google Scholar] [CrossRef]
  195. Kulhanek, N.; Martin, N.; Göttlich, R. Highly Versatile Preparation of Imidazo[1,5-a]Quinolines and Characterization of Their Photoluminescent Properties. Eur. J. Org. Chem. 2024, 27, e202301007. [Google Scholar] [CrossRef]
  196. Nitha, P.R.; Soman, S.; John, J. Indole Fused Heterocycles as Sensitizers in Dye-Sensitized Solar Cells: An Overview. Mater. Adv. 2021, 2, 6136–6168. [Google Scholar] [CrossRef]
  197. Giordano, M.; Volpi, G.; Bonomo, M.; Mariani, P.; Garino, C.; Viscardi, G. Methoxy-Substituted Copper Complexes as Possible Redox Mediators in Dye-Sensitized Solar Cells. New J. Chem. 2021, 45, 15303–15311. [Google Scholar] [CrossRef]
  198. Prostota, Y.; Kachkovsky, O.D.; Reis, L.V.; Santos, P.F. New Unsymmetrical Squaraine Dyes Derived from Imidazo[1,5-a]Pyridine. Dye. Pigment. 2013, 96, 554–562. [Google Scholar] [CrossRef]
  199. Irfan, A.; Chaudhry, A.R.; Al-Sehemi, A.G.; Assiri, M.A.; Hussain, A. Charge Carrier and Optoelectronic Properties of Phenylimidazo[1,5-a]Pyridine-Containing Small Molecules at Molecular and Solid-State Bulk Scales. Comput. Mater. Sci. 2019, 170, 109179. [Google Scholar] [CrossRef]
  200. Colombo, G.; Cinco, A.; Ardizzoia, G.A.; Brenna, S. Long-Alkyl Chain Functionalized Imidazo[1,5-a]Pyridine Derivatives as Blue Emissive Dyes. Colorants 2023, 2, 179–193. [Google Scholar] [CrossRef]
  201. Zhu, S.; Lin, X.; Ran, P.; Xia, Q.; Yang, C.; Ma, J.; Fu, Y. A Novel Luminescence-Functionalized Metal-Organic Framework Nanoflowers Electrochemiluminesence Sensor via “on-off” System. Biosens. Bioelectron. 2017, 91, 436–440. [Google Scholar] [CrossRef]
  202. Wu, X.; Wang, R.; Kwon, N.; Ma, H.; Yoon, J. Activatable Fluorescent Probes for in Situ Imaging of Enzymes. Chem. Soc. Rev. 2022, 51, 450–463. [Google Scholar] [CrossRef] [PubMed]
  203. Mohana Roopan, S.; Patil, S.M.; Palaniraja, J. Recent Synthetic Scenario on Imidazo[1,2-a]Pyridines Chemical Intermediate. Res. Chem. Intermed. 2016, 42, 2749–2790. [Google Scholar] [CrossRef]
  204. Ravi, C.; Adimurthy, S. Synthesis of Imidazo[1,2-a]Pyridines: C-H Functionalization in the Direction of C-S Bond Formation. Chem. Rec. 2017, 17, 1019–1038. [Google Scholar] [CrossRef] [PubMed]
  205. Ghosh, S.; Laru, S.; Hajra, A. Ortho C−H Functionalization of 2-Arylimidazo[1,2-a]Pyridines. Chem. Rec. 2022, 22, e202100240. [Google Scholar] [CrossRef] [PubMed]
  206. Bower, J.D.; Ramage, G.R. Heterocyclic Systems Related to Pyrrocoline. Part I. 2: 3a-Diazaindene. J. Chem. Soc. 1955; 2834–2837. [Google Scholar] [CrossRef]
  207. Palazzo, G.; Picconi, G. Derivatives if imidazol (1,5-a) pyridin-3(2H)-one. Farm. Sci. 1975, 30, 197–207. [Google Scholar]
  208. Volpi, G.; Garino, C.; Breuza, E.; Gobetto, R.; Nervi, C. Dipyridylketone as a Versatile Ligand Precursor for New Cationic Heteroleptic Cyclometalated Iridium Complexes. Dalton Trans. 2011, 41, 1065–1073. [Google Scholar] [CrossRef] [PubMed]
  209. Volpi, G.; Garino, C.; Nervi, C. Exploring Synthetic Pathways to Cationic Heteroleptic Cyclometalated Iridium Complexes Derived from Dipyridylketone. Dalton Trans. 2012, 41, 7098–7108. [Google Scholar] [CrossRef] [PubMed]
  210. Andreo, L.; Volpi, G.; Rossi, F.; Benzi, P.; Diana, E. Two-Step Synthesis of a New Twenty-Membered Macrocycle: Spectroscopic Characterization and Theoretical Calculations. ChemistrySelect 2022, 7, e202202564. [Google Scholar] [CrossRef]
  211. Volpi, G.; Garino, C.; Priola, E.; Barolo, C. A New Auspicious Scaffold for Small Dyes and Fluorophores. Dye. Pigment. 2022, 197, 109849. [Google Scholar] [CrossRef]
  212. Rahmati, A.; Zahra, K. One-Pot Three-Component Synthesis of Imidazo[1,5-a]Pyridines. Int. J. Org. Chem. 2011, 01, 15–19. [Google Scholar] [CrossRef]
  213. Crawforth, J.M.; Paoletti, M. A One-Pot Synthesis of Imidazo[1,5-a]Pyridines. Tetrahedron Lett. 2009, 50, 4916–4918. [Google Scholar] [CrossRef]
  214. Siddiqui, S.; Potewar, T.; Lahoti, R.; Srinivasan, K. Ionic Liquid Promoted Facile One-Pot Synthesis of 1-Pyridylimidazo[1,5-a]Pyridines from Dipyridylketone and Aryl Aldehydes. Synthesis 2006, 2006, 2849–2854. [Google Scholar] [CrossRef]
  215. Aksenov, D.A.; Arutiunov, N.A.; Maliuga, V.V.; Aksenov, A.V.; Rubin, M. Synthesis of Imidazo[1,5-a]Pyridines via Cyclocondensation of 2-(Aminomethyl)Pyridines with Electrophilically Activated Nitroalkanes. Beilstein J. Org. Chem. 2020, 16, 2903–2910. [Google Scholar] [CrossRef] [PubMed]
  216. Fulwa, V.K.; Sahu, R.; Jena, H.S.; Manivannan, V. Novel Synthesis of 2,4-Bis(2-Pyridyl)-5-(Pyridyl)Imidazoles and Formation of N-(3-(Pyridyl)Imidazo[1,5-a]Pyridine)Picolinamidines: Nitrogen-Rich Ligands. Tetrahedron Lett. 2009, 50, 6264–6267. [Google Scholar] [CrossRef]
  217. Volpi, G.; Magistris, C.; Garino, C. FLUO-SPICES: Natural Aldehydes Extraction and One-Pot Reaction to Prepare and Characterize New Interesting Fluorophores. Educ. Chem. Eng. 2018, 24, 1–6. [Google Scholar] [CrossRef]
  218. Volpi, G.; Magistris, C.; Garino, C. Natural Aldehyde Extraction and Direct Preparation of New Blue Light-Emitting Imidazo[1,5-a]Pyridine Fluorophores. Nat. Prod. Res. 2018, 32, 2304–2311. [Google Scholar] [CrossRef] [PubMed]
  219. Joshi, A.; Mohan, D.C.; Adimurthy, S. Copper-Catalyzed Denitrogenative Transannulation Reaction of Pyridotriazoles: Synthesis of Imidazo[1,5-a]Pyridines with Amines and Amino Acids. Org. Lett. 2016, 18, 464–467. [Google Scholar] [CrossRef] [PubMed]
  220. Ramírez-Gómez, A.; Gutiérrez-Hernández, A.I.; Alvarado-Castillo, M.A.; Toscano, R.A.; Ortega-Alfaro, M.C.; López-Cortés, J.G. Selenoamides as Powerful Scaffold to Build Imidazo[1,5-a]Pyridines Using a Grinding Protocol. J. Organomet. Chem. 2020, 919, 121315. [Google Scholar] [CrossRef]
  221. Wang, L.-S.; Zhou, Y.; Lei, S.-G.; Yu, X.-X.; Huang, C.; Wu, Y.-D.; Wu, A.-X. Iodine-Mediated Multicomponent Cascade Cyclization and Sulfenylation/Selenation: Synthesis of Imidazo[2,1-a]Isoquinoline Derivatives. Org. Lett. 2022, 24, 4449–4453. [Google Scholar] [CrossRef]
  222. Sahoo, S.; Pal, S. Rapid Access to Benzimidazo[1,2-a]Quinoline-Fused Isoxazoles via Pd(II)-Catalyzed Intramolecular Cross Dehydrogenative Coupling: Synthetic Versatility and Photophysical Studies. J. Org. Chem. 2021, 86, 4081–4097. [Google Scholar] [CrossRef]
  223. Yamaguchi, E.; Shibahara, F.; Murai, T. Direct Sequential C3 and C1 Arylation Reaction of Imidazo[1,5-a]Pyridine Catalyzed by a 1,10-Phenanthroline–Palladium Complex. Chem. Lett. 2011, 40, 939–940. [Google Scholar] [CrossRef]
  224. Santra, S.; Mitra, S.; Bagdi, A.K.; Majee, A.; Hajra, A. Iron(III)-Catalyzed Three-Component Domino Strategy for the Synthesis of Imidazo[1,2-a]Pyridines. Tetrahedron Lett. 2014, 55, 5151–5155. [Google Scholar] [CrossRef]
  225. Albano, S.; Olivo, G.; Mandolini, L.; Massera, C.; Ugozzoli, F.; Di Stefano, S. Formation of Imidazo[1,5-a]Pyridine Derivatives Due to the Action of Fe2+ on Dynamic Libraries of Imines. J. Org. Chem. 2017, 82, 3820–3825. [Google Scholar] [CrossRef]
  226. Wang, L.; Zheng, X.; Zheng, Q.; Li, Z.; Wu, J.; Gao, G. Thioether-Assisted Cu-Catalyzed C5–H Arylation of Imidazo[1,5-a]Pyridines. Org. Lett. 2022, 24, 3834–3838. [Google Scholar] [CrossRef] [PubMed]
  227. Wang, H.; Xu, W.; Wang, Z.; Yu, L.; Xu, K. Copper-Catalyzed Oxidative Amination of Sp(3) C-H Bonds under Air: Synthesis of 1,3-Diarylated Imidazo[1,5-Alpha]Pyridines. J. Org. Chem. 2015, 80, 2431–2435. [Google Scholar] [CrossRef]
  228. Li, M.; Xie, Y.; Ye, Y.; Zou, Y.; Jiang, H.; Zeng, W. Cu(I)-Catalyzed Transannulation of N-Heteroaryl Aldehydes or Ketones with Alkylamines via C(Sp3)–H Amination. Org. Lett. 2014, 16, 6232–6235. [Google Scholar] [CrossRef] [PubMed]
  229. Lopes, E.F.; Saraiva, M.T.; Debia, N.P.; Silva, L.; Chaves, O.A.; Stieler, R.; Iglesias, B.A.; Rodembusch, F.S.; Lüdtke, D.S. Imidazo[1,2-a]Pyridine-Based Hybrids. Copper-Catalyzed Cycloaddition Synthesis, Photophysics, Docking, and Interaction Studies with Biomacromolecules. Dye. Pigment. 2023, 214, 111212. [Google Scholar] [CrossRef]
  230. Bluhm, M.E.; Ciesielski, M.; Görls, H.; Döring, M. Copper-Catalyzed Oxidative Heterocyclization by Atmospheric Oxygen. Angew. Chem. Int. Ed. 2002, 41, 2962–2965. [Google Scholar] [CrossRef]
  231. Bluhm, M.E.; Ciesielski, M.; Görls, H.; Walter, O.; Döring, M. Complexes of Schiff Bases and Intermediates in the Copper-Catalyzed Oxidative Heterocyclization by Atmospheric Oxygen§. Inorg. Chem. 2003, 42, 8878–8885. [Google Scholar] [CrossRef]
  232. Bluhm, M.E.; Folli, C.; Pufky, D.; Kröger, M.; Walter, O.; Döring, M. 3-Aminoiminoacrylate, 3-Aminoacrylate, and 3-Amidoiminomalonate Complexes as Catalysts for the Dimerization of Olefins. Organometallics 2005, 24, 4139–4152. [Google Scholar] [CrossRef]
  233. Wang, H.; Xu, W.; Xin, L.; Liu, W.; Wang, Z.; Xu, K. Synthesis of 1,3-Disubstituted Imidazo[1,5-a]Pyridines from Amino Acids via Catalytic Decarboxylative Intramolecular Cyclization. J. Org. Chem. 2016, 81, 3681–3687. [Google Scholar] [CrossRef] [PubMed]
  234. Lee, S.W.; Dao, P.D.Q.; Lim, H.-J.; Cho, C.S. Recyclable Magnetic Cu-MOF-74-Catalyzed C(Sp2)-N Coupling and Cyclization under Microwave Irradiation: Synthesis of Imidazo[1,2-c]Quinazolines and Their Analogues. ACS Omega 2023, 8, 16218–16227. [Google Scholar] [CrossRef] [PubMed]
  235. Qian, P.; Yan, Z.; Zhou, Z.; Hu, K.; Wang, J.; Li, Z.; Zha, Z.; Wang, Z. Electrocatalytic Intermolecular C(Sp3)–H/N–H Coupling of Methyl N-Heteroaromatics with Amines and Amino Acids: Access to Imidazo-Fused N-Heterocycles. Org. Lett. 2018, 20, 6359–6363. [Google Scholar] [CrossRef] [PubMed]
  236. Joshi, A.; Mohan, D.C.; Adimurthy, S. Lewis Acid-Catalyzed Denitrogenative Transannulation of Pyridotriazoles with Nitriles: Synthesis of Imidazopyridines. J. Org. Chem. 2016, 81, 9461–9469. [Google Scholar] [CrossRef] [PubMed]
  237. Wang, Q.; Zha, M.; Zhou, F.; Chen, X.; Wang, B.; Liu, G.; Qian, L. CBr 4 Mediated [4 + 1] Dehydrocyclization for the Synthesis of Functionalized Imidazo[1,5-a]Heterocycles from Pyridin-2-Ylmethanamines and Aldehydes. ACS Omega 2021, 6, 20303–20308. [Google Scholar] [CrossRef] [PubMed]
  238. Bori, J.; Manivannan, V. A New Synthetic Route for Synthesis of 3-substituted Imidazo[1,5-a]Pyridines. J. Heterocycl. Chem. 2022, 59, 1073–1078. [Google Scholar] [CrossRef]
  239. Grigg, R.; Kennewell, P.; Savic, V.; Sridharan, V. X=Y-Zh Systems as Potential 1,3-Dipoles Part 38. 1,5-Electrocyclization of Vinyl-Azomethine and Iminyl-Azomethine Ylides—2-Azaindolizines and Pyrrolo-Dihydroisoquinolines. Tetrahedron 1992, 48, 10423–10430. [Google Scholar] [CrossRef]
  240. Yan, Y.; Zhang, Y.; Zha, Z.; Wang, Z. Mild Metal-Free Sequential Dual Oxidative Amination of C(Sp3)–H Bonds: Efficient Synthesis of Imidazo[1,5-a]Pyridines. Org. Lett. 2013, 15, 2274–2277. [Google Scholar] [CrossRef] [PubMed]
  241. Hu, Z.; Hou, J.; Liu, J.; Yu, W.; Chang, J. Synthesis of Imidazo[1,5-a]Pyridines via I 2 -Mediated Sp 3 C–H Amination. Org. Biomol. Chem. 2018, 16, 5653–5660. [Google Scholar] [CrossRef] [PubMed]
  242. Shibahara, F.; Sugiura, R.; Yamaguchi, E.; Kitagawa, A.; Murai, T. Synthesis of Fluorescent 1,3-Diarylated Imidazo[1,5-a]Pyridines: Oxidative Condensation−Cyclization of Aryl-2-Pyridylmethylamines and Aldehydes with Elemental Sulfur as an Oxidant. J. Org. Chem. 2009, 74, 3566–3568. [Google Scholar] [CrossRef]
  243. Qin, M.; Tian, Y.; Guo, X.; Yuan, X.; Yang, X.; Chen, B. I2/TBPB Mediated Oxidative Reaction to Construct of Imidazo[1,5-α]Pyridines under Metal-Free Conditions. Asian J. Org. Chem. 2018, 7, 1591–1594. [Google Scholar] [CrossRef]
  244. Cui, T.; Zhan, Y.; Dai, C.; Lin, J.; Liu, P.; Sun, P. Electrochemical Oxidative Regioselective C–H Cyanation of Imidazo[1,2-a]Pyridines. J. Org. Chem. 2021, 86, 15897–15905. [Google Scholar] [CrossRef] [PubMed]
  245. da Silva, R.B.; Coelho, F.L.; Rodembusch, F.S.; Schwab, R.S.; Schneider, J.M.F.M.; da Silveira Rampon, D.; Schneider, P.H. Straightforward Synthesis of Photoactive Chalcogen Functionalized Benzimidazo[1,2-a]Quinolines. New J. Chem. 2019, 43, 11596–11603. [Google Scholar] [CrossRef]
  246. Qian, P.; Yan, Z.; Zhou, Z.; Hu, K.; Wang, J.; Li, Z.; Zha, Z.; Wang, Z. Electrocatalytic Tandem Synthesis of 1,3-Disubstituted Imidazo[1,5-a]Quinolines via Sequential Dual Oxidative C(Sp3)–H Amination in Aqueous Medium. J. Org. Chem. 2019, 84, 3148–3157. [Google Scholar] [CrossRef] [PubMed]
  247. Kim, S.-H.; Kim, Y.-M.; Park, B.-R.; Kim, J.-N. Synthesis of Imidazo[1,5-α]Quinolines and Imidazo[5,1-α]Isoquinolines via the In-Mediated Allylation of Reissert Compounds. Bull. Korean Chem. Soc. 2010, 31, 3031–3034. [Google Scholar] [CrossRef]
  248. Pelletier, G.; Charette, A.B. Triflic Anhydride Mediated Synthesis of Imidazo[1,5-a]Azines. Org. Lett. 2013, 15, 2290–2293. [Google Scholar] [CrossRef] [PubMed]
  249. Anderson, S.; Glover, E.E.; Vaughan, K.D. The N-Amination and Subsequent Oxidation by Bromine of Imidazo[1,5-a]Pyridines. J. Chem. Soc. Perkin Trans. 1 1976, 16, 1722–1724. [Google Scholar] [CrossRef]
  250. Fulwa, V.K.; Manivannan, V. Synthesis of Some 1,3-Disubstituted Imidazo[1,5-a]Pyridines Using 2-Cyanopyridine. Tetrahedron Lett. 2012, 53, 2420–2423. [Google Scholar] [CrossRef]
  251. Moulin, A.; Garcia, S.; Martinez, J.; Fehrentz, J.-A. Synthesis of Substituted Imidazo[1,5-a]Pyridines Starting from N-2-Pyridylmethylamides Using Lawesson’s Reagent and Mercury(II) Acetate. Synthesis 2007, 17, 2667–2673. [Google Scholar] [CrossRef]
  252. Nguyen, H.T.H.; Nguyen, O.T.K.; Truong, T.; Phan, N.T.S. Synthesis of Imidazo[1,5-a]Pyridines via Oxidative Amination of the C(Sp(3))-H Bond under Air Using Metal-Organic Framework Cu-MOF-74 as an Efficient Heterogeneous Catalyst. RSC Adv. 2016, 6, 36039–36049. [Google Scholar] [CrossRef]
  253. Zimmer, H.; Glasgow, D.G.; McClanahan, M.; Novinson, T. Imidazo[5,1-a]Isochinoline. Tetrahedron Lett. 1968, 9, 2805–2807. [Google Scholar] [CrossRef]
  254. Bori, J.; Behera, N.; Mahata, S.; Manivannan, V. Synthesis of Imidazo[5,1-a]Isoquinoline and Its 3-Substituted Analogues Including the Fluorescent 3-(1-Isoquinolinyl)Imidazo[5,1-a]Isoquinoline. ChemistrySelect 2017, 2, 11727–11731. [Google Scholar] [CrossRef]
  255. Middleton, R.W.; Wibberley, D.G. Synthesis of Imidazo[4,5-b]- and [4,5-c]Pyridines. J. Heterocycl. Chem. 1980, 17, 1757–1760. [Google Scholar] [CrossRef]
  256. Altaib, M.; Doganc, F.; Kaşkatepe, B.; Göker, H. Synthesis of Some New 2-(Substituted-Phenyl)Imidazo[4,5-c] and [4,5-b]Pyridine Derivatives and Their Antimicrobial Activities. Mol. Divers. 2023, 1–13. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Imidazopyridine family: principal reported heterocyclic skeletons obtained by the union of imidazole and pyridine units.
Figure 1. Imidazopyridine family: principal reported heterocyclic skeletons obtained by the union of imidazole and pyridine units.
Molecules 29 02668 g001
Figure 2. Main imidazo[1,2-a]pyridine-based drugs (the imidazo[1,2-a]pyridine core is represented here in blue).
Figure 2. Main imidazo[1,2-a]pyridine-based drugs (the imidazo[1,2-a]pyridine core is represented here in blue).
Molecules 29 02668 g002
Figure 3. Principal imidazo[1,5-a]pyridine biologically active derivatives (the imidazo[1,5-a]pyridine core is represented here in green, the names of the represented products are listed in the text according to the given numbers).
Figure 3. Principal imidazo[1,5-a]pyridine biologically active derivatives (the imidazo[1,5-a]pyridine core is represented here in green, the names of the represented products are listed in the text according to the given numbers).
Molecules 29 02668 g003
Figure 4. Principal imidazopyridine nuclei and the corresponding positions for possible derivatization and introduction of substituent groups (different imidazopyridine cores filled with the colors used in Figure 1).
Figure 4. Principal imidazopyridine nuclei and the corresponding positions for possible derivatization and introduction of substituent groups (different imidazopyridine cores filled with the colors used in Figure 1).
Molecules 29 02668 g004
Figure 5. Main structures of imidazo[1,5-a]pyridinic (green) and imidazo[1,2-a]pyridinic (blue) derivatives employed as N-O, N-N, or multi-dentate ligands.
Figure 5. Main structures of imidazo[1,5-a]pyridinic (green) and imidazo[1,2-a]pyridinic (blue) derivatives employed as N-O, N-N, or multi-dentate ligands.
Molecules 29 02668 g005
Figure 6. Early reported synthetic approaches to obtain substituted imidazo[1,2-a]pyridine core (blue), inspired by [45,82].
Figure 6. Early reported synthetic approaches to obtain substituted imidazo[1,2-a]pyridine core (blue), inspired by [45,82].
Molecules 29 02668 g006
Figure 7. Complete reaction mechanism of one of the most used synthetic approaches involving substituted ketones and 2-aminopyridine to obtain imidazo[1,2-a]pyridine derivatives (blue), mechanism reported in [43,45].
Figure 7. Complete reaction mechanism of one of the most used synthetic approaches involving substituted ketones and 2-aminopyridine to obtain imidazo[1,2-a]pyridine derivatives (blue), mechanism reported in [43,45].
Molecules 29 02668 g007
Figure 8. Early reported synthetic approaches to obtain substituted imidazo[1,5-a]pyridine core (green), inspired by [12].
Figure 8. Early reported synthetic approaches to obtain substituted imidazo[1,5-a]pyridine core (green), inspired by [12].
Molecules 29 02668 g008
Figure 9. Complete reaction mechanism of one of the most used synthetic approaches to obtain imidazo[1,5-a]pyridine derivatives involving aldehydes and pyridyl-2-ketones, mechanism reported in [12,126].
Figure 9. Complete reaction mechanism of one of the most used synthetic approaches to obtain imidazo[1,5-a]pyridine derivatives involving aldehydes and pyridyl-2-ketones, mechanism reported in [12,126].
Molecules 29 02668 g009
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

Volpi, G.; Laurenti, E.; Rabezzana, R. Imidazopyridine Family: Versatile and Promising Heterocyclic Skeletons for Different Applications. Molecules 2024, 29, 2668. https://doi.org/10.3390/molecules29112668

AMA Style

Volpi G, Laurenti E, Rabezzana R. Imidazopyridine Family: Versatile and Promising Heterocyclic Skeletons for Different Applications. Molecules. 2024; 29(11):2668. https://doi.org/10.3390/molecules29112668

Chicago/Turabian Style

Volpi, Giorgio, Enzo Laurenti, and Roberto Rabezzana. 2024. "Imidazopyridine Family: Versatile and Promising Heterocyclic Skeletons for Different Applications" Molecules 29, no. 11: 2668. https://doi.org/10.3390/molecules29112668

Article Metrics

Back to TopTop