This article provides a comprehensive overview for researchers and drug development professionals on the strategic exploration of natural product (NP) chemical space to uncover novel therapeutic leads.
This article provides a comprehensive overview for researchers and drug development professionals on the strategic exploration of natural product (NP) chemical space to uncover novel therapeutic leads. It covers the foundational concept of the biologically relevant chemical space (BioReCS), where NPs occupy unique and underexplored regions compared to synthetic libraries. The piece details cutting-edge methodological approaches, including AI-driven screening, genomics, and high-throughput assays, and addresses significant challenges such as supply, characterization, and regulatory hurdles. By validating the success of NPs through approved drugs and comparative analyses, the article underscores NPs' irreplaceable role in addressing unmet medical needs, particularly in antimicrobial and anticancer therapy, and outlines a future roadmap for integration with innovative technologies.
The concept of chemical space (CS), also referred to as the "chemical universe," is a foundational concept in modern drug discovery and many other chemical disciplines [1]. While often used intuitively, chemical space is formally defined as a multidimensional space where molecular propertiesâboth structural and functionalâdefine coordinates and relationships between compounds [1]. Within this vast universe, the Biologically Relevant Chemical Space (BioReCS) comprises the subset of molecules with biological activity, spanning both beneficial compounds (therapeutics) and detrimental ones (toxins) [1].
The exploration of chemical space is particularly crucial for drug discovery, as the theoretical number of possible small organic molecules below 500 Da is estimated to exceed 10^60 structures [2]. This immense size makes comprehensive experimental screening impossible, necessitating intelligent navigation strategies to identify promising regions for bioactive molecule discovery, especially those inspired by or derived from natural products [3].
BioReCS encompasses all molecules that interact with biological systems, creating a complex landscape of chemical subspaces (ChemSpas) distinguished by shared structural or functional features [1]. This space includes not only drug-like molecules but also agrochemicals, flavor and odor chemicals, food components, and natural products [1]. A critical aspect of BioReCS is that it includes compounds with both desirable and undesirable biological effects, including promiscuous binders, poly-active molecules, and toxic compounds [1].
Systematic study of BioReCS requires molecular descriptors that define the dimensionality of the space, with the choice of descriptors depending on project goals, compound classes, and dataset characteristics [1]. The rise of machine learning has further driven the development of novel molecular representations that can efficiently navigate these complex spaces [1].
Chemical compound databases serve as essential resources for exploring BioReCS. The table below summarizes major public databases covering different regions of the biologically relevant chemical space.
Table 1: Representative Public Compound Databases Covering Different Regions of BioReCS
| Database Name | Primary Focus | Key Applications |
|---|---|---|
| ChEMBL [1] | Bioactive small molecules, primarily organic compounds | Major source for poly-active and promiscuous structures; drug discovery |
| PubChem [1] | Bioactive small molecules with extensive annotations | Biological activity analysis; chemical biology research |
| InertDB [1] | Curated and AI-generated inactive compounds | Defining non-biologically relevant chemical space; negative data for machine learning |
| Dark Chemical Matter [1] | Compounds repeatedly inactive in HTS assays | Understanding chemical features associated with lack of bioactivity |
The exploration of BioReCS has been uneven, with certain regions receiving extensive attention while others remain largely uncharted:
The vastness of chemical space necessitates sophisticated computational approaches for efficient navigation. Several algorithmic strategies have been developed to handle trillion-sized compound collections:
Table 2: Key Algorithmic Approaches for Chemical Space Exploration
| Algorithm | Search Principle | Key Applications |
|---|---|---|
| FTrees [4] | Fuzzy pharmacophore similarity | Identifying close analogs with similar pharmacophore properties |
| SpaceLight [4] | Molecular fingerprint similarity (ECFP/CSFP) | High-throughput similarity screening using Tanimoto metrics |
| SpaceMACS [4] | Maximum common substructure (MCS) | Scaffold-based similarity searching and analysis |
These algorithms enable researchers to identify close neighbors of known bioactive compounds within massive virtual chemical spaces. For example, screening FDA-approved drugs against the eXplore chemical space (containing 2.8 trillion virtual molecules) demonstrated that these methods can retrieve high-similarity analogs for a significant percentage of known drugs, providing starting points for drug optimization campaigns [4].
Natural products represent a privileged region of BioReCS, having evolved through biological selection processes to interact with macromolecular targets [3]. Strategies for natural product-informed exploration of chemical space include:
These approaches have enabled the discovery of novel bioactive molecules that might not have been identified through traditional screening methods, providing access to distinctive regions of BioReCS [3].
The structural diversity across BioReCS presents challenges for consistent chemical space analysis using traditional descriptors optimized for specific compound classes [1]. Ongoing efforts aim to develop universal molecular descriptors that can accommodate diverse chemical types:
The following diagram illustrates a generalized workflow for exploring chemical space in drug discovery, particularly emphasizing natural product-inspired approaches:
Figure 1: Workflow for Natural Product-Informed Drug Discovery
A critical consideration in BioReCS exploration is the pH-dependent nature of many bioactive compounds [1]. Most chemoinformatics analyses assume neutral charge states, yet approximately 80% of contemporary drugs are ionizable under physiological conditions [1]. This ionization significantly impacts solubility, permeability, absorption, distribution, toxicity, and target binding, necessitating methods that account for charged species in chemical space analysis [1].
The high-dimensional nature of chemical space requires dimensionality reduction techniques for visualization and interpretation [1]. Common approaches include:
These visualization approaches enable researchers to identify clusters of compounds with similar properties, locate sparsely populated regions of chemical space that may represent opportunities for novel discovery, and understand the relationship between natural products and synthetic compounds [5] [2].
Table 3: Essential Research Tools for Chemical Space Exploration
| Tool/Category | Specific Examples | Function in BioReCS Exploration |
|---|---|---|
| Chemical Databases | ChEMBL, PubChem, ZINC, GDB [1] [2] | Source of annotated chemical structures and bioactivity data |
| Similarity Search Tools | FTrees, SpaceLight, SpaceMACS [4] | Identify analogs and nearby compounds in chemical space |
| Molecular Descriptors | ECFP, MAP4, Molecular Quantum Numbers [1] | Numeric representations encoding chemical structure |
| Visualization Platforms | Chemical cartography tools, SOM implementations [5] | 2D/3D projection of high-dimensional chemical space |
| Virtual Screening | Docking, Pharmacophore screening [6] | Computational prioritization of compounds for testing |
Modern drug discovery increasingly leverages network-based multi-omics integration to understand complex biological systems and their interaction with chemical space [7]. These approaches combine various molecular data types (genomics, transcriptomics, proteomics) with biological networks (protein-protein interaction, drug-target interaction) to better predict drug responses, identify novel targets, and facilitate drug repurposing [7].
For natural product research, this means positioning natural compounds within broader biological context networks, connecting their chemical structures to target networks, metabolic pathways, and phenotypic effects [7]. Method categories include:
Research has demonstrated that natural product-informed exploration of chemical space enables the discovery of distinctive and novel bioactive small molecules [3]. These approaches help focus molecular discovery on biologically relevant regions of chemical space, increasing the likelihood of identifying useful chemical probes and therapeutic candidates [3].
The relationship between natural products, chemical space exploration, and drug discovery can be visualized as follows:
Figure 2: Natural Product-Informed BioReCS Exploration
The exploration of BioReCS faces several important challenges and opportunities:
As these challenges are addressed, the systematic exploration of biologically relevant chemical space, particularly regions inspired by natural products, will continue to drive innovation in drug discovery and chemical biology.
Natural products (NPs) from plants, animals, and microorganisms have served as a cornerstone of pharmacotherapy throughout human history, providing a rich source of structurally diverse and biologically active compounds for treating human diseases [10] [11]. These secondary metabolites represent an invaluable chemical resource, with over half of approved small-molecule drugs originating directly or indirectly from natural product scaffolds [12] [10]. The structural complexity and evolutionary optimization of natural products for biological interaction make them exceptionally suited for drug discovery, particularly for challenging targets such as protein-protein interactions [1] [12].
Within the framework of exploring natural product chemical space for drug discovery research, this review examines the biologically relevant chemical space (BioReCS) of natural products, which encompasses molecules with both beneficial and detrimental biological activities [1]. Current databases document over 1.1 million natural products that display high structural diversity and complexity, frequently featuring glycosylation and halogenation patterns that distinguish them from synthetic compounds [12]. Despite a declining discovery rate of novel structures, natural products continue to offer unique scaffolds that occupy broader chemical spaces than synthetic compounds, positioning them as an indispensable resource for addressing current therapeutic challenges [12] [10].
The relationship between natural products and human medicine dates back to ancient healing traditions, with well-documented use in Ayurvedic medicine, Traditional Chinese Medicine (TCM), Japanese Kampo, and European phytotherapy [10]. These traditional systems provided the initial framework for exploring nature's pharmacopeia, with many modern drugs tracing their origins to ethnobotanical and ethnopharmacological knowledge [11].
The pharmaceutical industry's engagement with natural products has experienced significant fluctuations over recent decades. The 1990s witnessed a "Green Rush" in natural product research, driven by advancements in high-throughput screening (HTS) and isolation technologies that enabled systematic exploration of biodiversity [11]. This period saw substantial investment in bioprospecting initiatives targeting terrestrial and marine organisms for novel drug leads. However, in the early 2000s, most major pharmaceutical companies terminated or significantly reduced their HTS and natural product discovery programs in favor of combinatorial chemistry and rational drug design approaches [11].
Contemporary analysis reveals that the relatively low productivity of purely synthetic approaches has quietly repositioned pharmacognosy back into the drug discovery mainstream [11]. Current estimates indicate that approximately 50% of FDA-approved medications between 1981â2006 were natural products or synthetic derivatives inspired by natural products, highlighting their enduring impact despite fluctuating industrial interest [13]. This reemergence recognizes that natural products offer structural complexity and biological relevance that remains challenging to replicate through purely synthetic approaches [12] [11].
Table 1: Therapeutic Areas Significantly Influenced by Natural Product-Derived Drugs
| Therapeutic Area | Representative Drugs | Natural Source | Clinical Significance |
|---|---|---|---|
| Oncology | Paclitaxel, Docetaxel, Trabectedin | Pacific Yew Tree, European Yew, Marine Tunicate | Taxanes represent cornerstone therapies for various cancers; marine-derived agents offer novel mechanisms |
| Infectious Diseases | Penicillins, Tetracyclines, Erythromycin | Fungi, Soil Bacteria | Foundation of anti-infective therapies with diverse mechanisms against pathogens |
| Immunosuppression | Cyclosporine, Fingolimod | Soil Fungus, Fungus Isaria sinclairii | Revolutionized organ transplantation; advanced multiple sclerosis treatment |
| Neurological Disorders | Galantamine, Huperzine A | Daffodil bulbs, Chinese Herb Huperzia serrata | Acetylcholinesterase inhibition for Alzheimer's management |
Table 2: Structural and Property Comparisons Between Natural Products and Synthetic Compounds
| Property | Natural Products | Synthetic Compounds | Biological Implications |
|---|---|---|---|
| Structural Complexity | High (multiple chiral centers, intricate ring systems) | Moderate to Low | Enhanced target selectivity and novel binding modes |
| Molecular Weight | Broader distribution, including bRo5 space | Typically focused on lower MW | Access to challenging target classes like PPIs |
| Oxygen Atoms | Higher count | Lower count | Improved hydrogen bonding capacity |
| Stereochemical Complexity | High | Variable to Low | Biological specificity and metabolic stability |
| Chemical Space Coverage | Broader, underexplored regions | Narrower, focused on drug-like space | Access to novel bioactive scaffolds |
Analysis of natural product chemical space reveals distinct structural characteristics that contribute to their biological success. Natural products frequently exhibit higher stereochemical complexity, greater abundance of oxygen atoms, and more varied ring systems compared to synthetic compounds [12]. These properties enable natural products to interact with complex biological targets through unique binding modes often inaccessible to synthetic libraries [1] [12]. Marine natural products, for instance, demonstrate particularly novel scaffolds with potent bioactivities, exemplified by the development of trabectedin from a marine tunicate [11].
Systematic exploration of natural product chemical space requires robust chemoinformatic approaches to characterize structural diversity, bioactivity patterns, and source-related characteristics. Natural products exhibit distinct chemical features based on their biological origins, with marine-derived compounds generally displaying higher molecular weight and hydrophobicity compared to terrestrial counterparts [12]. NPs from extreme environments such as deep-sea ecosystems and extremophiles frequently reveal novel scaffolds with unique bioactivities, highlighting the value of biodiversity exploration in drug discovery [12].
The concept of the biologically relevant chemical space (BioReCS) provides a framework for understanding natural products' privileged status in therapeutic development. BioReCS encompasses all molecules with biological activityâboth beneficial and detrimentalâspanning drug discovery, agrochemistry, sensory chemistry, and toxicological domains [1]. Within this framework, natural products occupy regions characterized by high structural diversity and complexity, often distinct from synthetic compounds [1] [12].
Key chemoinformatic analyses have revealed that natural products contain a higher prevalence of unique ring systems with different atom compositions and connectivity compared to synthetic molecules [12]. This structural novelty translates to diverse biological interactions and mechanisms of action. Furthermore, natural products frequently undergo specific biochemical modifications such as glycosylation and halogenation that enhance their biological activities and target affinity [12].
Despite extensive research, significant regions of natural product chemical space remain underexplored, presenting opportunities for future discovery. Several compound classes are notably underrepresented in current databases and drug discovery efforts:
These structurally complex natural products often fall into the beyond Rule of 5 (bRo5) category, presenting challenges for synthesis and optimization but offering unique opportunities for addressing difficult therapeutic targets [1]. Recent studies have begun systematically characterizing these underrepresented regions, including peptides, agrochemicals, metallodrugs, macrocycles, and PPI modulators [1].
The systematic investigation of natural products for drug discovery follows established experimental workflows that integrate traditional knowledge with modern analytical techniques. The process typically begins with source selection guided by ethnobotanical knowledge, ecological considerations, or biodiversity surveys, followed by careful specimen collection and authentication [10] [11].
Table 3: Key Methodologies in Natural Product Isolation and Characterization
| Method Category | Specific Techniques | Applications in NP Drug Discovery |
|---|---|---|
| Extraction & Fractionation | Bioassay-guided fractionation, Solvent-solvent partitioning, Liquid-liquid chromatography | Selective enrichment of bioactive compounds from complex mixtures |
| Compound Isolation | High-performance liquid chromatography (HPLC), Countercurrent chromatography, Flash chromatography | Purification of individual natural products from crude extracts |
| Structure Elucidation | NMR spectroscopy (1D/2D), Mass spectrometry (MS), X-ray crystallography | Determination of molecular structure and stereochemistry |
| Bioactivity Screening | High-throughput screening (HTS), Phenotypic assays, Target-based assays | Identification of biologically active natural products |
Bioassay-guided fractionation represents a cornerstone approach, wherein biological activity tracking directs the isolation of active constituents from complex natural extracts [13]. This method ensures that purification efforts focus on compounds with relevant biological effects, increasing the efficiency of lead identification. Advances in analytical technologies, particularly NMR and mass spectrometry, have dramatically accelerated the structure elucidation process, enabling determination of complex structures with minimal material [13] [11].
The following workflow diagram illustrates the integrated experimental and computational approach for natural product-based drug discovery:
The integration of computational methods has transformed natural product research, enabling more efficient exploration of chemical space and prediction of bioactivity. Computer-aided drug design (CADD) approaches, particularly artificial intelligence (AI) and machine learning (ML), have demonstrated significant utility in navigating the complex chemical space of natural products [13].
AI-driven approaches include:
Machine learning algorithms, including support vector machines (SVMs), neural networks, and decision trees, enable pattern recognition in complex structure-activity relationship data [13]. Deep learning approaches, particularly convolutional neural networks (CNNs) and recurrent neural networks (RNNs), facilitate analysis of molecular structures and prediction of bioactive conformations [13]. Natural language processing (NLP) techniques further enhance these approaches by extracting relevant information from scientific literature, patents, and natural product databases [13].
The following diagram illustrates the integration of AI technologies in natural product drug discovery:
The transition from initial bioactive natural product hits to viable lead compounds requires systematic approaches to evaluate and optimize chemical structures. Lead identification begins with validating biological activity through dose-response experiments and specificity assessments, followed by comprehensive characterization of absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties [14].
High-throughput screening (HTS) and ultra-high-throughput screening (UHTS) methodologies enable efficient evaluation of extensive natural product libraries, with capacity reaching up to 100,000 assays per day using automated robotic systems [14]. These approaches offer significant advantages over traditional screening methods, including enhanced automation, reduced sample volumes, improved sensitivity, and cost savings in reagents and culture media [14].
Hit validation involves rigorous assessment of:
Confirmed hits progress to lead optimization, where medicinal chemistry strategies enhance desirable properties while mitigating limitations. The lead optimization phase involves synthesis and characterization of analog structures, evaluation using biochemical assays (e.g., Irwin's test for neurobehavioral assessment, Ames test for genotoxicity), and detailed analysis of drug-induced metabolism through metabolic profiling [14].
Structure-activity relationship (SAR) studies form the foundation of natural product optimization, systematically exploring how structural modifications influence biological activity and drug-like properties. SAR analysis identifies critical pharmacophoric elementsâthe specific molecular features essential for biological activityâand guides strategic modifications to enhance efficacy and reduce toxicity [15].
Key strategies in natural product analog design include:
The iterative process of analog design and optimization follows a cyclical approach of design, synthesis, testing, and refinement. This process continues until compounds achieve the optimal balance of potency, selectivity, and drug-like properties required for preclinical development [15].
Table 4: Key Research Reagent Solutions for Natural Product Drug Discovery
| Reagent/Category | Specific Examples | Function in NP Research |
|---|---|---|
| Analytical Standards | Certified reference materials, Deuterated solvents, Quantitative NMR standards | Compound identification, quantification, method validation |
| Bioassay Kits | Enzyme inhibition assays, Cell viability assays, Receptor binding assays | Biological activity assessment, mechanism elucidation |
| Chromatography Materials | HPLC columns, Solid-phase extraction cartridges, Countercurrent chromatography solvents | Compound separation, purification, and enrichment |
| Molecular Biology Reagents | Protein expression systems, Enzyme substrates, Reporter gene assays | Target identification and validation, mechanism studies |
| Computational Tools | Molecular docking software, QSAR programs, Cheminformatics platforms | Virtual screening, property prediction, SAR analysis |
The field of natural product drug discovery is experiencing significant transformation through the integration of emerging technologies and interdisciplinary approaches. Several key trends are shaping the future of this field:
Artificial Intelligence and Cheminformatics: AI-driven approaches are revolutionizing natural product research through enhanced pattern recognition in complex chemical and biological data [13]. Chemical language models and neural network embeddings generate chemically meaningful representations that can reconstruct molecular structures or predict properties, accelerating the identification of promising bioactive molecules [1]. The development of universal molecular descriptors, such as molecular quantum numbers and the MAP4 fingerprint, enables more consistent analysis of natural product chemical space across diverse compound classes [1].
Integration of Multi-Omics Technologies: Genomic, transcriptomic, and metabolomic approaches provide unprecedented insights into biosynthetic pathways and ecological functions of natural products [10]. These technologies facilitate the identification of gene clusters responsible for natural product biosynthesis, enabling heterologous expression and engineering of novel analogs [12].
Exploration of Underexplored Biodiversity: Research continues to focus on extreme environments (deep-sea, deserts, polar regions) and symbiotic relationships (endophytic fungi, microbial symbionts) as sources of novel natural products with unique scaffolds and bioactivities [12]. These ecosystems offer chemical diversity distinct from traditional sources, with marine natural products particularly promising for anticancer and antiviral applications [13].
Despite promising advances, natural product drug discovery faces several persistent challenges that require innovative solutions:
Supply and Sustainability Issues: Many natural products occur in minute quantities in their source organisms, creating supply challenges for development and large-scale production [13]. Sustainable sourcing strategies, including cultivation, partial synthesis, and biotechnology approaches, are essential for addressing ecological concerns and ensuring consistent supply [11].
Technical Complexities in Characterization: The structural complexity of natural products presents challenges for synthesis, structural elucidation, and optimization [13]. Advances in synthetic methodologies, analytical technologies, and computational prediction are gradually overcoming these barriers, making complex natural products more accessible for drug discovery [15].
Data Integration and Quality: The lack of standardized data quality and reporting in natural product research hampers data mining and reproducibility [10]. Initiatives to improve data curation, implement standardized protocols, and develop integrated databases are critical for advancing the field [1] [12].
The historical legacy of natural products as a pillar of pharmacotherapy continues to evolve through the integration of traditional knowledge with contemporary scientific approaches. As technological innovations provide new tools for exploring natural product chemical space, the unique structural features and biological relevance of natural products ensure their continued importance in addressing current and future therapeutic challenges. By leveraging advances in AI, omics technologies, and synthetic biology, researchers can unlock the full potential of nature's chemical diversity for the development of next-generation therapeutics.
Natural products (NPs) and their derivatives have historically been a cornerstone of pharmacotherapy, accounting for over 60% of all small-molecule drugs approved between 1981 and 2014 [16] [17]. Despite this proven utility, synthetic compounds (SCs) dominate most commercial screening libraries, constrained by decade-old conventions like Lipinski's Rule of Five and synthetic accessibility [16]. This preference persists even as challenging biological targetsâsuch as protein-protein interactions, nucleic acid complexes, and antibacterial modalitiesâoften remain recalcitrant to libraries of drug-like molecules [18].
The fundamental advantage of NPs lies in their evolutionary origin. As products of natural selection, they have co-evolved to interact with biological macromolecules, encoding inherent biological relevance and an ability to explore a broader swath of biologically relevant chemical space [19] [20]. Consequently, NPs exhibit structural featuresâsuch as increased molecular complexity, higher fractions of sp³-hybridized carbons, and greater stereochemical densityâthat are often underrepresented in synthetic libraries [18] [16]. This manuscript demonstrates how Principal Component Analysis (PCA) serves as a powerful computational tool to visualize and quantify this superior diversity, providing a compelling rationale for reintegrating NPs into modern drug discovery pipelines.
In chemoinformatics, chemical space is defined as a multi-dimensional descriptor space where each molecule is represented by a numerical vector encoding aspects of its structure or physicochemical properties [21]. The concept of a chemical multiverse acknowledges that the chemical space of a single dataset is not unique; it is a "group of multiple chemical spaces, each defined by a given set of descriptors" [21]. The visual representation of this space, therefore, depends critically on the chosen descriptors and dimensionality reduction techniques.
PCA is a mathematical method for dimensionality reduction that transforms a multidimensional dataset into a new set of orthogonal axes called principal components (PCs) [22]. These components are linear combinations of the original descriptors, with the first PC (PC1) capturing the maximum variance in the data, the second PC (PC2) capturing the next highest variance, and so on [22]. By projecting high-dimensional data onto a two- or three-dimensional plot, PCA allows for intuitive visualization of similarities, differences, and patterns within compound collections with minimal loss of information [22]. When applied to collections of NPs and SCs, PCA vividly reveals the distinct regions these classes occupy and their relative diversity.
A comprehensive, time-dependent chemoinformatic analysis comparing NPs from the Dictionary of Natural Products with SCs from 12 databases reveals distinct evolutionary trajectories. NPs discovered over time have become larger, more complex, and more hydrophobic [19]. Specifically, descriptors of molecular sizeâincluding molecular weight, molecular volume, and the number of heavy atomsâshow a consistent upward trend in NPs, a phenomenon attributed to advances in separation and purification technologies [19].
Conversely, the physicochemical properties of SCs have been constrained within a narrower range, largely governed by drug-like rules and synthetic accessibility [19]. Table 1 summarizes key differentiating properties based on analyses of hundreds of thousands of compounds [19] [16].
Table 1: Key Physicochemical and Structural Differences Between Natural Products and Synthetic Compounds
| Property | Natural Products (NPs) | Synthetic Compounds (SCs) |
|---|---|---|
| Molecular Size | Generally larger; size increasing over time [19] | Smaller; constrained by drug-like rules [19] |
| Fraction of sp³ Carbons (Fsp3) | Higher, indicating more 3D character [16] | Lower, indicating more flat, aromatic structures [18] |
| Stereochemical Complexity | Higher number of stereocenters [19] [16] | Fewer stereocenters [18] |
| Ring Systems | More rings, larger fused rings, more non-aromatic rings [19] | More aromatic rings (e.g., benzene derivatives) [19] |
| Oxygen & Nitrogen Content | More oxygen atoms [19] | More nitrogen atoms [19] |
| Biological Relevance | High, due to evolutionary selection [19] [20] | Broader synthetic pathways but declining relevance [19] |
A PCA analysis utilizing 16 two-dimensional structural descriptors on a combined ~390,000 NPs and SCs clearly demonstrates the greater structural variability of NPs [16]. The NPs occupy a broader, more dispersed region in the PCA plot, particularly evident in properties like the fraction of sp³ carbon atoms (Fsp3), a key metric of molecular complexity [16].
Another powerful visualization tool is the Tree MAP (TMAP), a two-dimensional tree-based clustering algorithm built for large-scale data. When clusters are generated using molecular fingerprints (MHFP), NPs occupy vast structural areas that are largely unexplored by synthetic molecules [16]. The TMAP visualization further corroborates that NPs are structurally more complex, not only in Fsp3 but also in features like the number of spiroatoms [16].
This section provides a detailed methodology for reproducing the chemical space comparisons described in this review.
1. Source Natural Product Databases:
2. Source Synthetic Compound Database:
3. Data Cleaning and Standardization:
Calculate the following 16 two-dimensional molecular descriptors for each standardized compound. This can be accomplished using software such as ChemAxon's Instant JChem or the RDKit library in Python [22] [16] [21].
Table 2: Essential Molecular Descriptors for PCA of Chemical Space
| Descriptor | Description | Interpretation in NP/SC Context |
|---|---|---|
| MW | Molecular Weight | NPs are generally larger [19]. |
| LogP | Partition coefficient (octanol/water) | Measures lipophilicity [22]. |
| TPSA | Topological Polar Surface Area | Related to polarity and hydrogen bonding [22]. |
| a_acc | Number of hydrogen bond acceptors | NPs often have more oxygen atoms [19]. |
| a_don | Number of hydrogen bond donors | NPs often have more donors [22]. |
| a_heavy | Number of heavy atoms | Indicator of molecular size [19]. |
| b_rotR | Fraction of rotatable bonds | Related to molecular flexibility [22]. |
| a_nN | Number of nitrogen atoms | SCs are often richer in nitrogen [19]. |
| a_nO | Number of oxygen atoms | NPs are often richer in oxygen [19]. |
| FCharge | Sum of formal charges | Influences solubility and interactions. |
| a_aro | Number of aromatic atoms | SCs typically have more aromatic character [19]. |
| chiral | Number of chiral centers | NPs have higher stereochemical complexity [16]. |
| rings | Number of rings | NPs tend to have more ring systems [19]. |
| stereo | Number of stereocenters | Key indicator of NP complexity [22] [16]. |
| fsp3 | Fraction of sp³ hybridized carbons | Critical measure of 3D complexity; higher in NPs [18] [16]. |
| a_spiro | Number of spiro atoms | Indicator of complex ring fusions; higher in NPs [16]. |
The following workflow diagram summarizes the experimental protocol for chemical space analysis:
Table 3: Key Software and Resources for Chemical Space Analysis
| Tool/Resource | Type | Function in Analysis |
|---|---|---|
| RDKit | Open-source Cheminformatics Library | Data standardization, descriptor calculation, and fingerprint generation [16] [21]. |
| Instant JChem | Commercial Cheminformatics Platform | Management of chemical data, batch calculation of physicochemical parameters [22]. |
| R / Python (scikit-learn) | Programming Environments | Performing Principal Component Analysis and statistical computations [22]. |
| VCC Lab ALOGPS | Web Service | Calculating additional properties like logP and aqueous solubility [22]. |
| MolVS | Open-source Library | Standardizing molecular structures (tautomers, charges, fragments) [21]. |
| FooDB | Public Database | Source of natural product structures, particularly food-related chemicals [21]. |
| ZINC Database | Public Database | Source of commercially available synthetic compound structures [16]. |
| Kakuol | Kakuol, CAS:18607-90-4, MF:C10H10O4, MW:194.18 g/mol | Chemical Reagent |
| 2,4,6-Trihydroxybenzaldehyde | 2,4,6-Trihydroxybenzaldehyde, CAS:487-70-7, MF:C7H6O4, MW:154.12 g/mol | Chemical Reagent |
The clear visualization of NP diversity has direct, practical implications for drug discovery. The finding that NPs explore vast, biologically relevant regions of chemical space that SCs do not reach provides a strong rationale for designing new libraries that capture these underrepresented features [18] [20]. Several strategies have emerged to bridge this gap:
PCA can guide these efforts by quantifying how well a new library penetrates NP-like regions of chemical space. As demonstrated in one study, analyzing the component loadings can identify which structural parameters (e.g., number of oxygen atoms, stereochemical density, Fsp3) most influence the separation between NPs and SCs. Chemists can then target these specific parameters through synthetic modification to "shift" their compounds towards the NP region of the PCA plot [22]. This data-driven approach enables a more rational and effective exploration of nature's vast chemical repertoire for drug discovery.
Principal Component Analysis provides an unambiguous visual and quantitative demonstration of the superior structural diversity inherent in natural products compared to synthetic chemical libraries. The broader distribution of NPs in chemical space, characterized by greater molecular complexity, stereochemical richness, and distinct physicochemical properties, underscores their immense and irreplaceable value for drug discovery. By leveraging PCA as a guide for library design and analysis, researchers can move beyond the constraints of traditional drug-like chemical space, harnessing the evolutionary wisdom encoded in natural products to develop novel therapeutics for the most challenging biological targets.
Natural products (NPs) represent a vast and structurally diverse resource for drug discovery, comprising over 173,000 known structures that have evolved to interact with biological systems [23]. The concept of "chemical space" refers to a multidimensional universe where molecular properties define coordinates and relationships between compounds, with the biologically relevant chemical space (BioReCS) encompassing molecules with demonstrated biological activity [1]. Within this framework, natural products occupy a strategic position, as they largely adhere to the Rule of Five while simultaneously exploring regions of chemical space not covered by synthetic compounds and available screening collections [24]. This renders them a valuable, unique, and necessary component of screening libraries used in drug discovery. Analyses of 10,495 natural products and 5,757 trade drugs reveal that natural products possess 1,748 different ring systems compared to 807 different ring systems found in trade drugs, demonstrating their superior structural diversity [23]. Despite this proven potential, significant portions of the natural product chemical universe remain underexplored, creating opportunities for discovering novel bioactive compounds with resistance-breaking properties and new mechanisms of action, particularly in challenging therapeutic areas like antimicrobial resistance [25].
The systematic organization of natural products enables effective navigation of their chemical space. Several classification approaches have been developed, with structural classification of natural products (SCONP) emerging as a powerful organizing principle [26]. SCONP arranges the scaffolds of natural products in a tree-like fashion, providing both an analysis- and hypothesis-generating tool for the design of natural product-derived compound collections [26]. This approach facilitates the identification of biologically relevant subfractions of chemical space and has been successfully applied in the development of novel inhibitor classes, such as selective and potent inhibitors of 11β-hydroxysteroid dehydrogenase type 1 with cellular activity [26].
Alternative classification systems group natural products according to recurring structural features. For instance, flavonoid compounds are oxygenated derivatives of a specific aromatic ring structure, while alkaloids containing an indole ring are classified as indole alkaloids [27]. These structural classifications complement biosynthetic organization systems, which categorize compounds based on their metabolic pathways of origin within producing organisms [27]. Each classification approach offers distinct advantages for drug discovery, with structural systems enabling scaffold-based diversity analysis and biosynthetic systems facilitating genomics-guided discovery.
Computational methods have become indispensable for mapping and navigating natural product chemical space. ChemGPS-NP and Scaffold Hunter represent two widely used tools that enable researchers to explore biologically relevant NP chemical space in a focused and targeted fashion [24]. These cheminformatics platforms help bridge the gap between computational methods and compound library synthesis, integrating cheminformatics and chemical space analyses with synthetic chemistry and biochemistry to successfully identify novel small molecule modulators of protein function [24].
The analytical power of these tools stems from their ability to process multidimensional molecular descriptors that define the dimensionality of chemical space [1]. Recent advances include the development of more universal molecular descriptors, such as MAP4 fingerprints and neural network embeddings from chemical language models, which can accommodate entities ranging from small molecules to biomolecules [1]. These tools are particularly valuable for identifying "holes" in existing screening data setsâregions of chemical space that can and should be explored by chemistry and biology to discover new bioactive compounds [24].
Table 1: Structural and Property-Based Comparison of Natural Products and Trade Drugs
| Characteristic | Natural Products | Trade Drugs | Data Source |
|---|---|---|---|
| Average Molecular Weight | 356 | 360 | [23] |
| Average log P value | 2.9 | 2.5 | [23] |
| Number of Ring Systems | 1,748 | 807 | [23] |
| Rule-of-5 Violations | Similar percentage | Similar percentage | [23] |
| Hydrogen Bond Donors | Fewer per molecule | More per molecule | [23] |
| Bridgehead Atoms | Much higher number | Lower number | [23] |
| Chiral Centers | Many more per molecule | Fewer per molecule | [23] |
Table 2: Heavily Explored vs. Underexplored Regions of NP Chemical Space
| Aspect | Heavily Explored Regions | Underexplored Regions | Research Implications |
|---|---|---|---|
| Structural Classes | Flavonoids, indole alkaloids, opium alkaloids, common scaffold systems | Macrocycles, RiPPs (ribosomally synthesized and post-translationally modified peptides), metallodrugs | New structural motifs with potentially novel mechanisms of action [27] [28] [1] |
| Source Organisms | Soil-derived actinomycetes, terrestrial plants | Microbes from extreme environments, marine symbionts, cyanobacteria, hot sulfur springs | Unique biosynthetic pathways and enzymatic transformations [25] |
| Chemical Space Properties | Drug-like properties, Rule-of-5 compliance, well-characterized pharmacology | Beyond Rule of 5 (bRo5) compounds, protein-protein interaction inhibitors, PROTACs | Challenges in synthesis and optimization, but potential for targeting difficult therapeutic areas [28] [1] |
| Discovery Approaches | Bioactivity-guided fractionation, traditional natural product chemistry | Genome mining, metabolomics, bioengineering, synthetic biology | Access to cryptic biosynthetic gene clusters and previously inaccessible chemical diversity [25] [23] |
The data reveal that while natural products share many drug-like properties with trade drugs, they explore significantly more structural diversity, particularly in complex ring systems and stereochemistry [23]. This structural complexity contributes to their biological relevance but also presents challenges for synthesis and modification. The underexplored regions of NP chemical space are characterized by structural classes that fall outside traditional drug-like property space, source organisms from extreme or difficult-to-access environments, and novel biosynthetic pathways [25] [28] [1].
Certain classes of natural products have been extensively investigated due to their historical therapeutic success and relative accessibility. Flavonoids and alkaloids represent two such heavily explored families, with well-established biosynthetic pathways, known pharmacological activities, and extensive structure-activity relationship data [27]. These compounds typically exhibit favorable drug-like properties, with molecular weights and log P values falling within ranges comparable to approved drugs [23]. The structural classification of natural products (SCONP) has further illuminated that certain molecular scaffolds recur frequently among known natural products, creating regions of chemical space that have been systematically explored for drug discovery [26].
The heavy exploration of these regions is evidenced by the fact that more than 100 marketed macrocycle drugs are almost exclusively derived from natural products, yet this structural class remains poorly explored within targeted drug discovery efforts [28]. Similarly, natural products have contributed significantly to approved drugs across multiple therapeutic areas: 78% of antibacterial drugs, 75% of platelet aggregation inhibitors, 61% of anticancer drugs, 48% of anti-hypotensive drugs, 47.6% of antiulcer drugs, and 32.5% of anti-inflammatory drugs have a natural origin [23]. This extensive exploration has generated robust structure-activity relationship data for these compound classes but has also led to diminishing returns in discovering truly novel chemotypes from traditional sources.
The heavy focus on specific natural product classes and source organisms has resulted in significant redundancy in discovery efforts. Recent analyses indicate that although the total number of characterized natural products has increased over the last decades, only a small percentage of recently discovered compounds possess previously unknown chemical structures [25]. This repetition stems from several factors: the repeated isolation of known compounds from related species, the focus on easily cultivable microorganisms from similar ecological niches, and the application of standardized extraction and isolation procedures that selectively capture certain chemical classes while missing others.
This redundancy presents a substantial challenge for drug discovery, particularly in areas like antibiotic development where structurally new chemicals are urgently required for resistance-breaking properties [25]. The known natural product chemical space likely represents only "the tip of the iceberg," with significant biosynthetic potential remaining concealed in underexplored organisms, environments, and biosynthetic pathways [25]. Overcoming this limitation requires deliberate exploration of untapped regions of NP chemical space through innovative approaches and technologies.
Several structural classes of natural products remain underexplored despite their significant potential for drug discovery. Macrocycles, defined as compounds containing rings of 12 or more atoms, represent a particularly promising yet underexploited structural class [28]. These compounds provide diverse functionality and stereochemical complexity in a conformationally pre-organized ring structure, which can result in high affinity and selectivity for protein targets while preserving sufficient bioavailability to reach intracellular locations [28]. Macrocycles have demonstrated repeated success when addressing targets that have proved highly challenging for standard small-molecule drug discovery, especially in modulating macromolecular processes such as protein-protein interactions [28].
Other underexplored structural classes include ribosomally synthesized and post-translationally modified peptides (RiPPs), which exhibit remarkable structural diversity and bioactivities [25]. Recent research has identified ribosomally derived lipopeptides containing distinct fatty acyl moieties as a promising area for exploration [25]. Additionally, metal-containing natural products represent a structurally and functionally important class that is commonly excluded from standard chemoinformatics analyses due to modeling challenges [1]. The difficulty of modeling these regions of BioReCS should not justify their exclusion from systematic exploration, as they may offer unique therapeutic opportunities.
The biosynthetic potential of certain microbial groups and extreme environments remains largely untapped. Cyanobacteria and microbes that colonize extreme habitats represent talented but neglected natural product producers [25]. These organisms often possess unique biosynthetic pathways evolved to produce specialized metabolites under challenging environmental conditions, resulting in chemical structures not found in organisms from conventional sources.
Recent advances in metagenomics have revealed that the wealth of publicly available (meta)genomes conceals significant biosynthetic potential that has yet to be elucidated [25]. One comprehensive study of the global ocean microbiome uncovered extensive biosynthetic diversity, with thousands of new biosynthetic gene clusters identified in marine microorganisms [25]. The isolation of natural products from habitats and organisms previously thought to lack natural product biosynthesis potential (e.g., hot sulfur springs) further supports the hypothesis that known natural product chemical space represents only a fraction of what exists in nature [25].
A particularly intriguing underexplored region of biologically relevant chemical space consists of so-called "dark chemical matter" â compounds that have repeatedly failed to show activity in high-throughput screening assays [1]. These molecules represent the non-biologically relevant portions of chemical space and provide crucial boundary conditions for understanding bioactivity. Recent efforts have led to the development of InertDB, a curated collection of 3,205 experimentally confirmed inactive compounds supplemented with 64,368 putative inactive molecules generated using deep generative artificial intelligence models [1]. Understanding why these compounds lack activity can provide equally valuable insights for drug discovery as studying successful bioactive molecules.
The integration of genomic information with natural product chemistry has emerged as a powerful approach for targeted exploration of underexplored regions of NP chemical space. The following protocol outlines a genomics-guided discovery workflow:
Table 3: Research Reagent Solutions for Genomics-Guided NP Discovery
| Research Reagent | Function/Application | Experimental Role |
|---|---|---|
| Metagenomic DNA Libraries | Source of biosynthetic gene clusters from unculturable microorganisms | Provides access to genetic potential of microbial communities without cultivation [25] |
| Heterologous Expression Systems | Host organisms for expressing foreign biosynthetic gene clusters | Enables production of natural products from unculturable or genetically intractable organisms [25] |
| Bioinformatics Tools (e.g., antiSMASH) | Identification and analysis of biosynthetic gene clusters in genomic data | Guides target selection and predicts structural features of encoded natural products [25] |
| Mass Spectrometry Platforms | Detection and structural characterization of novel natural products | Links biosynthetic gene clusters to their metabolic products through metabolomics [23] |
Protocol Steps:
Bioengineering provides powerful methods to access underexplored regions of natural product chemical space through targeted modification of biosynthetic pathways:
Protocol Steps:
The systematic exploration of underexplored regions of natural product chemical space requires an integrated approach combining multiple scientific disciplines and methodologies. The following diagram illustrates the workflow for discovering novel natural products from underexplored sources, highlighting the interdisciplinary nature of modern natural product research:
Table 4: Essential Research Tools and Resources for NP Chemical Space Exploration
| Tool/Resource Category | Specific Examples | Application in NP Research |
|---|---|---|
| Chemical Space Navigation | ChemGPS-NP, Scaffold Hunter | Guide exploration of biologically relevant NP chemical space in a focused and targeted fashion [24] |
| Bioinformatics Platforms | antiSMASH, PRISM, MIBiG | Identify and analyze biosynthetic gene clusters in genomic and metagenomic data [25] |
| Analytical Technologies | LC-HRMS, MS Imaging, NMR | Detect, characterize, and visualize natural products in complex biological matrices [23] |
| Genomic Resources | Metagenomic libraries, Heterologous expression systems | Access biosynthetic potential of unculturable microorganisms and engineer biosynthetic pathways [25] |
| Specialized Compound Libraries | Macrocyclic libraries, RiPP libraries, Dark chemical matter collections | Focus screening efforts on underexplored regions of chemical space [28] [1] |
The systematic exploration of natural product chemical space represents a crucial frontier in drug discovery, particularly as resistance to existing therapies grows and challenging targets require innovative chemical solutions. While heavily explored regions of NP chemical space have provided numerous therapeutic agents, they face diminishing returns in yielding truly novel chemotypes. In contrast, underexplored regionsâincluding macrocycles, RiPPs, metabolites from extreme environments, and cryptic biosynthetic pathwaysâoffer significant opportunities for discovering compounds with resistance-breaking properties and novel mechanisms of action. The integrated application of genomics, bioinformatics, synthetic biology, and advanced analytical technologies provides powerful methods to navigate and populate these underexplored regions of natural product chemical space. As computational tools continue to evolve and our understanding of biosynthetic pathways expands, targeted exploration of these underexplored regions will play an increasingly important role in addressing unmet medical needs through natural product-inspired drug discovery.
The concept of the biologically relevant chemical space (BioReCS) provides a foundational framework for modern drug discovery. BioReCS encompasses all molecules with biological activityâboth beneficial and detrimentalâspanning diverse application areas including drug discovery, agrochemistry, and natural product research [1]. This chemical universe is vast, with estimates suggesting the existence of up to 10^60 drug-like compounds, creating a fundamental challenge for researchers seeking to identify novel therapeutic agents [29]. Within this expansive universe, natural products (NPs) occupy a particularly privileged region, characterized by unique structural complexity and high relevance to human biology. Analyses reveal that over half of approved small-molecule drugs originate directly or indirectly from natural products, highlighting their enduring importance [12].
The structural and physicochemical properties of natural products differ significantly from typical synthetic compounds. Natural products often feature greater stereochemical complexity, higher sp³-hybridized carbon counts, more oxygen atoms, and intricate ring systems that confer sophisticated three-dimensional architectures [30]. These characteristics enable natural products to interact with challenging biological targets, including protein-protein interactions, which have proven difficult to modulate with conventional synthetic compounds [30]. Despite the known structural diversity of natural products, current databases document approximately 1.1 million natural products, with only about 10% readily obtainable for experimental testing, creating a significant accessibility challenge [12]. This gap between known structures and readily testable compounds represents a critical bottleneck in natural product-based drug discovery.
Table 1: Key Characteristics of Natural Product Chemical Space
| Property | Natural Products | Synthetic/Drug-like Compounds |
|---|---|---|
| Structural Complexity | High (multiple stereocenters, complex ring systems) | Variable, often lower |
| sp³ Hybridized Carbons | Higher fraction (Fsp³) | Lower fraction |
| Oxygen Content | High | Variable |
| Number of Aromatic Rings | Generally lower | Generally higher |
| Relevance to Drug Discovery | >50% of approved drugs NP-derived | Foundation of combinatorial libraries |
| Readily Accessible Compounds | ~10% of known structures | High percentage |
Systematic exploration of natural product chemical space requires specialized computational tools that can map its complex topography. Platforms such as ChemGPS-NP and Scaffold Hunter enable researchers to navigate biologically relevant regions in a focused manner, identifying both densely populated and sparsely explored areas [24]. These cheminformatic tools employ dimensionality reduction techniques to project high-dimensional chemical descriptor data into visualizable and interpretable formats, allowing researchers to identify structural patterns and anomalies across large compound collections [1].
Recent advances in molecular representation have been critical for effective chemical space analysis. While traditional descriptors were optimized for small organic molecules, newer approaches like the MAP4 fingerprint and neural network embeddings from chemical language models offer more universal representations that can accommodate diverse molecular classes ranging from small molecules to peptides and even metallodrugs [1]. These improved descriptors facilitate more meaningful comparisons across different regions of chemical space and enable the identification of truly novel scaffolds with potential bioactivity.
Analysis of natural product chemical space reveals several underexplored regions with high potential for drug discovery. Certain structural classes remain underrepresented in current screening collections, including metal-containing molecules, large and complex natural products, macrocycles, protein-protein interaction (PPI) modulators, PROTACs, and mid-sized peptides [1]. Many of these compounds fall into the "beyond Rule of 5" (bRo5) category, presenting both challenges and opportunities for drug development [1].
Marine natural products represent another distinctive region of chemical space, characterized by larger molecular weights and greater hydrophobicity compared to their terrestrial counterparts [12]. Particularly interesting are natural products derived from deep-sea and extremophile organisms, which often display novel scaffolds and notable bioactivities honed by adaptation to unique environmental conditions [12]. The continued discovery of such structurally distinct compounds highlights the value of exploring diverse biological sources.
Beyond these structural classes, the concept of "dark chemical matter"âcompounds that consistently show no activity in high-throughput screensâprovides valuable negative data that helps define the boundaries of BioReCS [1]. Similarly, databases of curated inactive compounds, such as InertDB, which includes both experimentally determined and AI-generated putative inactive molecules, contribute to our understanding of the structural features that separate bioactive from non-bioactive chemical space [1].
Table 2: Key Public Databases for Exploring Natural Product Chemical Space
| Database | Scope | Key Features |
|---|---|---|
| COCONUT (Collection of Open Natural Products) | Comprehensive NP collection | >400,000 fully characterized natural products [31] |
| ChEMBL | Bioactive drug-like molecules | Extensive biological activity annotations [1] |
| PubChem | Chemical substances and bioactivities | Large repository with screening data [1] |
| Super Natural II | Natural products | Includes predicted bioactivity and pathways [12] |
| NAPRORE-CR | Costa Rican natural products | Geographically focused NP database [12] |
| PeruNPDB | Peruvian natural products | Regional focus for drug screening [12] |
Several sophisticated synthesis strategies have emerged to systematically populate promising regions of natural product chemical space with novel compounds. These approaches leverage the structural information encoded in natural products while introducing significant diversity to explore surrounding chemical space.
Biology-Oriented Synthesis (BIOS) proceeds from the premise that natural products are "privileged structures" with inherent biological relevance. This strategy employs natural products as starting points for designing focused libraries that retain core structural elements of the original bioactive compound while introducing strategic modifications [32] [30]. For example, Waldmann's development of an oxepane-based library inspired by bioactive natural products like heliannuol B and zoapatanol led to the discovery of novel Wnt signaling modulators that interact with the previously undrugged target Vangl1 [32]. BIOS libraries typically contain fewer compounds than traditional combinatorial libraries but demonstrate higher hit rates due to their foundation in evolutionarily validated scaffolds.
Diversity-Oriented Synthesis (DOS) aims to generate broad structural diversity through branching reaction pathways that produce compounds with varied skeletons and stereochemistries from common intermediates [32] [30]. Unlike target-oriented synthesis, DOS employs forward synthetic analysis to create structurally complex and diverse libraries that populate expansive regions of chemical space. A prominent example includes Schreiber's work generating a library of 2,070 macrolactone-based small molecules, which led to the discovery of robotnikininâa potent inhibitor of the Hedgehog signaling pathway with potential applications in cancer treatment [30]. DOS libraries are particularly valuable for phenotypic screening campaigns where the biological targets may not be fully characterized.
Pharmacophore-Directed Retrosynthesis (PDR) represents a more recent strategy that integrates synthetic planning with the identification of key structural features essential for bioactivity [32]. This approach begins with a retrospective analysis of structure-activity relationships to identify critical pharmacophoric elements, then designs synthetic routes that maximize opportunities to generate analogs exploring variations in these key features. PDR aims to balance the efficiency of total synthesis with the systematic investigation of structure-activity relationships throughout the synthetic process.
Diagram 1: NP-Inspired Synthesis Strategies
Beyond comprehensive library synthesis strategies, recent methodological advances enable precise molecular editing that can dramatically expand accessible chemical space from advanced intermediates. These approaches are particularly valuable for lead optimization phases where subtle structural modifications can significantly improve drug properties.
Skeletal Editing techniques allow direct modification of molecular frameworks through atom insertion, deletion, or exchange. A groundbreaking example is the development of sulfenylcarbene-mediated carbon atom insertion into N-heterocycles, which enables the transformation of existing drug scaffolds into new candidates by adding just one carbon atom at room temperature under metal-free conditions [33]. This method achieves yields up to 98% and is compatible with DNA-encoded library technology, making it particularly valuable for late-stage diversification of lead compounds [33]. The ability to perform such precise molecular surgery represents a paradigm shift in medicinal chemistry, potentially reducing drug development costs by enabling efficient renovation of existing molecular structures rather than requiring de novo synthesis.
Ring Distortion of Natural Products capitalizes on the complex ring systems found in many natural products by subjecting them to reaction conditions that dramatically rearrange their core structures. This approach can generate diverse, natural product-like compounds that would be challenging to access through conventional synthesis. The resulting libraries maintain the three-dimensional complexity and fraction of sp³-hybridized carbons characteristic of bioactive natural products while exploring unprecedented structural space around the original scaffold.
Hybrid Natural Products combine structural elements from two or more biologically active natural products to create novel compounds with potentially enhanced or dual activities. This strategy mimics nature's own evolutionary approach, as exemplified by the potent anticancer natural product vincristine, which represents a hybrid of the simpler alkaloids vindoline and catharanthine [30]. Synthetic hybridization enables the exploration of chemical space between known bioactive regions, potentially yielding compounds with novel mechanisms of action.
Table 3: Experimental Approaches for Chemical Space Exploration
| Methodology | Key Reagents/Techniques | Applications | Experimental Considerations |
|---|---|---|---|
| Skeletal Editing | Sulfenylcarbene reagents, metal-free conditions | Late-stage functionalization, lead optimization | Bench-stable reagents, room temperature operation, compatibility with DNA-encoded libraries [33] |
| DOS | Branching pathways, multicomponent reactions, complexity-generating transformations | Library generation for phenotypic screening | Aim for â¤5 steps, incorporate stereochemical diversity, use pluripotent intermediates [30] |
| BIOS | Natural product-inspired building blocks, target-oriented synthesis | Focused library for specific target classes | Prioritize privileged NP scaffolds, employ divergent synthesis from common intermediates [32] |
| Ring Distortion | Lewis acids, oxidants, rearranging conditions | Generating complexity and diversity from NP starting materials | Use stable, readily available NPs, employ multiple reaction conditions on single substrate [30] |
The effective exploration of natural product chemical space requires tight integration between computational prediction and experimental validation. Advanced screening protocols leverage the complementary strengths of both approaches to efficiently navigate vast molecular libraries toward promising bioactive compounds.
Active Learning with Alchemical Free Energy Calculations represents a powerful workflow that combines machine learning with physics-based binding affinity predictions. In this approach, an initial set of compounds is evaluated using computationally intensive alchemical free energy calculations, which provide highly accurate binding affinity estimates [29]. These data then train machine learning models that can rapidly predict affinities for much larger compound libraries. The most promising compounds from these predictions (selected through strategies like greedy selection or mixed uncertainty sampling) are subsequently validated through additional free energy calculations, creating an iterative refinement cycle that efficiently converges toward high-affinity binders [29]. This approach dramatically reduces the computational resources required to screen large libraries while maintaining high prediction accuracy.
High-Throughput Synthetic Platform technologies have emerged to accelerate the production and analysis of compound libraries. For instance, Blair and colleagues developed an approach that simplified the analysis of thousands of simultaneous reactions by focusing on molecular fragments, reducing analysis time from two months to a single day while generating 5,000 new chemicals through 20,000 reactions [34]. Such platforms address the critical bottleneck in chemical space exploration by enabling rapid synthesis and characterization of diverse compound collections, making large-scale investigation of underexplored chemical regions practically feasible.
Diagram 2: Integrated Screening Workflow
Artificial intelligence has revolutionized natural product discovery by enabling the generation of novel natural product-like structures that expand beyond known chemical space. Deep generative models, particularly recurrent neural networks (RNNs) with long short-term memory (LSTM) units trained on natural product SMILES representations, can produce vast libraries of novel yet biologically relevant structures [31]. One such effort generated 67 million natural product-like moleculesâa 165-fold expansion over the approximately 400,000 known natural productsâwhile maintaining distributions of natural product-likeness scores similar to authentic natural products [31].
These AI-generated compound libraries significantly expand the accessible chemical space for drug discovery, populating regions with structural novelty while maintaining the favorable physicochemical properties associated with natural products. The generated compounds exhibit expanded physiochemical and structural space compared to known natural products, as visualized through t-SNE projections of molecular descriptors [31]. This approach effectively inverts the traditional discovery process by first generating promising virtual structures that can then be prioritized for synthesis and testing, potentially uncovering entirely new classes of bioactive compounds.
Table 4: Essential Research Reagents and Materials for Chemical Space Exploration
| Reagent/Material | Function | Application Examples |
|---|---|---|
| Sulfenylcarbene Reagents | Carbon atom insertion into N-heterocycles | Late-stage skeletal editing of drug candidates [33] |
| DNA-Encoded Library (DEL) Components | Facilitating high-throughput screening of billions of compounds | Building diverse libraries for protein binding screens [33] |
| Pluripotent Building Blocks | Branching point substrates for DOS | Generating diverse scaffolds from common intermediates [30] |
| Solid-Supported Phosphonates | Facilitating parallel synthesis and purification | DOS library generation with simplified workup [30] |
| Natural Product Fragment Libraries | Starting points for BIOS and hybrid molecules | Exploring NP-inspired regions of chemical space [12] |
| Bench-Stable Carbene Precursors | Safe, metal-free carbene generation | Sustainable skeletal editing compatible with DEL [33] |
| Chromatin/Nucleosome Assembly Kits | Creating biologically relevant screening substrates | Targeting epigenetic mechanisms in drug discovery [34] |
| Perfluorotetradecanoic acid | Perfluorotetradecanoic acid, CAS:376-06-7, MF:C13F27COOH, MW:714.11 g/mol | Chemical Reagent |
| Thielavin A | Thielavin A|IDO Inhibitor|CAS 71950-66-8 | Thielavin A is a fungal depside with research value as an indoleamine 2,3-dioxygenase (IDO) inhibitor. This product is For Research Use Only. Not for human use. |
The systematic exploration of natural product chemical space represents a paradigm shift in drug discovery, moving from serendipitous finding to rational design. By integrating advanced cheminformatic analysis with innovative synthetic methodologies, researchers can now navigate the vast landscape of possible drug-like molecules with unprecedented precision and efficiency. The strategies outlinedâfrom biology-oriented and diversity-oriented synthesis to skeletal editing and AI-generated molecular designâprovide a comprehensive toolkit for populating underexplored yet biologically relevant regions of chemical space.
Future advances will likely focus on improving the integration between computational prediction and experimental validation, further accelerating the discovery cycle. Additionally, as synthetic methodologies continue to evolve, particularly those enabling late-stage functionalization and skeletal editing, the ability to fine-tune molecular properties while maintaining core bioactivity will become increasingly sophisticated. The ongoing development of open-access natural product databases and analysis tools will further democratize access to chemical space exploration, potentially unlocking novel therapeutic opportunities for challenging disease targets. Through the continued bridging of chemical space analysis and novel synthesis, drug discovery can more effectively harness the rich structural diversity evolved in nature while expanding into entirely new regions of chemical space with designed synthetic compounds.
The exploration of natural product (NP) chemical space represents a frontier of untapped therapeutic potential, historically plagued by insurmountable complexity and scale. The process of identifying bioactive compounds and predicting their properties from millions of candidate structures has been a protracted, resource-intensive endeavor. The integration of Artificial Intelligence (AI) and Machine Learning (ML) is now fundamentally reshaping this landscape, transforming NP-based drug discovery from a slow, empirical process into a predictive, data-driven science [35] [13]. These technologies are enabling researchers to navigate the vast, intricate chemical space of NPsâa space estimated to contain over 1060 drug-like moleculesâwith unprecedented speed and precision [36] [37]. This whitepaper provides an in-depth technical guide to the core AI/ML methodologies revolutionizing target identification and property prediction within the context of natural product drug discovery, detailing experimental protocols, benchmarking data, and essential computational tools for the modern research scientist.
Natural products are chemical compounds or substances produced by living organisms, including plants, animals, and microorganisms [13]. They have served as a rich source of biologically active compounds, with approximately 50% of FDA-approved medications between 1981 and 2006 being NPs or their synthetic derivatives [13]. However, the discovery of drugs derived from NPs presents numerous challenges, including the limited availability of bioactive molecules, the complexity of molecular structures, low yields of promising compounds, and the labor-intensive process of isolation and structural elucidation [13].
The accelerating growth of make-on-demand and virtual chemical libraries provides unprecedented opportunities but also creates a fundamental bottleneck. While these libraries now contain >70 billion readily available molecules, the number of possible drug-like molecules is estimated to be more than 1060, exceeding the size of chemical libraries evaluated in early drug discovery by many orders of magnitude [37]. This disparity highlights the critical need for more efficient virtual screening approaches capable of evaluating these vast chemical libraries [37].
Table 1: Key Challenges in Natural Product Drug Discovery and AI-Driven Solutions
| Challenge | Traditional Approach | AI/ML Solution | Impact |
|---|---|---|---|
| Dereplication | Manual literature review & experimental comparison | AI-powered database mining & pattern recognition [38] | Reduces redundant discovery efforts |
| Target Identification | Bioassay-guided fractionation | Predictive bioactivity modeling & reverse docking [13] [38] | Accelerates hypothesis generation |
| Property Prediction | Empirical structure-activity relationship (SAR) studies | Quantitative Structure-Activity/Property Relationship (QSA/PR) models [39] [38] | Enables in silico ADMET profiling |
| Chemical Space Exploration | Limited library screening | Deep generative models & latent space navigation [40] [41] | Expands access to novel scaffolds |
AI's role in this domain is rapidly expanding. Analysis of the CAS Content Collection, the largest human-curated collection of published scientific information, found over 600,000 scientific publications related to natural product research since 2010, with a notable increase in AI applications [38]. The most common AI application in natural products is in anti-tumor agents, followed by antiviral and antibacterial agents [38].
Structure-based virtual screening of ultralarge libraries has identified ligands of important therapeutic targets, but evaluating massive libraries requires substantial computational resources [37]. A breakthrough strategy combining machine learning and molecular docking enables rapid virtual screening of databases containing billions of compounds, reducing the computational cost by more than 1,000-fold [37].
The core protocol involves training a classification algorithm to identify top-scoring compounds based on molecular docking of a subset (e.g., 1 million compounds) to the target protein. The conformal prediction framework then makes selections from the multi-billion-scale library, drastically reducing the number of compounds requiring explicit docking scoring [37]. In application to a library of 3.5 billion compounds, this protocol successfully identified ligands of G protein-coupled receptors (GPCRs), one of the most important families of drug targets [37].
Table 2: Performance Benchmark of ML-Guided Docking Workflow [37]
| Metric | Standard Docking | ML-Guided Docking | Improvement |
|---|---|---|---|
| Library Size | 3.5 billion compounds | 3.5 billion compounds | - |
| Compounds Docked | 3.5 billion | ~25-30 million | >100-fold reduction |
| Computational Cost | ~493 trillion complexes predicted | Not specified | >1,000-fold reduction |
| Sensitivity | Baseline (100%) | 87-88% | Maintains high recall |
| Experimental Hit Rate | Target-dependent | Successfully identified GPCR ligands | Confirmed utility |
Objective: To identify potential ligands for a target protein from an ultralarge chemical library while reducing computational requirements by >100-fold.
Input Requirements:
Methodology:
The predictive power of AI in property prediction hinges on effective molecular representations. Quantitative Structure-Activity Relationship (QSAR) modeling correlates numerical molecular descriptors with biological activity or physicochemical properties [39]. These descriptors are classified by dimensions:
Modern AI approaches have expanded these to include learned representations. Deep learning techniques generate "deep descriptors" derived from molecular graphs or SMILES strings without manual engineering, capturing abstract and hierarchical molecular features [39].
Table 3: Molecular Representations in AI-Driven Drug Discovery [39] [43]
| Representation | Description | AI Applications | Advantages | Limitations |
|---|---|---|---|---|
| SMILES Strings | 1D linear notation of chemical structure | RNN, LSTM, Transformers (ChemBERTa) [43] | Simple, compact, widely supported | Non-unique, sensitive to syntax errors |
| Molecular Fingerprints | Bit vectors indicating substructure presence (ECFP4, Morgan) | Classical ML, Deep Learning [37] [43] | Fixed-length, suitable for similarity search | Lack 3D stereochemical detail |
| Molecular Graphs | Atom-bond networks with nodes and edges | Graph Neural Networks (GNN, GCN, GAT) [39] [43] | Preserves atomic connectivity and topology | Computationally expensive |
| 3D Representations | Atomic coordinates and spatial relationships | SchNet, DimeNet, GeoMol [43] | Captures stereochemistry and shape | Requires conformer generation |
Objective: To develop a predictive QSAR model for ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) properties or bioactivity of natural products.
Data Curation:
Feature Engineering & Model Training:
Model Validation & Interpretation:
The therapeutic potential of natural products is often confined to specific regions of chemical space. Deep generative models provide an alternative approach to explore wider drug-like chemical spaces [40] [41]. These models can generate novel molecular structures with desired properties, capturing the chemical space of known drugs while expanding into unexplored territories [40].
Conditional generative models, such as the Conditional Randomized Transformer with molecular fingerprints as a condition, can perform guided exploration in drug-like chemical space [40]. The combination of quantitative estimation of drug-likeness (QED) and quantitative estimate of protein-protein interaction targeting drug-likeness (QEPPI) can cover a larger drug-like space than either metric alone [40].
Objective: To generate novel, synthetically accessible natural product-inspired compounds with optimized properties.
Model Selection & Training:
Generation & Optimization:
Validation & Synthesis Planning:
Successful implementation of AI-driven NP discovery requires access to specialized computational tools, databases, and experimental reagents. The following table catalogs essential resources referenced in contemporary literature.
Table 4: Essential Research Reagents & Computational Tools for AI-Driven NP Discovery
| Resource Name | Type | Function/Application | Reference |
|---|---|---|---|
| Enamine REAL Space | Chemical Library | >70 billion make-on-demand compounds for virtual screening | [37] |
| CAS Content Collection | Database | Human-curated collection of published scientific information on NPs | [38] |
| CETSA (Cellular Thermal Shift Assay) | Experimental Method | Validates direct target engagement in intact cells and tissues | [42] |
| CatBoost | ML Algorithm | Gradient boosting classifier optimal for molecular fingerprint data | [37] |
| RDKit | Cheminformatics | Open-source toolkit for descriptor calculation & cheminformatics | [37] [39] |
| NRPSpredictor2 | Web Server | Predicts substrate specificity of NP biosynthetic enzymes using ML | [38] |
| AutoDock, SwissADME | Software Platform | Molecular docking and ADMET property prediction | [42] |
| QSARINS | Software | Development and validation of classical QSAR models | [39] |
| NuBBE Database | Database | Specialized natural product database from Brazilian biodiversity | [38] |
| Leucomycin A6 | Leucomycin A6, CAS:18361-48-3, MF:C40H65NO15, MW:799.9 g/mol | Chemical Reagent | Bench Chemicals |
| Bepridil | Bepridil|Calcium Channel Blocker|For Research Use | Bepridil is a multi-target calcium channel blocker for cancer, virology, and cardiology research. For Research Use Only. Not for human consumption. | Bench Chemicals |
The integration of AI and ML into natural product research marks a paradigm shift from serendipitous discovery to rational, predictive exploration. The emerging "lab-in-a-loop" concept, where AI algorithms are continuously refined using real-world experimental data, promises a future of autonomous, adaptive, and exponentially accelerating drug discovery [43]. This closed-loop, self-improving ecosystem represents the next frontier, transforming drug development from a linear, human-driven process into a cyclical, AI-driven process with human oversight [43].
However, challenges remain in the widespread adoption of these technologies. Data quality and standardization continue to be significant hurdles, particularly for natural products with complex stereochemistry and limited available data [38]. Model interpretability, regulatory acceptance, and the need for large-scale experimental validation of AI-generated designs are additional areas requiring continued focus [39] [41]. As these challenges are addressed, AI-driven exploration of natural product space will undoubtedly unlock novel therapeutic avenues, harnessing the best of what nature has to offer to address human disease.
High-Throughput Screening (HTS) represents an automated, robust approach to rapidly testing large collections of molecules for bioactivity, holding particular promise in antibacterial drug discovery where over 50% of marketed antibiotics originate from natural products (NPs) [44]. The screening of natural product libraries (NPLs) presents unique challenges and opportunities due to the complex chemical nature of NP extracts, which contain a plethora of molecules at varying concentrations with potential for antagonistic or synergistic biological activities [44]. Within this paradigm, two primary screening philosophies have emerged: cellular target-based HTS (CT-HTS) and molecular target-based HTS (MT-HTS). The selection between these approaches carries significant implications for hit identification, validation, and subsequent development within the broader context of exploring natural product chemical space for drug discovery research. This technical guide examines both methodologies, their experimental protocols, and their application in modern drug discovery pipelines.
Definition and Principle: CT-HTS, also referred to as whole cell-based or phenotypic screening, utilizes intact living cells to identify compounds that produce a desired phenotypic response, such as bacterial cell death or inhibition of viral replication, without prior knowledge of the specific molecular target [44] [45].
Key Characteristics:
Definition and Principle: MT-HTS employs isolated molecular targets â typically purified proteins, enzymes, or nucleic acids â to identify compounds that interact with these specific biomolecules through binding or functional modulation [44] [45].
Key Characteristics:
Table 1: Comparative Analysis of Cellular vs. Molecular Target HTS Approaches
| Parameter | Cellular Target HTS (CT-HTS) | Molecular Target HTS (MT-HTS) |
|---|---|---|
| Screening System | Whole living cells (bacterial, fungal, mammalian) | Purified proteins, enzymes, or nucleic acids |
| Target Knowledge | Not required; phenotypic outcome driven | Required prior to screening |
| Physiological Context | Full physiological context maintained | Minimal to no physiological context |
| Hit Rate for NPs | ~0.3% (with polyketides) [44] | Variable; typically lower than CT-HTS |
| Primary Advantage | Identifies compounds with cellular activity | Reveals specific molecular mechanisms |
| Primary Challenge | Target deconvolution required | May not translate to cellular activity |
| Secondary Screening | Eliminate non-specific cytotoxics | Eliminate PAINS and promiscuous binders |
A hybrid approach has emerged that combines advantages of both CT-HTS and MT-HTS through mechanism-informed phenotypic screening, most commonly implemented as reporter gene assays [44]. These assays utilize cells engineered with reporter constructs that produce measurable signals (e.g., luminescence, fluorescence) when specific pathways of interest are modulated. For example, the ATAD5-luciferase HTS assay identifies genotoxic compounds by exploiting the stabilization of ATAD5 protein following DNA damage [46]. This approach maintains physiological context while providing information about the signaling pathways with which hits interact, effectively bridging the gap between purely phenotypic and purely target-based screening [44].
Other innovative reporter systems include:
Diagram 1: HTS Approaches for NP Library Screening. This workflow illustrates the three main strategies for screening natural product libraries, with their respective advantages and limitations.
Objective: Identify natural product extracts that inhibit growth of pathogenic bacterial strains.
Materials and Reagents:
Procedure:
Cell Seeding and Compound Exposure:
Incubation and Signal Detection:
Data Analysis:
Objective: Identify natural product extracts that inhibit specific bacterial enzyme targets (e.g., DNA gyrase, topoisomerase, transpeptidases).
Materials and Reagents:
Procedure:
Screening Reaction Setup:
Reaction Initiation and Detection:
Data Analysis:
Table 2: Key Research Reagent Solutions for NP HTS Campaigns
| Reagent Category | Specific Examples | Function in HTS | Application Notes |
|---|---|---|---|
| Detection Systems | Resazurin, ATP-lite, GFP reporters | Cell viability and metabolic activity assessment | Resazurin preferred for bacterial screens due to linear range [46] |
| Reporters | Luciferase, β-lactamase, fluorescent proteins | Pathway-specific reporter gene assays | ATAD5-luciferase for genotoxicity screening [46] |
| Cellular Systems | ESKAPE pathogens, DT40 cell lines, primary cells | Physiological context for screening | DNA-repair-deficient DT40 lines for genotoxin screening [46] |
| Molecular Targets | Purified enzymes, protein-protein interactions | Target-specific screening | Fluorescence anisotropy for lipid II binding proteins [44] |
| Automation Tools | Liquid handlers, plate stackers, detectors | Enable high-throughput processing | Robotic systems can test >100,000 compounds daily [47] |
The successful implementation of HTS for natural product libraries requires careful attention to workflow design and quality control measures throughout the process.
Diagram 2: HTS Workflow with Quality Control Gates. The screening process incorporates quality control checkpoints at each stage to ensure identification of high-quality hits from natural product libraries.
Assay Robustness Metrics:
Hit Selection Criteria:
The strategic selection between cellular and molecular target approaches for HTS of natural product libraries represents a fundamental decision point in drug discovery. CT-HTS offers the advantage of physiological relevance and identification of cellularly active compounds, while MT-HTS provides mechanistic clarity and target engagement information. The emerging paradigm of mechanism-informed phenotypic screening bridges these approaches, offering both physiological context and pathway-specific information. As natural products continue to play a crucial role in addressing the antibiotic resistance crisis and other therapeutic challenges, the intelligent application of these HTS methodologies â coupled with robust quality control and hit validation protocols â will maximize the potential of exploring natural product chemical space for drug discovery research. Future directions will likely see increased integration of artificial intelligence, advanced bioinformatics, and innovative screening technologies to further enhance the efficiency and success of NP-based drug discovery campaigns.
The declining efficiency of purely target-based drug discovery has catalyzed a resurgence in phenotypic screening. However, the limited translatability of simple phenotypic observations has necessitated an evolution in strategy. This whitepaper details the paradigm of Mechanism-Informed Phenotypic Drug Discovery (MIPDD), a hybrid approach that integrates the physiological relevance of phenotypic observation with molecular-level mechanistic insight. Framed within the critical context of exploring Natural Product (NP) chemical space, we demonstrate how MIPDD leverages the unique physicochemical properties of NPs to identify novel therapeutic leads. This technical guide provides a comprehensive overview of the conceptual foundation, experimental methodologies, and practical implementation of MIPDD, with a specific focus on its application in antiviral and anticancer drug discovery.
Modern drug discovery has been dominated by target-based approaches, but their high attrition rates have prompted a critical re-evaluation. Analysis of cancer drug origins reveals that while the majority of approved small-molecule drugs originated from target-based discovery, very few were discovered entirely by 'classical' phenotypic screening [48]. This highlights a fundamental challenge: traditional phenotypic screens, often reliant on nonspecific readouts like cytotoxicity, are insufficient for identifying drugs with novel, therapeutically translatable mechanisms [48].
This realization has spurred the development of Mechanism-Informed Phenotypic Drug Discovery (MIPDD), defined as the use of phenotypic assays designed around specific molecular pathways and targets, employing disease-relevant cellular models [48]. MIPDD aims to determine the causal relationships between target inhibition and phenotypic effects, opening new avenues for understanding cancer biology and discovering drugs with optimal molecular mechanisms of action [48].
Concurrently, the exploration of biologically relevant chemical space (BioReCS) has identified natural products as occupants of unique regions not represented by synthetic medicinal chemistry compounds [49] [50] [24]. Their structural rigidity, lower aromaticity, and high degree of stereochemistry make them exceptional starting points for MIPDD campaigns, providing a strategic advantage in identifying first-in-class therapeutics [49].
Table 1: Comparison of Drug Discovery Approaches
| Feature | Classical Phenotypic Screening | Target-Based Screening | Mechanism-Informed Phenotypic Screening (MIPDD) |
|---|---|---|---|
| Primary Focus | Observable phenotypic change without prior target knowledge [51] | Modulation of a predefined molecular target [51] | Phenotypic change informed by underlying molecular mechanisms [48] |
| Discovery Bias | Unbiased, allows novel target identification [51] | Hypothesis-driven, limited to known pathways [51] | Hypothesis-guided, informed by disease biology |
| Mechanism of Action | Often unknown initially, requires deconvolution [51] | Defined from the outset [51] | Informed by pathway context, facilitating deconvolution |
| Chemical Library Strategy | Diverse libraries, emphasis on natural products [52] [49] | Focused libraries for specific target classes | Biased diversity, leveraging NP chemical space for specific phenotypes [52] |
Mechanism-informed phenotypic screening represents a neoclassic strategy that merges the best attributes of phenotypic and target-based approaches. Its core principle is the use of mechanistically defined cellular models for therapeutically translatable cancer phenotypes [48]. This involves:
Natural products are a critical component for populating MIPDD screening libraries. Computational analyses using tools like ChemGPS-NP have demonstrated that NPs and synthetic bioactive compounds "differ notably in coverage of chemical space" [49] [50]. Key characteristics of NP chemical space include:
Table 2: Key Characteristics of Natural Products in Chemical Space
| Property | Finding | Implication for MIPDD |
|---|---|---|
| Rule of Five Compliance | ~60% of unique NPs have no Ro5 violations; NP-derived drugs are equally split between Ro5 compliant and violators [49] | NPs are a valuable source for both conventional oral drugs and "beyond Rule of 5" therapeutics |
| Chemical Space Coverage | NPs cover regions not represented by synthetic medicinal chemistry databases (e.g., WOMBAT) [49] [50] | Provides access to novel scaffolds and mechanisms not found in standard synthetic libraries |
| Structural Features | NPs are less flexible and contain fewer aromatic rings than synthetic medicinal chemistry compounds [49] | Favors binding to challenging target classes like protein-protein interactions |
The following workflow, adapted from a study on human coronavirus 229E, exemplifies a modern MIPDD approach for identifying host-targeting antivirals [54].
Protocol Details:
In cancer drug discovery, MIPDD moves beyond simple proliferation assays. Key strategies include [48]:
Successful implementation of MIPDD relies on a suite of specialized reagents and tools. The following table details key components for establishing these assays.
Table 3: Essential Research Reagents for MIPDD Assays
| Reagent / Solution | Function / Purpose | Application Example |
|---|---|---|
| Cell Painting Dye Set | Multiplexed fluorescent staining for capturing a wide spectrum of morphological features [54] | General phenomic profiling; antiviral phenomics [54] |
| Anti-Viral Nucleoprotein Antibody | Specific detection of virus-infected cells at a single-cell level within a phenotypic assay [54] | Classifying infection status in host-cell morphological profiling [54] |
| 3D Organoid / Spheroid Cultures | Physiologically relevant models that better mimic tissue architecture and disease context [51] | Oncology screening using more predictive in vitro models [48] [51] |
| iPSC-Derived Cell Models | Patient-specific disease modeling and drug screening for complex diseases [51] | Neurological disease modeling, personalized medicine applications |
| ChemGPS-NP / Scaffold Hunter | Computational tools for navigating and analyzing natural product chemical space [24] | Guiding the selection of NP-enriched screening libraries [49] [24] |
| High-Content Imaging System | Automated microscopy and image analysis for quantitative multiparametric data extraction | Essential for all image-based phenotypic screening workflows [51] [54] |
| Tinoridine | Tinoridine, CAS:24237-54-5, MF:C17H20N2O2S, MW:316.4 g/mol | Chemical Reagent |
| 5-Fluoroindole | 5-Fluoroindole, CAS:399-52-0, MF:C8H6FN, MW:135.14 g/mol | Chemical Reagent |
The MIPDD approach is highly applicable to antimicrobial discovery, particularly in targeting virulence mechanisms. This shifts the focus from essential pathogen viability to disarming its ability to cause disease, a strategy that may impose less selective pressure for resistance.
In antimalarial research, this has translated to developing robust phenotypic screens against diverse lifecycle stages beyond the symptomatic asexual blood stage, such as exoerythrocytic stages and transmission-blocking gametocytes [53]. The core logic of this approach is mapped below:
This strategy has been successfully implemented to discover compounds like the spiroindolone KAE609, which targets P-type cation-transporter ATPase4 (PfATP4) and demonstrates rapid parasiticidal activity [53].
Mechanism-Informed Phenotypic Drug Discovery represents a powerful and necessary evolution in the search for novel therapeutics. By integrating the physiological relevance of phenotypic observation with growing molecular understanding of disease pathways, MIPDD increases the probability of identifying high-quality leads with novel mechanisms of action. The strategic integration of this approach with the systematic exploration of natural product chemical space creates a synergistic partnership. NPs provide a source of unique, pre-validated scaffolds that populate biologically relevant but otherwise sparsely occupied regions of chemical space, while MIPDD offers a sophisticated framework to effectively probe and decode their complex biological activities. As technological advances in high-content imaging, automated image analysis, and biologically complex model systems continue, the implementation and success of MIPDD are poised to expand, firmly establishing its role in the future of drug discovery.
The relentless pursuit of new therapeutic compounds has driven researchers to delve into the vast chemical space of natural products, which have served as a cornerstone for drug development for decades. Plant natural products, or specialized metabolites, play a vital role in this endeavor, with many clinically important drugs such as the anticancer agents topotecan (derived from camptothecin) and etoposide (derived from podophyllotoxin) originating from plant sources [55]. Historically, the discovery of these compounds relied on bioactivity-guided fractionation approaches, which are increasingly hampered by the high rate of compound re-discovery [56]. The natural products discovery field has therefore begun a decisive shift away from these traditional methods toward strategies that capitalize on large-scale -omics technologies [56].
This transformation is powered by the integration of genomics and metabolomics, which provides a comprehensive framework for elucidating biosynthetic pathways. Genomics reveals the blueprint of an organism's biosynthetic potential, while metabolomics captures the chemical expression of that potential under specific conditions [56]. The convergence of these disciplines generates vast datasets, and the application of advanced computational tools, machine learning, and data analytics has become crucial for processing and interpreting this information to uncover intricate regulatory networks and identify key components of biosynthetic pathways [55]. This in-depth technical guide explores how the power of genomics and metabolomics is being harnessed to unlock biosynthetic pathways, framing these advancements within the critical context of exploring natural product chemical space for modern drug discovery research.
Genomics provides the foundational blueprint for biosynthetic pathway discovery by enabling the identification and annotation of Biosynthetic Gene Clusters (BGCs)âgenomic loci that co-localize genes encoding the enzymes responsible for producing a specialized metabolite. The first step in genomic exploration involves obtaining high-quality sequence data. While Illumina next-generation sequencing (NGS) offers high fidelity and low cost, its short reads can result in fragmented assemblies. Advanced single-molecule sequencing technologies like Pacific Biosciences (PacBio) and Oxford Nanopore generate longer reads, which are invaluable for assembling complete BGCs, despite their higher per-base error rates [56].
Once a genome is sequenced and assembled, the critical task of BGC identification begins. This is accomplished using sophisticated bioinformatic algorithms that scan genomic data for signature biosynthetic genes. Several tools have been developed for this purpose, each with distinct strengths and applications [56].
Table 1: Key Computational Tools for Biosynthetic Gene Cluster Identification
| Tool Name | Primary Application | Methodology Overview |
|---|---|---|
| antiSMASH [56] | Bacteria, Fungi, Plants | Uses a library of profile Hidden Markov Models (pHMMs) to detect >50 classes of BGCs; widely considered a industry standard. |
| plantiSMASH [57] | Plants | A specialized derivative of antiSMASH using modified rules tailored to plant genomes. |
| PRISM [56] | Bacteria & Fungi | Employs pHMMs and machine learning to predict BGCs and the chemical structures of their products. |
| SMURF [56] | Fungi | pHMM-based tool designed for fungal genome mining. |
| CO-OCCUR [56] | Phylogenetically diverse fungi | Identifies accessory biosynthetic genes through frequency and co-occurrence analysis around core genes, complementing other tools. |
These tools function by identifying core biosynthetic genes, such as those for polyketide synthases (PKSs) and non-ribosomal peptide synthetases (NRPSs), and then scanning the surrounding genomic region for additional genes encoding tailoring enzymes, transporters, and regulators [56]. The output is a map of an organism's biosynthetic potential, which often reveals a surprising abundance of uncharacterized BGCs, even in well-studied organisms [56]. This highlights the vastness of unexplored natural product chemical space and provides a genetic starting point for discovery.
1. DNA Extraction and Sequencing:
2. Genome Assembly and Annotation:
3. BGC Identification and Analysis:
Diagram 1: Genomic workflow for BGC identification.
Metabolomics delivers the complementary chemical phenotype by providing a snapshot of the entire set of metabolites in a biological system at a given time. In the context of natural product research, it focuses on the secondary metabolites actually produced by the organism, offering a direct readout of biosynthetic pathway activity [56]. The workflow is typically divided into pre-analytical, analytical, and post-analytical stages, with careful standardization at each phase being critical for robust and reproducible results [58].
The pre-analytical phase involves sample collection, handling, and storage. Factors such as collection tubes, centrifugation steps, freeze-thaw cycles, and storage conditions must be standardized using Standardized Operating Procedures (SOPs) to minimize variability and ensure data accurately reflects endogenous metabolite levels [58]. For MS-based metabolomics, sample preparation involves extracting metabolites from proteins and other matrix components, a process that should be automated where possible to reduce human error [58].
The analytical heart of modern metabolomics is mass spectrometry (MS), often coupled with separation techniques like liquid chromatography (LC) or gas chromatography (GC). MS is favored for its high sensitivity and specificity, allowing for the measurement of thousands of metabolites in small sample volumes [58]. Two primary analytical approaches are employed:
Table 2: Core Metabolomics Instrumentation and Reagents
| Category / Item | Function / Description | Application in Pathway Elucidation |
|---|---|---|
| LC-MS / GC-MS System | Separates (chromatography) and detects (mass spectrometry) complex metabolite mixtures. | Workhorse platform for profiling metabolite extracts; enables detection of thousands of features. |
| Biocrates AbsoluteIDQ p180 Kit [59] | Standardized kit for targeted quantification of 188 plasma metabolites. | Provides highly reproducible quantitative data for defined metabolite classes; used for biomarker studies. |
| High-Resolution MS (Orbitrap, FTICR-MS) [57] | Mass spectrometers with very high mass accuracy and resolution. | Critical for determining precise molecular formulae of unknown metabolites from untargeted data. |
| Nuclear Magnetic Resonance (NMR) [58] | Non-destructive, quantitative analytical technique. | Useful for structural elucidation and quantifying abundant metabolites; complements MS data. |
| Ion Mobility Spectrometry [58] | Separates ions based on their size, shape, and charge. | Adds an additional separation dimension, helping to resolve structurally similar isomers. |
1. Sample Preparation and Extraction:
2. LC-MS Data Acquisition:
3. Data Pre-processing and Metabolite Annotation:
Diagram 2: Metabolomics workflow for chemical phenotyping.
The true power of -omics approaches is realized when genomics and metabolomics datasets are integrated, moving beyond correlation to predict causal relationships within biosynthetic pathways. This integration allows researchers to simultaneously identify expressed secondary metabolites and link them to their biosynthetic machinery [56]. One of the primary strategies for integration is co-expression analysis, which identifies genes and metabolites that show correlated abundance patterns across different samples (e.g., different tissues, treatments, or time points) [57]. A gene whose expression profile closely mirrors the accumulation of a specific metabolite is a strong candidate for encoding an enzyme involved in that metabolite's biosynthesis.
Cutting-edge computational tools are now automating this integration process. A leading example is MEANtools, a systematic and unsupervised computational workflow that predicts candidate metabolic pathways de novo [57]. MEANtools integrates mass features from metabolomics data and transcripts from transcriptomics data. It uses a mutual rank-based correlation method to identify highly correlated metabolite-transcript pairs and then leverages known biochemical reaction rules from databases like RetroRules to assess whether correlated transcripts encode enzymes that can catalyze reactions between correlated metabolites [57]. This allows the pipeline to construct putative biosynthetic pathways from the integrated data, generating testable hypotheses.
Another powerful integration method is the Genome-Wide Association Study (GWAS) of metabolites, known as mQTL (metabolite Quantitative Trait Loci) mapping. This approach identifies genomic regions associated with natural variation in metabolite levels [59]. In a study on pigs, mQTL mapping successfully identified 97 genomic loci associated with the levels of 126 metabolites, directly linking genetic variants to metabolic phenotypes and uncovering genes involved in specific metabolic pathways [59].
1. Paired Sample Collection and Multi-Omics Data Generation:
2. Data Integration and Pathway Prediction with MEANtools:
3. Hypothesis Testing and Validation:
Diagram 3: Multi-omics integration for pathway prediction.
The application of integrated genomics and metabolomics is revolutionizing the drug discovery and development pipeline, offering powerful approaches from initial target identification to the realization of precision medicine.
In the target identification phase, metabolomics can reveal specific metabolic pathways that are altered in disease states. For instance, an unbiased discovery metabolomics approach can characterize the molecular heterogeneity of complex diseases like type 2 diabetes mellitus (T2DM), identifying distinct patient subtypes with different underlying metabolic disturbances [58]. This can pinpoint specific enzymes or metabolic regulators as novel therapeutic targets. Genomics complements this by identifying genetic variants associated with both disease risk and metabolite levels (mQTLs), providing orthogonal evidence for a target's validity and highlighting potential mechanisms of action [59].
In natural product-based drug discovery, the integration of -omics technologies directly addresses the challenge of efficiently linking bioactive compounds to their BGCs. By combining metabolomic profiling with genomic mining, researchers can prioritize uncharacterized BGCs that are active under specific conditions and associated with the production of novel chemical scaffolds [55] [56]. This strategy efficiently guides the isolation and characterization of new lead compounds from the vast "dark matter" of uncharacterized natural product space.
Finally, metabolomics plays a crucial role in biomarker discovery for patient stratification and treatment response predictionâa cornerstone of precision medicine. The identification of genetically influenced metabolites (GIMs) provides a powerful class of biomarkers. As demonstrated in pig models, these stable molecular phenotypes are highly heritable and can be used to dissect complex traits [59]. In humans, metabolomic signatures can define individual "metabotypes," enabling the stratification of patient populations to predict drug response and optimize therapeutic outcomes [58]. This ensures that the right natural product-derived drug or other therapy is delivered to the right patient.
Natural Products (NPs) represent an indispensable source of chemical diversity for drug discovery, providing greater structural variety than standard synthetic chemistry and unique opportunities for identifying novel low molecular weight lead compounds [60]. A detailed analysis of FDA-approved drugs between 1981 and 2019 reveals that natural products, their direct derivatives, or synthetic drugs incorporating pharmacophoric groups of active secondary metabolites constitute approximately 56.1% of all approved drugs, with particularly significant contributions to anticancer (69.6%), antibacterial (58%), and antiviral (37.6%) therapies [60]. This remarkable success stems from the evolutionary optimization of NPs for biological interaction, often resulting in complex three-dimensional structures rich in sp³-hybridized carbon atoms and stereocenters that cover chemical space regions largely inaccessible to purely synthetic compounds [61]. The screening of natural product libraries therefore offers significant advantages for finding novel therapeutic agents, but realizing this potential requires meticulous attention to library construction, curation, and management. This technical guide outlines comprehensive methodologies for building and maintaining high-quality NP libraries specifically framed within the context of systematically exploring natural product chemical space for drug discovery research.
Modern natural product libraries encompass diverse physical forms and structural types, each with distinct advantages for screening campaigns. These libraries typically include crude extracts from plants, marine organisms, and microorganisms; prefractionated extracts that reduce complexity while preserving synergistic interactions; and pure natural product compounds [62] [60]. A particularly promising approach involves the deconstruction of NPs into fragments and their recombination into unprecedented pseudo-natural product frameworks, which retain NP-inspired features while extending into novel structural and functional space [63]. When designing a library, careful consideration must be given to the balance between complexity and screening compatibility. Crude extracts offer the fullest representation of natural chemical diversity but may present challenges in dereplication and identification of active constituents, while pure compounds provide immediate structural information but require significant upfront investment in isolation and characterization.
Table 1: Composition of Representative Natural Product Libraries
| Library/Source | Type | Scale/Size | Key Characteristics | References |
|---|---|---|---|---|
| COCONUT 2.0 | Database (Virtual) | 695,133 distinct structures | Comprehensive collection of open natural products; extensive chemical space coverage | [61] |
| LANaPDB | Database (Virtual) | 13,578 compounds | Focus on Latin American biodiversity; non-duplicate natural products | [61] |
| MEDINA | Physical Library | >200,000 extracts | Microbial-derived natural products from diverse global environments | [62] |
| NCI Natural Products Repository | Physical Library | 230,000+ crude extracts; 400+ purified compounds | One of world's most comprehensive collections; includes traditional Chinese medicine extracts | [62] |
| University of Michigan Natural Products Discovery Core | Physical Library | 45,000+ natural product extracts (NPEs) | Metadata-enabled with chemical and genetic profiles; HTS-formatted | [62] |
| NatureBank, Griffith University | Physical Library | 18,000+ extracts; 90,000+ fractions; 100+ pure compounds | Australian biodiversity focus; lead-like enhanced libraries | [62] |
Fragment-based approaches have emerged as powerful tools for efficiently exploring NP chemical space. Recent research has generated comprehensive fragment libraries from large NP databases, with 2,583,127 fragments derived from COCONUT and 74,193 fragments from LANaPDB [61]. These fragments, typically obtained using methods like the Retrosynthetic Combinatorial Analysis Procedure (RECAP), adhere to the "rule of three" (RO3) for fragment-based drug design: molecular weight â¤300 Da, rotatable bonds â¤3, topological polar surface area â¤60 à ², LogP â¤3, hydrogen-bond acceptors â¤3, and hydrogen-bond donors â¤3 [61]. Analysis reveals that only 1.5% of COCONUT fragments and 2.5% of LANaPDB fragments fulfill all RO3 criteria, highlighting both the structural complexity of natural products and the need for careful curation to optimize fragment libraries for screening [61]. When compared to synthetic fragment libraries, NP-derived fragments occupy distinct regions of chemical space, often exhibiting greater stereochemical complexity and scaffold diversity that can provide unique starting points for drug discovery programs focused on challenging therapeutic targets.
Table 2: Performance Metrics of Natural Product Fragment Libraries
| Library | Initial Fragments | Fragments After Standardization | Fragments Fulfilling RO3 | Percentage Fulfilling RO3 |
|---|---|---|---|---|
| COCONUT | 2,583,127 | 2,583,127 | 38,747 | 1.5% |
| LANaPDB | 74,193 | 74,193 | 1,832 | 2.5% |
| CRAFT | 1,214 | 1,202 | 176 | 14.6% |
| Enamine | 12,505 | 12,496 | 8,386 | 67.1% |
| ChemDiv | 74,721 | 72,356 | 16,723 | 23.1% |
The access and use of biological resources for natural product library development must comply with international and national regulations governing genetic resources and associated traditional knowledge. The United Nations 1992 Convention on Biological Diversity (CBD) and its supplementary Nagoya Protocol on Access to Genetic Resources and the Fair and Equitable Sharing of Benefits (ABS) establish the legal framework requiring mutually agreed terms between source countries and users [60]. These agreements typically include provisions for prior informed consent, benefit-sharing arrangements, and respect for the rights of indigenous communities and traditional knowledge holders. In Brazil, for example, Law 13.123/15 and the National System for the Management of Genetic Resources and Associated Traditional Knowledge (SisGen) regulate research and development involving Brazilian biodiversity, requiring registration of activities and establishing that foreign researchers must collaborate with Brazilian institutions [60]. Similar frameworks exist in other biodiverse countries, creating a complex regulatory landscape that necessitates careful legal assessment during library planning. Negotiating appropriate access and benefit-sharing agreements can be time-consuming but represents an essential ethical and legal foundation for sustainable natural product research that respects national sovereignty and contributes to conservation efforts.
The construction of high-quality natural product libraries begins with meticulous sample preparation, which directly influences phytochemical composition and screening outcomes. For plant-based libraries, critical considerations include proper taxonomic identification by qualified botanists with voucher specimen deposition in recognized herbaria, optimal collection timing that accounts for seasonal and diurnal variation in metabolite production, and appropriate preservation methods such as freeze-drying or controlled drying to prevent degradation [60]. Extraction strategies should be designed to maximize chemical diversity while maintaining compatibility with screening platforms, typically employing a sequential approach with solvents of increasing polarity (e.g., hexane, dichloromethane, ethyl acetate, methanol, water) [60]. For microorganism-derived libraries, specialized isolation media and cultivation conditions are essential to access the full biosynthetic potential, as standard laboratory conditions may not activate silent gene clusters responsible for producing many bioactive metabolites. Advanced techniques such as co-cultivation, OSMAC (one strain many compounds) approaches, and genomic mining can significantly enhance chemical diversity from microbial sources.
Standardized formatting and rigorous quality control are essential for generating reproducible screening data from natural product libraries. Most modern libraries are formatted in 96-well or 384-well plates compatible with high-throughput screening robotics, with typical concentrations of 1-10 mg/mL for extracts and 1-10 mM for pure compounds in dimethyl sulfoxide (DMSO) [62]. Quality control measures should include chemical profiling using HPLC-UV/PDA/ELSD and/or LC-MS to verify composition and stability, determination of dry weight for extract normalization, and assessment of potential interferants such as tannins, pigments, or non-specific binding compounds that may produce false positives in certain assay formats [60]. For pure compound libraries, purity assessment (typically â¥95% by HPLC) and structural confirmation (via NMR and HRMS) are essential, along with curation of associated metadata including natural source, isolation method, physicochemical properties, and known biological activities [62]. The ChromaDex Natural Compound Libraries exemplify this approach, offering extensively characterized fractions that preserve cross-fraction synergy while providing detailed compositional data [62].
Affinity selection mass spectrometry has emerged as a powerful label-free biophysical method for identifying ligands from complex natural product libraries against various biological targets, including soluble proteins, membrane proteins, nucleic acids, and nucleic acid-protein complexes [64]. AS-MS interrogates non-covalent target-ligand complexes in a non-functional assay, simultaneously identifying multiple ligands with different mechanisms of action, including orthosteric and allosteric binders [64]. The methodology involves four critical stages: (1) static incubation of the target with the natural product library, typically with the target in molar excess to avoid competition effects; (2) separation of target-ligand complexes from unbound components; (3) dissociation of ligands from the complexes; and (4) identification of ligands by mass spectrometry [64]. This approach offers significant advantages over traditional bioactivity-guided fractionation by reducing false positives and avoiding activity loss through multiple fractionation steps.
Ultrafiltration represents a particularly effective solution-based AS-MS technique that separates target-ligand complexes from unbound molecules based on size exclusion through specialized membranes with controlled porosity [64]. In a typical implementation, the target protein is incubated with the natural product library at low micromolar concentrations optimal for detecting high-affinity ligands with specific binding interactions. Following equilibrium establishment, ultrafiltration membranes with molecular weight cutoffs between 500-500,000 Da retain the larger ligand-protein complexes while allowing unbound molecules to pass through [64]. Ligands are subsequently dissociated using denaturing conditions such as methanol or acetonitrile with volatile organic acids (e.g., formic acid), maintaining compatibility with subsequent LC-MS analysis. This approach has been successfully applied to identify bioactive natural products, such as the discovery of botulin, lanosterol, and quercetin as 5-lipoxygenase ligands from Inonotus obliquus extracts [64]. The methodology offers advantages in maintaining native protein conformation during screening and applicability to diverse target classes, though careful optimization of filtration conditions is necessary to prevent membrane fouling or non-specific binding.
Table 3: Key Research Reagent Solutions for Natural Product Library Screening
| Reagent/Resource | Function/Application | Implementation Example | Considerations | |
|---|---|---|---|---|
| Ultrafiltration Membranes | Separation of target-ligand complexes from unbound molecules | Molecular weight cutoffs 500-500,000 Da for protein-ligand complex retention | Pore size uniformity, minimal non-specific binding, chemical compatibility | [64] |
| Immobilization Supports | Ligand fishing with immobilized targets | Magnetic microbeads (MagMASS), chromatographic resins | Retention of target activity after immobilization, ligand accessibility | [64] |
| Mass Spectrometry Platforms | Detection and identification of bound ligands | LC-MS systems with high resolution and mass accuracy | Sensitivity, dynamic range, compatibility with dissociation solvents | [64] |
| Bioaffinity Chromatography Systems | Zonal or frontal chromatography for ligand disclosure | Solid-supported proteins for "functional chromatography" | Retention time precision, breakthrough curve analysis | [64] |
| Natural Product Libraries | Sources of diverse 3D molecular features for screening | Commercial sources (e.g., ChromaDex, MicroSource) or custom collections | Chemical diversity, annotation quality, regulatory compliance | [62] [60] |
| Fragment Libraries | Fragment-based drug design starting points | COCONUT, LANaPDB, CRAFT, commercial vendors | Rule of three compliance, synthetic accessibility, structural diversity | [61] |
| N-Acetyl-D-cysteine | N-Acetyl-D-cysteine, CAS:26117-28-2, MF:C5H9NO3S, MW:163.20 g/mol | Chemical Reagent | Bench Chemicals | |
| Palmitoyl 3-carbacyclic Phosphatidic Acid | Palmitoyl 3-carbacyclic Phosphatidic Acid, CAS:476310-22-2, MF:C20H39O5P, MW:390.5 g/mol | Chemical Reagent | Bench Chemicals |
Building and curating high-quality natural product libraries for drug discovery represents both a significant technical challenge and a substantial opportunity to access unique chemical space with proven therapeutic relevance. Success in this endeavor requires integrated expertise spanning taxonomy, natural product chemistry, analytical methodology, screening technology, and regulatory compliance. The future of natural product-based drug discovery will increasingly leverage computational approaches for virtual screening, chemical space analysis, and bioactivity prediction, while advanced screening technologies like AS-MS enhance the efficiency of ligand identification from complex mixtures. Furthermore, innovative strategies such as pseudo-natural product design, which recombines biosynthetically unrelated NP fragments into novel scaffolds, promise to extend accessible chemical space beyond naturally occurring structures while retaining desirable NP-like properties [63]. By applying the systematic approaches outlined in this guideâfrom ethical sourcing and standardized library construction to advanced screening methodologiesâresearch institutions and pharmaceutical companies can fully leverage the remarkable structural and functional diversity of natural products to address unmet medical needs through novel therapeutic agents.
The exploration of natural products (NPs) represents a cornerstone of drug discovery, with a significant portion of modern small-molecule drugs originating from or being inspired by natural compounds [65]. However, the traditional drug discovery pipeline is notoriously protracted, often exceeding 12 years and costing more than $1.8 billion USD on average [66]. The vastness of biologically relevant chemical space, coupled with challenges in sourcing and characterizing NPs, necessitates more efficient discovery approaches [49].
In silico technologies have emerged as powerful tools to address these challenges. Computer-Aided Drug Discovery (CADD) leverages computational power to streamline hit identification and optimization, dramatically reducing the time and cost associated with early-stage discovery [67] [66]. This technical guide details the core methodologies of virtual screening (VS) and molecular dynamics (MD) simulations, framing them within the strategic imperative to efficiently navigate the unique and biologically relevant chemical space occupied by natural products [68] [49].
Virtual screening is a computational technique for identifying potential hit compounds from vast digital libraries. Its application is particularly valuable for exploring NPs, which are often structurally complex and sparsely represented in conventional screening collections [49].
Natural products are pre-validated by nature, possessing evolutionary optimization for interaction with biological macromolecules. Analyses reveal that NPs occupy distinct regions of chemical space compared to synthetic medicinal chemistry compounds. They are typically more structurally rigid and possess a lower degree of aromaticity, offering access to novel scaffolds that can circumvent pre-existing intellectual property and overcome the limitations of conventional chemical libraries [49]. It is estimated that about two-thirds of modern small-molecule drugs are related to natural compounds [65].
Two primary VS approaches are employed, often in tandem, for efficient hit identification.
Ligand-Based Virtual Screening (LBVS): This method is used when the 3D structure of the target protein is unknown but a set of active ligands is available. It relies on comparing molecular descriptors or fingerprints to identify new compounds with similar properties [69]. Advanced LBVS platforms, such as the BIOPTIC B1 system, utilize transformer-based models trained on massive molecular datasets (e.g., 160 million molecules) to create potency-aware embeddings. These enable ultra-high-throughput screening of multi-billion compound libraries in mere minutes, demonstrating success in prospective campaigns for targets like LRRK2 for Parkinson's disease, yielding novel sub-micromolar binders (Kd = 110 nM) [69].
Structure-Based Virtual Screening (SBVS): This approach requires a 3D structure of the biological target, typically derived from X-ray crystallography, NMR, or homology modeling. The primary technique used in SBVS is molecular docking, which predicts the preferred orientation and binding affinity (scoring) of a small molecule within a target's binding site [67] [65]. Docking simulations help understand molecular-level interactions and are crucial for identifying hit leads from natural product libraries [67].
Table 1: Key Databases for Virtual Screening of Natural Products
| Database Name | Description | Content Focus | Utility in VS |
|---|---|---|---|
| ZINC15 [70] | Curated database of commercially available compounds. | 100+ million compounds in ready-to-dock, 3D formats. | Primary source for purchasable screening compounds. |
| ChEMBL [70] [71] | Manually curated database of bioactive molecules. | Drug-like molecules with bioactivity data. | Ligand-based screening and model training. |
| PubChem [70] | NCBI repository of chemical molecules and bioactivities. | Massive collection of compounds and bioassay results. | Similarity searching (2D/3D) and bioactivity data. |
| Dictionary of Natural Products (DNP) [70] [49] | Comprehensive and fully-edited NP database. | Over 170,000 compounds of natural origin. | Definitive source for natural product structures and data. |
| TCM Database [70] | Database on Traditional Chinese Medicine. | ~170,000 compounds with 2D/3D structural files. | Virtual screening of natural product libraries. |
The following diagram illustrates a synergistic VS workflow that integrates both ligand-based and structure-based methods to efficiently mine natural product chemical space.
While molecular docking provides a static snapshot of binding, Molecular Dynamics (MD) simulations model the dynamic behavior of molecules over time, offering critical insights into stability, conformational changes, and binding mechanisms that are essential for hit-to-lead optimization [65].
MD simulations numerically integrate Newton's second law of motion for all atoms in a system, typically involving a protein-ligand complex solvated in water. This allows researchers to observe the time-dependent evolution of the molecular system [72]. Key parameters must be carefully set to ensure simulation stability and accuracy:
A typical MD workflow for validating a natural product hit bound to its target protein involves the following steps [65] [72]:
Table 2: Critical MD Parameters and Recommended Settings for Stability
| Parameter | Description | Recommended Settings | Rationale |
|---|---|---|---|
| Time Step [72] | Interval for numerical integration. | 1-2 fs (systems with H-bonds); 5 fs (metallic systems). | Prevents instability; ensures energy conservation. |
| Force Field [73] | Mathematical functions for atomic interactions. | COMPASS II, CHARMM, AMBER, OPLS. | Determines accuracy of interatomic forces. |
| Temperature Control [72] | Thermostat for NVT/NpT ensembles. | Langevin, Nosé-Hoover, Bussi. | Correctly samples the canonical ensemble. |
| Pressure Control [72] | Barostat for NpT ensemble. | Berendsen, Parrinello-Rahman. | Maintains correct system density. |
| Simulation Length [65] | Duration of production run. | 50 - 500 ns (varies by system). | Captures relevant biological dynamics and stability. |
| Periodic Boundary Conditions (PBC) [72] | Mimics an infinite system. | Cubic or rhombic dodecahedron box. | Eliminates edge effects; uses a finite number of atoms. |
The following diagram outlines the sequential steps involved in setting up and running an MD simulation for a protein-ligand complex.
Successful implementation of in silico drug discovery relies on a suite of specialized software tools and databases.
Table 3: Essential In Silico Research Tools for Natural Product Drug Discovery
| Tool Category | Representative Examples | Key Function | Application in NP Discovery |
|---|---|---|---|
| Molecular Docking Software [67] [65] | AutoDock Vina, GOLD, Glide | Predicts binding pose and affinity of ligands. | Structure-based screening of NP libraries against therapeutic targets. |
| MD Simulation Engines [72] [73] | ASE (Atomic Simulation Environment), GROMACS, AMBER, NAMD | Performs molecular dynamics simulations. | Validating binding stability and mechanism of NP hits. |
| Cheminformatics & ML Libraries [71] | RDKit, Open Babel, scikit-learn | Handles chemical data and builds ML models. | Processing NP structures; building LBVS models with ECFP fingerprints. |
| Homology Modeling Tools [65] | MODELLER, SWISS-MODEL | Predicts 3D protein structures from amino acid sequences. | Generating target models for SBVS when experimental structures are unavailable. |
| Bioactivity Databases [70] [71] | ChEMBL, BindingDB, PubChem BioAssay | Provides experimental bioactivity data. | Training and validating machine learning models for target prediction. |
| Palmitoleoyl 3-carbacyclic Phosphatidic Acid | Palmitoleoyl 3-carbacyclic Phosphatidic Acid, MF:C20H37O5P, MW:388.5 g/mol | Chemical Reagent | Bench Chemicals |
| Riluzole hydrochloride | Riluzole hydrochloride, MF:C8H6ClF3N2OS, MW:270.66 g/mol | Chemical Reagent | Bench Chemicals |
The integration of virtual screening and molecular dynamics provides a powerful, synergistic framework for accelerating the discovery of bioactive hits from the vast and structurally diverse universe of natural products. By leveraging these in silico tools, researchers can efficiently navigate biologically relevant chemical space, prioritize the most promising NP-derived leads with novel scaffolds, and gain deep mechanistic insights into their interactions with therapeutic targets. This computational approach de-risks and informs subsequent experimental validation, paving the way for the development of new drugs inspired by nature's intricate chemistry. As these methodologies continue to advance, particularly with the integration of machine learning and increasing computational power, their role in unlocking the full potential of natural products for drug discovery will only become more profound.
The exploration of natural products represents a cornerstone in drug discovery, offering access to a vast and structurally diverse chemical space that is largely untapped by synthetic compound libraries [49]. Natural product libraries are a source of diverse 3D molecular features furnishing an array of biological functions and are resourceful in furnishing scaffolds for drug discovery research [64]. However, the very complexity that makes these mixtures so valuable also presents significant technical challenges throughout the discovery pipeline. The process of prospecting active molecules from these complex mixtures is classically performed by bio-guided isolation, but this is intensive work that can be hampered by false positive results and loss of activity through multiple fractionation steps and repetitive bioassays [64]. This whitepaper examines the principal technical barriers in screening, isolation, and characterization of complex natural product mixtures within the context of exploring biologically relevant chemical space (BioReCS) for drug discovery research, and details emerging technological solutions overcoming these limitations.
The concept of chemical space (CS) or chemical universe refers to the theoretical multidimensional space encompassing all possible chemical compounds, where molecular properties define coordinates and relationships between compounds [1]. Within this vast universe, the biologically relevant chemical space (BioReCS) comprises molecules with biological activityâboth beneficial and detrimental [1]. Natural products exhibit distinct and privileged occupancy within BioReCS, populating regions that often lack representation in synthetic medicinal chemistry databases [49].
Comprehensive analyses reveal that natural products possess distinctive physicochemical properties that differentiate them from synthetic compounds and drugs, while largely adhering to the Rule of Five, which renders them a valuable and necessary component of screening libraries for drug discovery [24]. Studies using the chemical space navigation tool ChemGPS-NP have demonstrated that natural products cover unique regions of chemical space not adequately explored by conventional medicinal chemistry compounds, indicating these regions represent promising yet underexplored territory for drug discovery [49].
Table 1: Chemical Space Comparison: Natural Products vs. Medicinal Chemistry Compounds
| Property | Natural Products | Medicinal Chemistry Compounds | Analysis Method |
|---|---|---|---|
| Structural Rigidity | Generally more structurally rigid [49] | Generally more flexible [49] | ChemGPS-NP Principal Component 4 (PC4) [49] |
| Aromaticity | Lower degree of aromaticity [49] | Higher degree of aromaticity [49] | ChemGPS-NP Principal Component 2 (PC2) [49] |
| Size & Lipophilicity | Similar distribution [49] | Similar distribution [49] | ChemGPS-NP Principal Components 1 & 3 (PC1, PC3) [49] |
| Scaffold Diversity | High scaffold diversity, unique topologies [50] | Limited scaffold diversity, focused around historical targets [50] | Scaffold topology analysis [50] |
Certain regions of BioReCS remain underexplored due to significant technical challenges in their investigation. These include:
The success of any screening campaign is fundamentally dependent on the quality of the chemical library. Constructing high-quality natural product librariesâwhether from microbial, plant, marine, or other sourcesâis a costly and technically challenging endeavor [74]. These libraries can be composed of crude extracts, semi-pure fractions, or single purified natural products, each design carrying distinctive advantages and disadvantages [74]. Crude extract libraries have lower resource requirements for sample preparation but demand significant effort for the subsequent identification of bioactive constituents. Pre-fractionated libraries balance preparation effort with a shortened active principle identification timeline, while purified natural product libraries require substantial upfront resources but simplify the hit detection process to that of synthetic single-component libraries [74].
A critical step in natural product screening is dereplicationâthe process of rapidly identifying known compounds present in a mixture to avoid redundant rediscovery [74]. This process is essential for prioritizing novel leads and allocating resources efficiently. The use of mass spectrometry and HPLC-mass spectrometry together with spectral databases serves as a powerful tool in the chemometric profiling of bio-sources for natural product production [74]. High-throughput, high-sensitivity flow NMR is also emerging as a valuable tool in this area [74].
To overcome the limitations of traditional bioassay-guided fractionation, advanced screening technologies have been developed that directly probe ligand-target interactions.
Affinity selection mass spectrometry (AS-MS) is a consolidate high-throughput screening (HTS) technique that interrogates non-covalent target-ligand complexes as a non-functional assay [64]. It is a label-free biophysical method that discloses binders solely by mass spectrometry data, providing conditions for chemical annotation of the identified ligands [64]. A key advantage is its ability to identify several ligands exhibiting multiple mechanisms of action against the same target, including orthosteric and allosteric ligands [64].
The AS-MS workflow involves four major stages [64]:
Figure 1: AS-MS Workflow for Natural Product Screening
AS-MS can be implemented in various formats, primarily categorized into solution-based and immobilized target approaches [64]. Each method presents distinct advantages and limitations, which must be considered when designing a screening campaign.
Table 2: AS-MS Methodologies: Comparative Analysis
| Method Type | Specific Techniques | Key Features | Considerations |
|---|---|---|---|
| Solution-Based | Size exclusion chromatography (SEC), Ultrafiltration, Vacuum filtration, Gel filtration [64] | Target remains in native state; Suitable for soluble proteins [64] | Potential for ligand loss with rapid off-rates [64] |
| Immobilized Target | Affinity capture MS (AC-MS), Magnetic microbeads (MagMass), Ligand-fishing [64] | Target can be recycled; Controlled washing conditions [64] | Potential for target denaturation during immobilization [64] |
Ultrafiltration-based AS-MS has been successfully applied to explore 5-lipoxygenase (5-LOX) ligands in Inonotus obliquus, leading to the identification of botulin, lanosterol, and quercetin as promising molecules [64].
NMR spectroscopy offers unique opportunities for screening complex mixtures due to its unbiased nature and rich structural information content [75]. Unlike mass spectrometry, NMR is less biased toward specific compound classes, providing relatively uniform detection across diverse metabolites [75]. Techniques such as SAR by NMR and STD-NMR have been effectively utilized to screen molecular libraries directly in mixtures, without the need for prior separation [74] [75].
A significant barrier in natural product research is the isolation of compounds that are chemically unstable or present in minute quantities. Traditionally, achieving high purity through chromatographic fractionation was deemed essential for successful NMR characterization, but this approach excluded metabolites not robust enough to survive chromatography [75]. Furthermore, activity-guided isolation may overlook biologically important compounds that act synergistically rather than individually [75].
NMR spectroscopy has undergone a paradigm shift, evolving from a technique relegated primarily to pure compounds to a powerful tool for characterizing complex metabolite mixtures [75]. This approach is particularly valuable for identifying otherwise inaccessible small molecules, such as compounds prone to chemical decomposition that cannot be isolated [75].
Pioneering work analyzing unfractionated biofluids has demonstrated the power of this approach. For example:
The various available hyphenated techniques (e.g., GC-MS, LC-PDA, LC-MS, LC-FTIR, LC-NMR, LC-NMR-MS, CE-MS) have made possible the pre-isolation analyses of crude extracts or fractions from different natural sources [76]. These integrated systems enable:
Ultra-high-performance liquid chromatography coupled with quadrupole-Orbitrap high-resolution mass spectrometry has been successfully applied for comprehensive chemical constituent analysis of complex natural products like Ranunculus sceleratus L. [77].
Success in navigating the technical barriers of natural product research requires specialized reagents, materials, and instrumentation. The following toolkit details essential resources for effective screening, isolation, and characterization.
Table 3: Research Reagent Solutions for Natural Product Research
| Tool/Reagent | Function/Application | Technical Specifications |
|---|---|---|
| Ultrafiltration Membranes | Separation of target-ligand complexes from unbound molecules in AS-MS [64] | Molecular weight cutoffs 500-500,000 Da; Compatible with centrifugal force, vacuum, or pressure [64] |
| Immobilized Target Platforms | Ligand fishing using immobilized biological targets on solid supports [64] | Magnetic microbeads (MagMass); Functionalized chromatography resins [64] |
| Cryogenic NMR Probes | Enhanced sensitivity for NMR analysis of limited samples or low-abundance compounds [75] | 1-mm HTS cryogenic probes providing 25-fold greater sensitivity than conventional probes [75] |
| Hyphenated System Columns | Chromatographic separation prior to mass spectrometric or NMR detection [76] [77] | UHPLC columns compatible with high-resolution MS systems (e.g., Quadrupole-Orbitrap) [77] |
| Chemical Dereplication Databases | Rapid identification of known compounds to avoid redundant rediscovery [74] | Spectral databases for MS and NMR; Dictionary of Natural Products (DNP) [74] [49] |
| L-Nio dihydrochloride | L-Nio dihydrochloride, MF:C7H17Cl2N3O2, MW:246.13 g/mol | Chemical Reagent |
| Cerbinal | Cerbinal, MF:C11H8O4, MW:204.18 g/mol | Chemical Reagent |
To maximize efficiency in exploring natural product chemical space, an integrated workflow incorporating advanced technologies at each stage is essential. The following diagram illustrates a modern approach that combines chemical and biological profiling for functional annotation of complex natural product mixtures.
Figure 2: Integrated Workflow for Natural Products Research
This workflow emphasizes the integration of chemical profiling and biological screening data early in the process, enabling informed prioritization of leads before committing to resource-intensive isolation efforts. Advances in chemoinformatics tools and molecular networking (MN) allow researchers to relate the presence or absence of specific metabolites to observations of biological phenotypes in profiling assays [78]. This integrated systems biology approach provides a broad perspective on the biological roles of all metabolites in complex samples, ultimately accelerating the identification of novel therapeutic candidates from nature's chemical treasure trove.
The technical barriers in screening, isolation, and characterization of complex natural product mixtures remain significant, yet technological advances are rapidly overcoming these historical limitations. Methods such as affinity selection mass spectrometry and NMR-based mixture screening are transforming the screening landscape, while hyphenated analytical techniques and advanced chemoinformatic tools are accelerating the isolation and characterization process. By adopting integrated workflows that combine chemical and biological profiling, researchers can more effectively navigate the biologically relevant chemical space occupied by natural products, bridging the gap between computational methods and experimental validation. As these technologies continue to evolve, natural products will maintain their essential role in drug discovery, providing novel chemical scaffolds with unique properties that continue to elude conventional synthetic approaches.
Natural products (NPs) and their derivatives have historically been a prolific source of therapeutic agents, accounting for a significant proportion of FDA-approved small-molecule drugs [79] [12] [80]. These compounds, derived from plants, microorganisms, and marine organisms, exhibit remarkable structural diversity and complexity that often surpasses synthetic chemical libraries [12]. However, a critical challenge persists in natural product-based drug discovery: the resupply problem. Transitioning a natural compound from a "screening hit" through a "drug lead" to a "marketed drug" creates exponentially increasing demands for compound amount, which frequently cannot be met by re-isolation from original biological sources due to limited availability, environmental concerns, and unsustainable harvesting practices [79].
This whitepaper examines how sustainable sourcing and synthetic biology approaches are solving the natural product resupply problem within the broader context of exploring natural product chemical space for drug discovery. With over 1.1 million natural products currently documented in databasesâonly approximately 10% of which are readily purchasableâthe scientific community faces significant challenges in accessing these compounds for comprehensive research and development [12]. We explore integrated strategies that combine advanced biotechnology, bioinformatics, and engineering principles to create reliable, scalable, and environmentally responsible resupply pipelines, thereby enabling the continued utilization of nature's chemical richness in pharmaceutical development.
Natural products occupy a broader chemical space compared to synthetic compounds, characterized by higher structural complexity, increased stereochemical diversity, and distinct physicochemical properties [12] [80]. Analyses of natural product databases reveal that these compounds typically feature more chiral centers, higher oxygen content, greater molecular rigidity, and aliphatic ring systems that contrast with the predominance of aromatic rings in synthetic libraries [80]. This structural diversity directly contributes to their biological relevance and success as drug leads, but also complicates their chemical synthesis and resupply.
Table 1: Comparative Analysis of Natural Product Properties Versus Synthetic Compounds
| Property | Pure Natural Products (PNP) | Natural Products & Derivatives (SNP) | Synthetic Compounds |
|---|---|---|---|
| Molecular Weight | 393.9 | 409.2 | Typically <500 |
| ClogP | 2.3 | 3.7 | <5 |
| H-bond Donors | 2.7 | 1.4 | â¤5 |
| H-bond Acceptors | 6.6 | 6.4 | â¤10 |
| Ring Count | 3.6 | 3.5 | Variable |
| Rotatable Bonds | 5.2 | 6.1 | Variable |
| Chiral Atoms | 5.5 | 1.4 | Fewer |
| Lipinski Violations â¥2 | 18% | 10% | <10% |
Source: Adapted from Life Chemicals Natural Product-like Compound Library analysis [80]
The resupply challenge is compounded by several factors: limited natural availability, with many bioactive compounds present in minute quantities in their source organisms; environmental sustainability concerns regarding large-scale harvesting of sensitive species; and the structural complexity of natural products that often makes traditional chemical synthesis economically unviable [79] [12]. Furthermore, certain natural products originate from organisms that are difficult to cultivate or from extreme environments such as deep-sea ecosystems, presenting additional practical challenges for sustainable sourcing [12].
Synthetic biology applies engineering principles to biological systems, creating engineered biological platforms that can address the resupply challenge through multiple approaches. This field has reoriented natural product drug discovery by enabling the development of microbial biofactories and engineered biosynthetic pathways that can produce complex natural products sustainably and at scale [81].
The foundational approach in synthetic biology involves identifying and transferring entire biosynthetic gene clusters from native producers to heterologous hosts such as E. coli or S. cerevisiae. This strategy was pioneered with the discovery that giant biosynthetic units, such as the 28-protein module that synthesizes erythromycin in Actinomycetes, could be isolated and implemented in host organisms [81]. Success in this area requires extensive pathway engineering, including codon optimization, promoter engineering, and balancing enzyme expression levels to avoid burdening the host metabolism.
The artemisinin bioproduction project represents a landmark achievement in this domain. Through sophisticated metabolic engineering, researchers developed yeast strains capable of producing artemisinic acid, a precursor to the antimalarial drug artemisinin, which traditionally was extracted from the sweet wormwood plant (Artemisia annua) with significant supply chain limitations [81]. This project demonstrated the viability of synthetic biology for producing complex plant-derived natural products in microbial systems, establishing a blueprint for numerous subsequent efforts.
Advances in genome sequencing and bioinformatics have enabled genome mining approaches that identify cryptic biosynthetic gene clusters in microbial genomes, revealing enzymes capable of performing novel chemical transformations [82]. This strategy has been particularly valuable for discovering enzymes that catalyze stereodivergent transformations, providing access to diverse stereoisomers of natural product scaffolds that might be difficult to obtain through chemical synthesis [82].
Table 2: Key Bioinformatic Tools for Natural Product Biosynthetic Gene Cluster Analysis
| Tool Name | Primary Function | Application in Resupply Solutions |
|---|---|---|
| antiSMASH | Identification & analysis of biosynthetic gene clusters | Prediction of natural product pathways from genomic data |
| SMURF | Similar function to antiSMASH | Genome mining for secondary metabolite pathways |
| Natural Product-Likeness Scorer | Computational assessment of natural product similarity | Prioritization of compounds for library development |
| GDB-17 | Enumeration of possible organic molecules | Virtual exploration of synthesizable chemical space |
| SANCDB | Curated database of natural compounds & analogs | Resource for natural product discovery & optimization |
Source: Compiled from multiple references [36] [12] [81]
Genome mining has uncovered enzymes with noncanonical activities that exhibit unusual stereoselectivities, significantly expanding the toolbox available for biocatalytic production of natural products [82]. These enzymes can process diverse substrate scopes, enabling the generation of products with distinct stereochemical markers that are crucial for pharmaceutical efficacy. The discovery of such enzymes through genome mining provides new biocatalytic tools that can be integrated into synthetic biology platforms for natural product synthesis.
Synthetic biology employs engineered genetic circuits to create cellular factories with precisely controlled behaviors. These circuits typically comprise three elements: an inducer (small molecule, ligand, or light), a genetically encoded circuit that processes the input signal, and an output (such as a reporter gene or target natural product) [81]. Such systems can be designed for dynamic regulation of metabolic fluxes, improving titers of desired compounds by avoiding the accumulation of intermediate metabolites that might be toxic to the host cell.
Synthetic cellular models can also function as screening platforms for both target-based and phenotypic-based drug discovery approaches [81]. These systems can be designed to incorporate human drug targets or disease-relevant pathways, enabling direct screening of natural product libraries while simultaneously developing production strains for hit compounds. This integration of discovery and production represents a powerful paradigm for accelerating natural product-based drug development.
This protocol outlines the key steps for identifying novel natural product biosynthetic pathways through genome mining:
Genome Sequencing and Assembly: Sequence target organism genomes using Illumina, PacBio, or Oxford Nanopore technologies to obtain high-quality draft or complete genomes. For complex environmental samples, perform metagenomic sequencing.
Bioinformatic Analysis: Utilize specialized tools such as antiSMASH (Antibiotics & Secondary Metabolite Analysis Shell) or SMURF (Secondary Metabolite Unknown Regions Finder) to identify biosynthetic gene clusters (BGCs) encoding natural product pathways [81]. These tools detect characteristic signature sequences of polyketide synthases (PKS), non-ribosomal peptide synthetases (NRPS), and other biosynthetic systems.
Comparative Genomics: Perform phylogenomic analysis to identify unique or divergent BGCs by comparing against databases of known gene clusters. Prioritize clusters with novel architectures or in silent/silenced genomic regions.
Heterologous Expression: Clone prioritized BGCs into suitable expression vectors (e.g., BAC, cosmic, or artificial chromosome vectors) and introduce into heterologous hosts such as Streptomyces coelicolor, E. coli, or S. cerevisiae [81]. Optimize expression through promoter engineering and ribosome binding site modification.
Metabolite Analysis: Characterize compounds produced by recombinant strains using LC-MS/MS and NMR spectroscopy. Compare spectral data against natural product databases to identify novel compounds.
Pathway Engineering: Refactor the BGC for improved production titers by optimizing codon usage, removing regulatory elements, and balancing expression of individual genes.
This protocol provides a framework for engineering microbial hosts for natural product production:
Host Selection: Choose an appropriate microbial host (E. coli, S. cerevisiae, B. subtilis) based on the target natural product's biosynthetic requirements, including precursor availability, cofactor requirements, and potential toxicity.
Pathway Design: Design biosynthetic pathways using bioinformatic tools, breaking down the target molecule into biosynthetic steps and identifying or engineering enzymes for each transformation.
Genetic Construct Assembly: Assemble genetic constructs using modern DNA assembly methods (Golden Gate, Gibson Assembly, CRISPR/Cas9). Include appropriate regulatory elements (promoters, RBS, terminators) for balanced expression.
Host Transformation and Screening: Introduce constructs into the host organism and screen for production using analytical methods (HPLC, LC-MS). Employ high-throughput screening methods when applicable.
Strain Optimization: Implement iterative cycles of the Design-Build-Test-Learn paradigm to improve production titers. Strategies include:
Bioprocess Development: Scale up production from laboratory flasks to bioreactors, optimizing process parameters (pH, temperature, aeration, feeding strategies) for maximum yield and productivity.
The following diagrams illustrate key synthetic biology workflows for solving the natural product resupply problem.
Diagram Title: Biosynthetic Pathway Engineering Workflow
Diagram Title: Genetic Circuit Design Framework
Implementation of synthetic biology approaches for natural product resupply requires specialized research reagents and tools. The following table details key resources available to scientists working in this field.
Table 3: Essential Research Reagents and Tools for Natural Product Synthetic Biology
| Tool/Reagent Category | Specific Examples | Function & Application |
|---|---|---|
| Natural Product Libraries | Selleck Natural Product Library (3,673 compounds) [83]; Life Chemicals Natural Product-like Libraries (15,000+ compounds) [80] | High-throughput screening for bioactivity; source of lead compounds |
| Specialized Databases | Super Natural II; Coconut 2.0; Dictionary of Natural Products; SANCDB; NAPRORE-CR [12] | Cheminformatic analysis; chemical space exploration; target prediction |
| Genome Mining Tools | antiSMASH; SMURF; BAGEL; PRISM [81] | Identification of biosynthetic gene clusters; pathway prediction |
| Heterologous Host Systems | E. coli BAP1; S. cerevisiae; Streptomyces coelicolor; Bacillus subtilis [81] | Production chassis for heterologous expression of biosynthetic pathways |
| Genetic Engineering Tools | CRISPR-Cas9; Gibson Assembly; Golden Gate Assembly; SEVA plasmids [81] | Genetic manipulation; pathway construction; genome editing |
| Bioinformatic Resources | Natural Product-Likeness Calculator; Synthetic Accessibility Score (SAS) tools [36] [80] | Assessment of compound natural product similarity; synthetic feasibility evaluation |
| Metabolic Modeling Software | COBRApy; OptFlux; GEMs for host organisms [81] | Prediction of metabolic fluxes; identification of engineering targets |
Synthetic biology provides a powerful and expanding toolkit for addressing the longstanding resupply problem in natural product-based drug discovery. By leveraging advances in metabolic engineering, genome mining, and genetic circuit design, researchers can create sustainable production platforms for complex natural products that would otherwise be inaccessible for development as therapeutic agents. These approaches are complemented by sophisticated cheminformatic analyses that help prioritize the most promising natural product scaffolds for development [12].
The future of natural product resupply will likely involve increased integration of artificial intelligence and machine learning approaches to predict biosynthetic pathways, optimize enzyme function, and design engineered production strains [36] [84]. Additionally, the exploration of previously untapped natural sources, including organisms from extreme environments and microbial dark matter, will continue to expand the accessible natural product chemical space [12]. As these technologies mature, synthetic biology platforms will become increasingly central to natural product-based drug discovery, enabling reliable, scalable, and sustainable access to nature's chemical diversity for the development of next-generation therapeutics.
The Nagoya Protocol on Access to Genetic Resources and the Fair and Equitable Sharing of Benefits Arising from their Utilization (ABS) is an international agreement that establishes a transparent legal framework for the effective implementation of one of the three objectives of the Convention on Biological Diversity (CBD): the fair and equitable sharing of benefits arising from the utilization of genetic resources [85]. For researchers exploring natural products for drug discovery, this protocol has profound implications. Natural products (NPs) and their derivatives represent a significant source of therapeutic agents, accounting for approximately 56.1% of all drugs approved by the FDA between 1981 and 2019 [60]. The protocol recognizes that each country has sovereign rights over the genetic resources within its jurisdiction and aims to ensure that benefits arising from their use are shared fairly and equitably [85].
In practical terms, "genetic resources" within this context include any material of plant, animal, microbial, or other origin containing functional units of heredity that possesses actual or potential value, including their derivatives [85]. For drug discovery professionals, this encompasses the biological materials typically used in natural product research, from which novel bioactive compounds are often isolated. The protocol also covers "traditional knowledge" associated with genetic resources, defined as the knowledge, innovations, and practices of indigenous and local communities relevant for the utilization of genetic resources [85]. This dual coverage creates both obligations and opportunities for researchers seeking to explore the vast chemical space of natural products for therapeutic development.
The Nagoya Protocol operates on several foundational principles that researchers must understand:
The Nagoya Protocol entered into force on 12 October 2014 and has been implemented through various national legislations [85]. While the core principles remain consistent, specific requirements can vary significantly between countries. As of 2020, the CBD and Nagoya Protocol on ABS had been ratified, accorded to, approved, or accepted by 196 and 123 countries, respectively [88]. More recent developments include India's Biological Diversity (Access and Benefit Sharing) Regulation 2025, which has expanded its scope to include Digital Sequence Information (DSI) [86].
Brazil represents another important case study, having established its legal framework through Law 13.123/15 and the National System for the Management of Genetic Resources and Associated Traditional Knowledge (SisGen) [60]. A notable feature of Brazilian legislation is that foreign researchers can access native biodiversity only if associated with public or private Brazilian scientific and technological research institutions, which must take responsibility for registering the activity [60].
Table 1: Key Implementation Differences in Select Jurisdictions
| Jurisdiction | Governing Legislation | Unique Requirements | Special Considerations |
|---|---|---|---|
| European Union/UK | Regulation (EU) No 511/2014; The Nagoya Protocol (Compliance) Regulations 2015 | Due diligence declarations at research funding and commercialization stages | User compliance monitoring by Office for Product Safety and Standards [85] |
| India | Biological Diversity Act, 2002; ABS Regulation 2025 | Benefit sharing slabs based on annual turnover; includes DSI | Prior intimation to National Biodiversity Authority for resource access [86] |
| Brazil | Law 13.123/15; SisGen registry | Mandatory association with Brazilian institutions for foreign researchers | Registry replaces previous authorization system, reducing bureaucratization [60] |
Researchers must follow a systematic approach to determine their obligations under the Nagoya Protocol. The following workflow outlines the key decision points in the compliance process:
Depending on how genetic resources are obtained, researchers must follow different compliance pathways. The Nagoya Protocol distinguishes between direct access (obtaining resources directly from the country of origin) and indirect access (obtaining resources from a third party such as a collaborator or registered collection) [85].
For direct access, researchers must:
For indirect access, researchers must:
Compliance with the Nagoya Protocol requires meticulous record-keeping. Users of genetic resources accessed under the protocol are required to seek, keep, and transfer to subsequent users specific information for a period of 20 years following the end of the period of use [85]. Required records include:
Table 2: Essential Documentation for Nagoya Protocol Compliance
| Document Type | Purpose | When Required | Retention Period |
|---|---|---|---|
| Internationally Recognized Certificate of Compliance (IRCC) | Evidence that genetic resources were accessed in accordance with PIC and MAT | For all utilization of genetic resources covered by the protocol | 20 years after end of use [85] |
| Due Diligence Declaration | Declaration of compliance with ABS requirements to regulatory authorities | At research funding receipt and commercialization stages [85] | 20 years after end of use |
| Mutually Agreed Terms (MAT) | Contract specifying benefit-sharing arrangements | Before accessing genetic resources for utilization | 20 years after end of use |
| Prior Informed Consent (PIC) | Permission from provider country | Before accessing genetic resources | 20 years after end of use |
| Transfer Documentation | Records of any transfer of genetic resources to third parties | When providing genetic resources to other researchers | 20 years after end of use [85] |
The Nagoya Protocol requires fair and equitable sharing of benefits arising from the utilization of genetic resources. Benefit-sharing can take various forms, categorized as monetary and non-monetary benefits [85] [86].
Monetary benefits may include:
Non-monetary benefits may include:
Recent regulatory developments have introduced more precise frameworks for calculating benefit-sharing obligations. India's 2025 ABS Regulations, for example, delineate slabs based on the annual turnover of the person/industry utilizing the resources [86]. The regulations also specify that for biological resources having high conservation or economic value (such as red sanders or agarwood), the benefit sharing shall not be less than 5% of the proceeds of the auction or sale amount or the purchase price and could be more than 20% in case of commercial use [86].
For intellectual property commercialization, if a person commercializes a product based on IPR developed using biological resources, they must share a monetary benefit up to 1% of the annual gross ex-factory sale price (excluding taxes), depending on the sector and case specifics [86].
The drug discovery workflow for natural products must be adapted to incorporate ABS compliance at each stage. The following diagram illustrates a Nagoya-compliant natural product drug discovery pipeline:
The National Cancer Institute's (NCI) Program for Natural Product Discovery provides an exemplary model of implementing ABS principles in large-scale natural product library development. The NCI produces a library of 1,000,000 partially purified natural product fractions for distribution to the research community [88]. The program adheres to collection agreements based on the NCI Letter of Collection (LOC), which stipulates equitable benefit sharing from commercial products derived from discoveries, irrespective of whether a formal agreement has been signed by each participating source country [88].
For researchers creating smaller-scale libraries, essential practices include:
Modern cheminformatics approaches can support ABS compliance while facilitating natural product discovery. With over 1.1 million natural products documented in current databases, chemoinformatics analysis reveals that natural products occupy broader chemical spaces than synthetic compounds [12]. However, the limited availability of NPs (only ~10% purchasable) and redundancy in known scaffolds pose major challenges in NP research [12].
Specialized natural product databases provide valuable resources for research that can complement ABS compliance:
These databases enable similarity analysis and virtual screening approaches that can help researchers prioritize genetic resources for access, potentially reducing the need for extensive physical sampling while still exploring chemical diversity.
Table 3: Essential Research Tools for Nagoya-Compliant Natural Product Research
| Tool/Category | Specific Examples | Function in NP Drug Discovery | ABS Compliance Relevance |
|---|---|---|---|
| ABS Compliance Platforms | ABS Clearing-House [87] | Platform for exchanging information on access and benefit-sharing | Provides IRCCs, information on country requirements, and regulatory updates |
| Natural Product Databases | COCONUT, SuperNatural II, NPASS, LOTUS [89] | Chemical information on natural product structures and properties | Enables preliminary screening and reduces unnecessary physical access to resources |
| Cheminformatics Tools | ChemSAR [90], DeepAutoQSAR [91] | SAR modeling and molecular property prediction | Facilitates efficient use of accessed materials through in silico approaches |
| Molecular Representation | RDKit, CDK, OpenBabel, PaDEL [90] | Molecular descriptor calculation and fingerprint generation | Enables comprehensive analysis of chemical space from limited samples |
| Sample Management Systems | NCI's Natural Product Repository [88] | Physical storage and distribution of natural product fractions | Maintains chain of custody and transfer documentation |
| Regulatory Tracking | SisGen (Brazil), NBA (India) portals [60] [86] | Country-specific compliance documentation | Manages national regulatory requirements for specific jurisdictions |
The Nagoya Protocol and ABS regulations represent a critical framework that researchers must integrate into their natural product drug discovery workflows. While presenting additional complexity, these regulations enable ethical and sustainable exploration of natural product chemical space. Future developments in this area will likely include:
For researchers, success in this evolving landscape requires understanding both the scientific and regulatory dimensions of natural product discovery. By implementing robust compliance workflows from the outset and leveraging modern cheminformatics tools, drug discovery professionals can continue to explore the rich chemical space of natural products while ensuring fair and equitable benefit sharing with provider countries and communities.
The exploration of natural products for drug discovery is akin to searching for a needle in a haystack, with the added challenge that most of the easily discoverable "needles" have already been found. Rediscovery, the repeated identification of known compounds, represents a significant bottleneck in natural product-based drug discovery, consuming valuable resources and time. The underlying issue stems from the immense scale of chemical spaceâthe theoretical space containing all possible organic molecules. Research indicates that while known chemical databases contain millions of compounds, this represents only a tiny fraction (<0.1%) of the possible small molecule structures that could be synthesized and tested [93]. This vast unexplored territory offers tremendous opportunity but also presents formidable challenges in navigation. The concept of the chemotypeâa chemically distinct entity defined by its specific composition of secondary metabolitesâprovides a crucial framework for addressing this challenge [94]. By focusing on novel chemotype discovery rather than simply new compound isolation, researchers can implement strategic approaches to systematically explore uncharted regions of chemical space and minimize redundant rediscovery of known chemical scaffolds.
A chemotype is defined as a chemically distinct entity within a species that demonstrates consistent differences in its secondary metabolite profile, largely under genetic control [94]. Importantly, chemotypes may be morphologically indistinguishable, with their chemical differences arising from minor genetic or epigenetic variations that nonetheless produce significant changes in chemical phenotype. In practical terms, chemotypes are often classified based on the most abundant secondary metabolite produced by an individual organism. For instance, Thymus vulgaris (thyme) demonstrates seven distinct chemotypes characterized by whether thymol, carvacrol, linalool, geraniol, sabinene hydrate, α-terpineol, or eucalyptol dominates its essential oil composition [94]. This classification system, while useful, has limitations, as it may oversimplify complex chemical profiles where multiple compounds contribute significantly to biological activity.
The systematic analysis of chemotypes provides a powerful alternative to traditional molecular descriptor-based approaches for assessing chemical diversity. Chemotype-based diversity analysis offers several distinct advantages for addressing rediscovery. By focusing on molecular scaffolds or core structures, chemotype analysis enables researchers to quantify and maximize the structural diversity of compound libraries, ensuring that screening collections encompass the broadest possible range of chemical skeletons [95]. Studies have demonstrated that diversity selection algorithms based on chemotype analysis can outperform traditional methods using molecular fingerprints, retrieving a larger share of the chemotypes contained in a library when selecting subsets of compounds [95]. This approach is particularly valuable for designing general-purpose screening libraries against novel targets with limited prior structural information, as it maximizes the probability of identifying novel bioactive scaffolds with minimal compound throughput.
Table 1: Comparison of Diversity Assessment Methods
| Method | Basis | Advantages | Limitations |
|---|---|---|---|
| Chemotype Analysis | Molecular scaffolds/core structures | Intuitive interpretation; maximizes structural diversity; efficient library design | May oversimplify complex structures |
| Molecular Fingerprints | Binary representation of structural features | Comprehensive structural representation; well-established algorithms | Less intuitive; may miss scaffold diversity |
| Molecular Quantum Numbers (MQN) | 42 integer descriptors counting atom/bond types, polarity, topology | Simple, universal chemical space classification; easily identifiable features | Newer approach with limited track record |
Active learning represents a powerful machine learning strategy for efficiently navigating chemical space by iteratively selecting the most informative compounds for experimental evaluation. When combined with first-principles based alchemical free energy calculations, this approach enables targeted exploration of regions containing high-affinity binders while explicitly evaluating only a small subset of a large chemical library [96]. The protocol typically involves an iterative cycle where, at each iteration, a carefully chosen fraction of compounds undergoes computational evaluation, and the resulting affinity data trains machine learning models to improve predictions for subsequent rounds. This strategy has been successfully applied to identify high-affinity phosphodiesterase 2 (PDE2) inhibitors, robustly identifying a large fraction of true positives while dramatically reducing the computational resources required compared to exhaustive screening [96].
Dynamic hybrid pharmacophore models (DHPM) represent an innovative approach that addresses the limitations of conventional pharmacophore models by incorporating protein flexibility and multiple binding sites. Unlike conventional pharmacophore models generated from single binding sites, DHPMs capture the combined interaction features of different binding pockets, enabling identification of novel chemotypes that simultaneously engage multiple regions of a target [97]. The development of DHPMs typically involves molecular dynamics simulations of target structures with ligands bound to adjacent sites (e.g., cofactor binding site and substrate binding site), trajectory clustering to identify stable interaction features, and generation of pharmacophore hypotheses that represent the combined binding characteristics. This approach has demonstrated success in identifying structurally diverse compounds with improved binding strength and drug-like properties compared to those identified through conventional methods [97].
The comprehensive enumeration of chemical space from first principles provides a foundational approach for systematic exploration of novel chemotypes. Initiatives such as the Chemical Universe Generated Databases (GDB) have demonstrated that almost all small molecules (>99.9%) have never been synthesized and remain available for exploration [93]. The classification and representation of this chemical space using systems such as Molecular Quantum Numbers (MQN)â42 integer value descriptors that count elementary molecular features including atom and bond types, polar groups, and topological characteristicsâenable intuitive navigation and identification of underrepresented regions [93]. By mapping known natural products and synthetic compounds within this framework, researchers can identify "white spaces" in chemical space that represent opportunities for novel chemotype discovery, strategically targeting these regions through synthesis or focused natural product sourcing.
Table 2: Computational Strategies for Novel Chemotype Discovery
| Strategy | Key Methodology | Application in Novel Chemotype Discovery | Technical Requirements |
|---|---|---|---|
| Active Learning with Free Energy Calculations | Iterative ML model training with alchemical free energy calculations | Efficient navigation toward high-affinity chemotypes in large libraries | MD simulation expertise; ML infrastructure |
| Hybrid Dynamic Pharmacophore Models | MD simulations of multi-site binding; hybrid pharmacophore generation | Identification of chemotypes binding multiple target sites simultaneously | MD simulation capability; pharmacophore modeling tools |
| Chemical Space Enumeration & Mapping | Systematic enumeration using MQN descriptors; chemical space visualization | Targeted exploration of underrepresented chemical regions | Large-scale computing; cheminformatics expertise |
The initial extraction process critically influences the range of chemotypes accessible from biological source material. While traditional methods like maceration, percolation, and Soxhlet extraction remain valuable, contemporary extraction techniques offer improved efficiency, reduced extraction times, and decreased solvent consumption [98]. Key advanced methods include:
Ultrasound-assisted extraction: Utilizes ultrasonic energy to enhance cell wall disruption and improve solvent penetration, typically reducing extraction time and temperature requirements.
Microwave-assisted extraction: Employs microwave energy to rapidly heat the sample, accelerating desorption of compounds from the matrix while using less solvent than conventional methods.
Pressurized solvent extraction: Uses solvents at elevated temperatures and pressures to maintain them in liquid state above their normal boiling points, significantly improving extraction efficiency and speed.
These methods must be carefully optimized to prevent degradation of labile natural products, as temperature selection is crucial for maintaining compound stability [98]. The choice of extraction method should be guided by the chemical properties of the target compounds and the nature of the biological matrix.
The selection of extraction solvents plays a pivotal role in determining the quality, quantity, and selectivity of isolated compounds, thereby directly influencing the range of accessible chemotypes. Traditional organic solvents present significant disadvantages including volatility, toxicity, and environmental concerns [98]. The development of green solvent systems represents a crucial advancement for accessing novel chemotypes while addressing sustainability and safety concerns:
Natural deep eutectic solvents (NADES): These solvent systems typically consist of natural primary metabolites (e.g., choline chloride combined with sugars, organic acids, or alcohols) that form eutectic mixtures with superior extraction properties for various natural product classes.
Ionic liquids: These designer solvents offer tunable properties through selection of appropriate cation-anion combinations, enabling selective extraction of specific chemotype classes.
Supercritical and subcritical fluids: Supercritical COâ offers tunable solvating power by varying temperature and pressure, while subcritical water exhibits altered polarity and improved extraction efficiency for more polar compounds.
The principle of "like dissolves like" remains fundamental to solvent selection, with solvents having polarity values near the target solute's polarity generally performing better [98]. However, innovative solvent systems can overcome these traditional limitations, enabling access to previously challenging chemotypes.
Dereplicationâthe process of rapidly identifying known compounds in complex mixturesârepresents a critical strategy for minimizing rediscovery early in the isolation pipeline. Modern dereplication approaches typically combine sophisticated analytical techniques with database searching:
LC-MS/MS and LC-HRMS: Liquid chromatography coupled with tandem mass spectrometry or high-resolution mass spectrometry provides structural information that can be matched against natural product databases.
NMR-based dereplication: Advanced NMR techniques, including DOSY, LC-NMR, and microcryoprobe NMR, enable structure elucidation directly in complex mixtures with minimal purification.
Database integration: Automated searching against comprehensive natural product databases (e.g., AntiBase, Dictionary of Natural Products) facilitates rapid identification of known compounds.
Implementation of robust dereplication protocols at the earliest possible stage of extraction and fractionation enables researchers to prioritize fractions containing potentially novel chemotypes, effectively allocating resources to the most promising leads.
A comprehensive, integrated workflow is essential for systematic isolation of novel chemotypes while minimizing rediscovery. The following diagram illustrates a strategic approach that combines computational and experimental methods:
This integrated workflow emphasizes the critical importance of interdisciplinary collaboration between biologists, chemists, and computational scientists, which has been shown to significantly advance natural product research [98]. For example, partnerships between chemical engineers and biologists have clarified relationships between extraction methods and the biological activity of natural compounds like C-phycocyanin [98]. Similarly, collaboration between natural product chemists and computational chemists enables the effective application of chemical space mapping and virtual screening to guide experimental isolation efforts.
Table 3: Essential Research Reagents and Materials for Novel Chemotype Isolation
| Reagent/Material | Function/Application | Considerations for Novel Chemotype Discovery |
|---|---|---|
| Natural Deep Eutectic Solvents | Green extraction medium with tunable properties | Enhanced extraction of specific chemotype classes; reduced environmental impact |
| Ionic Liquids | Designer solvents for selective extraction | Customizable for target compound polarity; improved selectivity |
| Supercritical COâ | Non-polar extraction medium | Tunable solvating power; minimal solvent residues; temperature-sensitive compounds |
| Hybrid Silica Materials | Chromatographic stationary phases | Enhanced separation of complex natural product mixtures |
| Chiral Stationary Phases | Enantioseparation of natural products | Resolution of stereoisomers; access to enantiomerically pure chemotypes |
| Molecularly Imprinted Polymers | Selective solid-phase extraction | Target-specific isolation; reduced matrix interference |
| LC-MS/MS Systems | Dereplication and structure elucidation | Rapid identification of known compounds; prioritization of novel leads |
| Microcryoprobe NMR | Structure elucidation of limited samples | Enhanced sensitivity for rare or minor novel chemotypes |
The systematic isolation of novel chemotypes from natural sources requires a multifaceted strategy that integrates computational guidance with experimental innovation. By leveraging approaches such as active learning protocols, dynamic hybrid pharmacophore models, and comprehensive chemical space mapping, researchers can strategically navigate the vast terrain of unexplored chemistry [96] [93] [97]. Simultaneously, advances in extraction technologies, green solvent systems, and early-stage dereplication provide the experimental tools necessary to access and identify novel chemical entities efficiently [98]. The continuing development of interdisciplinary collaborations promises to further enhance our ability to explore natural product chemical space, addressing the persistent challenge of rediscovery while unlocking new sources of valuable bioactive compounds for drug discovery and other applications. As these methodologies evolve and integrate, they create a powerful framework for systematic exploration of nature's chemical diversity, ensuring that natural product research continues to contribute novel chemotypes to the drug discovery pipeline.
The high failure rate of drug candidates in late development stages poses a significant challenge for the pharmaceutical sector, with poor pharmacokinetics and toxicity accounting for numerous setbacks. Early integration of in silico Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) prediction now serves as a crucial strategy to mitigate this risk. This approach is particularly vital within the context of exploring natural product chemical space, a proven source of structurally diverse compounds with privileged biological activities. By employing computational ADMET tools at the initial stages of drug discovery, researchers can effectively filter out molecules with unfavorable pharmacokinetic profiles, prioritize promising candidates from complex natural product extracts, and guide the optimization of lead compounds. This whitepaper provides an in-depth technical guide to the methodologies, applications, and limitations of early in silico ADMET prediction, with a specific focus on its role in unlocking the therapeutic potential of natural products for drug discovery research [99] [24] [100].
Natural products represent a heterogeneous group of compounds with diverse molecular properties that often occupy regions of chemical space not explored by synthetic compound libraries [24]. These molecules largely adhere to the rule-of-five, rendering them a valuable and necessary component of screening libraries [24]. However, a significant hurdle in identifying secondary metabolites from medicinal microbes is the presence of "sleeping gene clusters" â silent biosynthetic pathways that remain unactivated under standard laboratory conditions [99]. Techniques such as abiotic stress, including exposure to heavy metals, can activate these clusters to elicit novel secondary metabolites with enhanced pharmacological profiles [99].
The high rate of medication failures underscores the importance of ADMET evaluation early in the drug design process [100]. Selecting appropriate experimental data for ADMET prediction and applying it effectively in the context of physiological characteristics remains challenging [100]. In silico ADMET models, built on verified experimental datasets using key classifying factors and molecular descriptors, provide a powerful solution to these challenges [100]. When framed within the exploration of natural product chemical space, early ADMET prediction becomes an indispensable tool for identifying and optimizing novel therapeutic agents from biological sources.
Computer-aided drug design (CADD) methods are broadly categorized into structure-based drug design (SBDD) and ligand-based drug design (LBDD) approaches [101]. These computational tools interpret and guide experiments to expedite the antibiotic drug design process, and by extension, the discovery of drugs from natural products [101].
SBDD methods analyze macromolecular target 3-dimensional structural information, typically of proteins or RNA, to identify key sites and interactions important for biological function [101]. This information guides the design of drugs that can compete with essential interactions involving the target [101]. Key SBDD methodologies include:
LBDD methods focus on known ligands for a target to establish a relationship between their physiochemical properties and biological activities, known as a structure-activity relationship (SAR) [101]. This information guides optimization of known drugs or design of new drugs with improved activity [101]. Key software packages covering both SBDD and LBDD capabilities include Discovery Studio, OpenEye, Schrödinger, and MOE [101].
Recent advancements in ADMET prediction include comprehensive benchmarking of predictors, particularly those leveraging foundation models [102]. Evaluation protocols now employ sophisticated data splitting strategies to test model generalization:
Technical paradigms for ADMET predictors include end-to-end deep learning models (e.g., GNNs and Transformers) that automatically learn feature representations, and feature-based classical models that rely on expert-engineered molecular descriptors [102]. Roughness Index variants (MODI, SARI, ROGI) help analyze model performance and dataset difficulty [102].
This section details a representative methodology for discovering natural products with optimized ADMET properties, combining metal stress elicitation with computational validation.
Based on research from Streptomyces sp. SH-1312 [99]
Objective: To elicit secondary metabolite production through heavy metal stress and evaluate their pharmacological activities.
Materials:
Methodology:
The following diagram illustrates the integrated experimental-computational workflow for natural product discovery with early ADMET optimization:
Diagram 1: Integrated discovery workflow for natural products with ADMET optimization.
Objective: To predict ADMET properties of candidate molecules using computational tools.
Materials:
Methodology:
Model Selection and Training:
Prediction and Validation:
The metal stress approach successfully elicited production of anhydromevalonolactone (MVL) from Streptomyces sp. SH-1312, a metabolite absent in normal culture conditions [99]. This case demonstrates the power of integrated methodology for discovering compounds with favorable ADMET profiles.
Table 1: Antioxidant and Cytotoxic Activities of Anhydromevalonolactone (MVL)
| Assay Type | Specific Assay | ICâ â Value (µg/mL) | Standard Used | Standard ICâ â (µg/mL) |
|---|---|---|---|---|
| Antioxidant | DPPH Scavenging | 19.65 ± 5.7 | Ascorbic Acid | 6.52 ± 4.92 |
| NO Inhibition | 15.49 ± 4.8 | Ascorbic Acid | 8.44 ± 4.17 | |
| OHâ Inhibition | 19.65 ± 5.22 | Gallic Acid | 6.26 ± 6.39 | |
| Iron Chelation | 19.38 ± 7.11 | EDTA | 10.20 ± 6.54 | |
| Cytotoxic (PC3 Cell Lines) | 24-hour exposure | 35.81 ± 4.2 | - | - |
| 48-hour exposure | 23.29 ± 3.8 | - | - | |
| 72-hour exposure | 16.25 ± 6.5 | - | - |
MVL exhibited remarkable antioxidant activities across multiple assays and demonstrated time-dependent cytotoxic activity against PC3 cancer cell lines [99]. Further mechanistic studies revealed that MVL exerts pharmacological efficacy by upregulation of P53 and BAX while downregulation of BCL-2 expression, indicating induction of apoptotic pathways [99].
Table 2: ADMET Profile and Molecular Docking of MVL
| Property Category | Specific Property | Result for MVL |
|---|---|---|
| Toxicity | Hepatotoxicity | Safer profile |
| Cytochrome Inhibition | Safer profile | |
| Cardiotoxicity | Non-cardiotoxic | |
| Molecular Docking | Target Protein | Binding Energy |
| P53 | Good binding energy in active region | |
| BAX | Good binding energy in active region |
During ADMET predictions, MVL displayed a favorable safety profile with no significant hepatotoxicity, cytochrome inhibition, or cardiotoxicity concerns [99]. Molecular docking studies confirmed that MVL binds in the active region of target proteins P53 and BAX [99]. The research triumphantly announced a prodigious effect of heavy metals on actinobacteria with fringe benefits as a key tool for MVL production with a strong pharmacological and pharmacokinetic profile [99].
Successful implementation of early ADMET optimization requires specific research reagents and computational resources. The following table details key components for establishing this workflow.
Table 3: Research Reagent Solutions for ADMET-Optimized Natural Product Discovery
| Category | Item | Function/Application |
|---|---|---|
| Biological Materials | Actinobacteria strains (e.g., Streptomyces sp.) | Source of diverse secondary metabolites with pharmaceutical potential [99] |
| Gause's Medium | Specialized culture medium for actinobacteria cultivation [99] | |
| Elicitors | Heavy metal ions (Co²âº, Zn²âº) | Abiotic stress agents to activate silent gene clusters and enhance metabolite production [99] |
| Analytical Tools | HPLC with PDA/UV detector | Metabolic profiling, purification, and quantification of elicited compounds [99] |
| NMR spectroscopy | Structural elucidation of novel natural products [99] | |
| Computational Resources | ADMET prediction tools (e.g., AutoGluon, TabPFNv2) | Automated training and prediction of pharmacokinetic and toxicity properties [102] |
| Molecular docking software (e.g., AutoDock Vina) | Prediction of ligand-target interactions and binding affinities [101] | |
| Chemical databases (e.g., ZINC) | Source of compound structures for virtual screening and model training [101] | |
| Specialized Software | MD simulation packages (e.g., CHARMM, AMBER) | Simulation of molecular dynamics and protein-ligand interactions [101] |
| CADD platforms (e.g., Schrödinger, MOE) | Integrated suites for computer-aided drug design [101] |
The following diagram illustrates the apoptotic mechanism identified for MVL, demonstrating how computational predictions align with experimental findings:
Diagram 2: MVL mechanism of action through apoptotic pathway regulation.
The integration of early in silico ADMET prediction into natural product drug discovery represents a paradigm shift in pharmaceutical development. By leveraging computational tools to evaluate pharmacokinetic and toxicity properties at initial stages, researchers can efficiently navigate the complex chemical space of natural products, prioritize candidates with the highest therapeutic potential, and reduce late-stage attrition rates. The case study of MVL production through metal stress elicitation demonstrates the power of this integrated approach, where compounds with favorable ADMET profiles can be identified and optimized before extensive laboratory investment. As ADMET prediction models continue to advance through benchmarked frameworks and sophisticated machine learning algorithms, their role in exploring biologically relevant natural product chemical space will become increasingly indispensable for discovering novel therapeutic agents with optimized pharmacological properties.
The exploration of natural product chemical space offers a powerful strategy for discovering novel therapeutic agents. Natural products (NPs) are a heterogeneous group of compounds with diverse molecular properties that often occupy regions of chemical space not explored by standard synthetic compounds while largely adhering to drug-like principles [24]. This chemical diversity makes them a valuable, unique, and necessary component of screening libraries for drug discovery. However, the full potential of this diversity can only be realized through rigorous standardization and quality control during library construction. Without these controls, the inherent complexity of natural product sources leads to irreproducible results, misidentified activities, and ultimately, failed discovery efforts.
This technical guide outlines the critical standardized processes and quality control measures required to construct biologically relevant natural product libraries that effectively populate and navigate chemical space for drug discovery research. By implementing these protocols, researchers can transform raw biodiversity into systematically organized, well-characterized libraries capable of supporting high-throughput screening (HTS) campaigns and generating reliable, reproducible data for downstream development.
The construction of natural product libraries begins with ethical and regulated sample acquisition. The access and use of biological resources must be mutually agreed upon between the researcher and the country of origin, which maintains sovereign rights over these resources [60]. The Convention on Biological Diversity (CBD) established at the 1992 United Nations Conference provides the foundational framework based on three pillars: conservation of biological diversity, sustainable use of its components, and fair and equitable sharing of benefits derived from genetic resources [60].
The Nagoya Protocol on Access to Genetic Resources and the Fair and Equitable Sharing of Benefits Deriving from Their Utilization supplements the CBD and provides a legal framework for benefit sharing [103]. As of 2020, the CBD and Nagoya Protocol have been ratified or accepted by 196 and 123 countries respectively [103]. Researchers must secure all necessary permitsâincluding collection, shipping, and export permitsâbefore initiating fieldwork, as this process can be time-consuming but is essential for legal and ethical compliance.
Countries rich in biodiversity have implemented specific legal frameworks to regulate access to their genetic resources. For example, Brazil established Law 13.123/15 and the National System for the Management of Genetic Resources and Associated Traditional Knowledge (SisGen) to facilitate compliance with CBD principles [60]. A significant aspect of this framework requires foreign researchers to collaborate with Brazilian scientific institutions, which must assume responsibility for registering the activity [60]. These regulations emphasize fair and equitable benefit-sharing arrangements that must be negotiated with source countries, potentially including technology transfer, royalty agreements, or capacity building initiatives.
Proper specimen collection and documentation are fundamental to creating traceable and reproducible natural product libraries. The collection process must include the creation of voucher specimens that are accurately tagged (e.g., with barcoded labels) and documented with essential metadata [103]. This documentation should include:
These voucher specimens must be deposited in recognized herbariums or collections to maintain taxonomic verification and enable future recollection efforts. Comprehensive metadata collection establishes the foundation for sample tracking databases that support both scientific reproducibility and compliance with regulatory requirements.
Following collection, biological samples require careful processing to preserve chemical integrity. While specific protocols vary by organism type (plant, marine, microbial), general principles include:
Standardization across these steps minimizes chemical variation between batches and ensures consistent quality throughout the library's lifetime.
Extraction protocols must balance comprehensive metabolite recovery with reproducibility and compatibility with downstream screening platforms. Recent methodological advances have improved extraction efficiency while streamlining workflow:
Table 1: Standardized Extraction Methods for Natural Product Libraries
| Method | Principle | Advantages | Applications |
|---|---|---|---|
| Pressurized Liquid Extraction | Uses high pressure and temperature | Reduced solvent usage, faster processing | Ideal for solid plant materials |
| Ultrasound-Assisted Extraction | Applies ultrasonic energy | Enhanced extraction efficiency, minimal thermal degradation | Suitable for thermolabile compounds |
| Microwave-Assisted Extraction | Uses microwave energy | Rapid, selective heating, reduced extraction time | Effective for polar compounds |
| Supercritical Fluid Extraction | Employ supercritical COâ | Solvent-free, tunable selectivity | Valuable for lipophilic compounds |
The US National Cancer Institute's Natural Product Repository, one of the world's largest collections, generates between 15,000 and 20,000 extracts annually through high-throughput processing methods, demonstrating the scalability of standardized approaches [103].
Prefractionation significantly improves screening outcomes by reducing complexity and concentrating minor metabolites. Various chromatographic techniques are employed:
Prefractionated libraries demonstrate improved screening performance through higher confidence in hit rates, enhanced biological activity from concentrated minor metabolites, sequestration of nuisance compounds, and streamlined downstream processes [103]. The NCI's Cancer Moonshot program exemplifies large-scale implementation, producing a library of approximately 1,000,000 partially purified natural product fractions in 384-well plates for distribution to the research community [103].
Robust quality control requires comprehensive analytical characterization to ensure batch-to-batch consistency and compound integrity. The following table outlines essential analytical methods:
Table 2: Analytical Quality Control Methods for Natural Product Libraries
| Method | Quality Control Parameters | Acceptance Criteria |
|---|---|---|
| LC-MS/MS | Metabolic profiling, identity confirmation | Retention time stability, characteristic mass fragments |
| NMR Spectroscopy | Structural confirmation, purity assessment | Signal-to-noise ratio, absence of contaminant peaks |
| HPLC-UV/ELSD | Chromatographic fingerprint, purity | Peak area consistency, resolution of critical pairs |
| Standardized Bioassay | Biological activity baseline | Activity within historical control ranges |
Implementing these analytical controls enables the detection of degradation, contamination, or other inconsistencies that could compromise screening results.
Systematic assessment of chemical space coverage ensures library diversity and drug-likeness. Computational analysis demonstrates that natural products cover a much larger volume of chemical diversity space than combinatorial compounds [60]. Principal Component Analysis (PCA) using descriptors including AlogP, molecular weight, hydrogen bond donors/acceptors, rotatable bonds, and ring systems reveals that natural products occupy distinct regions of chemical space often complementary to synthetic compounds [104]. Research indicates that approximately 52% of natural products comply with Lipinski's "Rule of Five" for drug-likeness, while 71.8% meet at least three of the four criteria [104], confirming their relevance to drug discovery.
The following diagram illustrates the complete standardized workflow for constructing natural product libraries:
Protocol: Standardized Solid-Phase Extraction for Natural Product Prefractionation
Background: This protocol describes a standardized solid-phase extraction method for fractionating natural product extracts into distinct chemical fractions based on polarity, reducing complexity for biological screening [103].
Materials and Reagents:
Equipment:
Procedure:
Critical Notes:
Validation: Validate the fractionation scheme using standard compounds with known polarity profiles. Assess fraction quality by TLC or LC-MS to confirm distinct chemical profiles between fractions.
Table 3: Essential Research Reagents for Natural Product Library Construction
| Category | Specific Items | Function & Importance |
|---|---|---|
| Extraction Solvents | Methanol, ethanol, ethyl acetate, hexane, water | Comprehensive extraction of diverse metabolite classes with varying polarity |
| Chromatography Media | C18 reversed-phase, silica gel, Sephadex LH-20, ion-exchange resins | Fractionation and purification based on chemical properties |
| Analytical Standards | Natural product standards, internal standards (e.g., umbelliferone) | Quality control, method validation, quantification |
| Stabilizing Agents | DMSO, glycerol, ascorbic acid, butylated hydroxytoluene (BHT) | Compound stabilization, prevention of degradation during storage |
| Storage Materials | 384-well plates, amber glass vials, septa, desiccants | Long-term sample integrity maintenance, prevention of moisture/light damage |
| Bioassay Reagents | Cell culture media, assay buffers, enzyme substrates, detection reagents | Standardized biological screening across library specimens |
Standardization and quality control transform the inherent chemical diversity of natural sources into reliable, screening-ready libraries that effectively explore natural product chemical space. By implementing the comprehensive framework outlined in this guideâencompassing ethical collection, standardized processing, rigorous quality control, and systematic documentationâresearchers can construct natural product libraries that deliver reproducible results and identify novel bioactive compounds with drug development potential. As computational tools like ChemGPS-NP and Scaffold Hunter continue to evolve [24], their integration with well-characterized physical libraries will further enhance our ability to navigate chemical space and address unmet medical needs through natural product-inspired drug discovery.
Natural Products (NPs) and their derivatives have been a cornerstone of medicine for centuries, evolving from ancient herbal remedies to the discovery of transformative drugs like morphine and quinine [105]. The mid-20th century marked a 'golden age' for antibiotic discovery from natural sources, which subsequently expanded into other therapeutic areas [105]. Despite a shift in focus towards technological advances and synthetic compound libraries in the late 20th century, natural products remain an indispensable source of molecular innovation. This whitepaper provides a quantitative analysis of the significant contribution of NPs to the pharmaceutical landscape, particularly to FDA-approved drugs, framing this contribution within the essential context of exploring natural product chemical space for modern drug discovery research. The extensive structural diversity and complexity of NPs, which frequently exhibit unique glycosylation and halogenation patterns, render them invaluable for probing biologically relevant chemical space and identifying novel therapeutic agents [12].
A comprehensive review of drugs approved globally between January 2014 and the end of 2024 provides a clear metric for the contribution of natural products. Among all 579 drugs approved in this period, 56, or 9.7%, were classified as NPs or NP-derived (NP-D) [105]. This total comprises 44 New Chemical Entities (NCEs), representing 7.6% of all approvals and 11.3% of all NCEs, and 12 NP-Antibody Drug Conjugates (NP-ADCs), accounting for 2.1% of all approvals and 6.3% of all New Biological Entities (NBEs) [105]. The annual number of new NP-D NCEs and NP-ADCs has fluctuated, averaging five approvals per year since 2014 [105]. This data underscores the consistent and vital role of NPs in filling pharmaceutical pipelines, even amidst a growing number of biological therapies.
Table 1: Global Drug Approvals and NP-Derived Contributions (2014-2024)
| Category | Total Approvals | NP-Derived Approvals | Percentage of Total | Percentage of Subcategory |
|---|---|---|---|---|
| All Drugs | 579 | 56 | 9.7% | - |
| New Chemical Entities (NCEs) | 388 | 44 | 7.6% | 11.3% |
| New Biological Entities (NBEs) | 191 | 12 (NP-ADCs) | 2.1% | 6.3% |
The momentum for NP-derived drugs continues. Between January 2014 and June 2025, a total of 58 NP-related drugs were launched globally [105]. This figure includes 45 NP and NP-D new chemical entities and 13 NP-antibody drug conjugates, highlighting the successful integration of natural product warheads with advanced biologic platforms [105]. Looking forward, the clinical pipeline remains robust. As of the end of December 2024, 125 NP and NP-D compounds were identified as undergoing clinical trials or in the registration phase [105]. Notably, thirty-three new pharmacophores not previously found in approved drugs are currently in development, signaling ongoing innovation in this field, although the discovery of truly novel pharmacophores has slowed, with only one discovered in the past 15 years [105].
The chemical space occupied by natural products is distinct from that of synthetic compounds. Current databases document over 1.1 million unique natural products, which display high structural diversity and complexity [12]. NPs frequently incorporate complex ring systems, glycosylation, and halogenation, features that are often underrepresented in synthetic screening libraries [12]. Chemoinformatic analyses consistently show that NPs occupy a broader chemical space than synthetic compounds while largely adhering to the Rule-of-Five, which predicts favorable oral bioavailability [24]. This makes them a valuable, unique, and necessary component of screening libraries for drug discovery [24].
The structural features of NPs are heavily influenced by their source organisms and environments, creating unique subspaces within the broader NP chemical universe:
Table 2: Key Databases for Navigating Natural Product Chemical Space
| Database Name | Scope and Specialization | Primary Application in Research |
|---|---|---|
| Super Natural II | A comprehensive database of natural products [12]. | Virtual screening and chemoinformatic analysis. |
| Dictionary of Marine Natural Products | Focuses on compounds isolated from marine organisms [12]. | Research on marine-derived chemical space. |
| Coconut | An open, curated database of natural products [12]. | Comparative analysis and accessible data for sourcing. |
| Natural Products Repository of Costa Rica (NAPRORE-CR) | Geographically focused open-access database [12]. | Exploring region-specific chemical diversity. |
| PeruNPDB | The Peruvian Natural Products Database for in silico screening [12]. | Drug discovery from traditionally sourced compounds. |
The journey from a natural source to a drug candidate involves a multidisciplinary workflow that integrates traditional techniques with modern technological approaches. The following diagram visualizes this integrated process.
This foundational protocol is critical for identifying active compounds from complex natural extracts [105].
This computational protocol characterizes the structural landscape of NPs to guide discovery [12].
Successful navigation of NP chemical space and development of NP-derived drugs rely on a suite of specialized reagents, databases, and tools.
Table 3: Essential Research Toolkit for NP Drug Discovery
| Tool / Reagent Category | Specific Examples | Function and Application |
|---|---|---|
| NP Sourcing & Databases | Dictionary of Marine Natural Products [12], Super Natural II [12], NAPRORE-CR [12] | Provides curated structural and source data for virtual screening and chemoinformatic analysis. |
| Chromatography Media | C18 reversed-phase silica gel, Sephadex LH-20 | Purification and fractionation of complex natural extracts. C18 separates by hydrophobicity; Sephadex LH-20 separates by size and polarity in organic solvents. |
| Analytical Standards | Commercially available NP libraries (e.g., MicroSource Spectrum) | Used as benchmarks in HPLC-MS for dereplication (early identification of known compounds to avoid rediscovery). |
| Cheminformatic Software | RDKit, ChemGPS-NP [24], Scaffold Hunter [24] | Calculates molecular properties, visualizes chemical space, and analyzes scaffold diversity to prioritize novel structures. |
| Target Prediction Tools | AI-driven platforms (e.g., SuperPred, SEA) | Predicts potential protein targets for a NP based on its structural similarity to known ligands, generating testable hypotheses for MoA studies. |
The quantitative data presented herein leaves no doubt: natural products continue to be a significant and indispensable source of new chemical entities and pharmacophores for the pharmaceutical industry. The approval of 45 NP-derived NCEs in just over a decade, coupled with a robust pipeline of 125 clinical-stage candidates, firmly establishes their enduring value [105]. However, the declining discovery rate of novel pharmacophores signals a need for a strategic shift [105]. The future of NP drug discovery lies in the sophisticated navigation of its vast chemical space. This requires a renewed emphasis on bioassay-guided isolation coupled with detailed mode of action studies to identify new drug leads [105], integrated with advanced chemoinformatic approaches to map diversity and target unexplored regions [12] [24]. Leveraging artificial intelligence for target prediction, exploring untapped species and extreme environments, and mitigating the challenges of compound redundancy and availability are the key strategies that will unlock the next generation of life-saving medicines from nature's chemical treasury [12].
Natural products (NPs) and their derivatives represent an invaluable resource in the anticancer drug discovery pipeline, accounting for over half of all approved anticancer medicines [106]. Their unparalleled chemical diversity provides a vast resource for discovering novel compounds with enhanced efficacy and safety profiles [107]. Drug repurposingâidentifying new therapeutic applications for existing drugsâhas emerged as a strategy to significantly shorten the traditional 13-15 year drug development pathway at a fraction of the cost [108]. This whitepaper explores prominent case studies of artemisinin, ivermectin, and other natural products undergoing investigation as anticancer agents, focusing on their mechanisms of action, experimental evidence, and research methodologies relevant to drug development professionals.
Artemisinin is a sesquiterpene lactone isolated from Artemisia annua L. (qinghao) and is characterized by a crucial endoperoxide bridge essential for its biological activity [109] [106]. Due to limitations in artemisinin's solubility and bioavailability, several derivatives have been developed, including dihydroartemisinin (DHA), artesunate (ART), artemether, and arteether [109] [106]. Artesunate is rapidly hydrolyzed to the active metabolite DHA under physiological conditions [106]. The peroxide group is activated by heme or intracellular iron, leading to the generation of cytotoxic reactive oxygen species (ROS) and carbon-centered radicals, which mediate both antimalarial and anticancer effects [109] [108].
Artemisinin derivatives exhibit multifaceted anticancer activity through several interconnected mechanisms:
Table 1: Anticancer Activity of Artemisinin and Its Derivatives Across Cancer Types
| Cancer Type | Compound | Experimental Model | Key Findings | IC50 / Effective Concentration |
|---|---|---|---|---|
| Breast Cancer | Dihydroartemisinin (DHA) | MCF-7 cells | Suppressed proliferation, induced autophagy and pyroptosis, targeted cancer stem cells [106]. | 129.1 μM (24 h) [106] |
| Artemisinin | MCF-7 cells | Suppressed cell growth, induced ferroptosis [106]. | 396.6 μM (24 h) [106] | |
| Artesunate | MCF-7 cells | Induced apoptosis [106]. | 83.28 μM (24 h) [106] | |
| Lung Cancer | Artemisinin | A549 cells | Inhibited proliferation and metastasis, induced apoptosis [106]. | 28.8 μg/mL [106] |
| Dihydroartemisinin | PC9 cells | Induced ferroptosis and apoptosis, inactivated STAT3 [106]. | 19.68 μM (48 h) [106] | |
| Artemisinin derivative 4 | H1299 cells | Antiproliferation effect, induced ferroptosis [106]. | 0.09 μM [106] | |
| Liver Cancer | Artemisinins | HepG2, PLC/PRF/5 cells | Induced G2/M cell cycle arrest [109]. | Varies by compound and cell line [106] |
| Dihydroartemisinin | Hepatic stellate cells | Induced S-phase cell cycle arrest [109]. | Varies by cell line [106] |
Objective: To evaluate the in vitro antiproliferative activity of an artemisinin derivative and determine its effect on the cell cycle.
Materials:
Methodology:
Cell Viability/Proliferation Assay (SRB Assay):
Cell Cycle Analysis by Flow Cytometry:
Diagram 1: Experimental workflow for cell viability and cycle analysis.
Ivermectin (IVM) is a macrolide antiparasitic drug derived from avermectin, composed of 80% 22,23-dihydroavermectin-B1a and 20% 22,23-dihydroavermectin-B1b [112]. Its discoverers won the Nobel Prize in Physiology or Medicine in 2015. Beyond its established role in treating river blindness and scabies, IVM has demonstrated potent anticancer effects in various in vitro and in vivo models [112].
Ivermectin exerts its anticancer activity through a multi-target mechanism:
Table 2: Documented Anticancer Effects of Ivermectin Across Cancer Models
| Cancer Type | Experimental Model | Key Findings | IC50 / Effective Dose |
|---|---|---|---|
| Cholangiocarcinoma (CCA) | KKU214 (Gem-sensitive) & KKU214GemR (Gem-resistant) cells | Inhibited proliferation & colony formation; Gem-resistant cells were more sensitive [111]. | KKU214: 11.41 μM (48 h); KKU214GemR: 4.05 μM (48 h) [111] |
| Colorectal Cancer | HCT-8/VCR (Vincristine-resistant) cells | Reversed chemoresistance in vitro and in vivo; reduced P-gp expression [113]. | In vivo: 2 mg/kg/day + VCR [113] |
| Breast Cancer | MCF-7/ADR (Adriamycin-resistant) cells | Reversed chemoresistance in vitro and in vivo via EGFR/ERK/Akt/NF-κB pathway [113]. | In vivo: 2 mg/kg/day + ADR [113] |
| Chronic Myeloid Leukemia | K562/ADR (Adriamycin-resistant) cells | Reversed chemoresistance in xenograft mouse model [113]. | In vivo: 2 mg/kg/day + ADR [113] |
| Gastric Cancer | MKN1, SH-10-TC (YAP1-high) cells | Inhibited proliferation in a YAP1-dependent manner [112]. | Sensitive in YAP1-high cells [112] |
Objective: To investigate the ability of ivermectin to reverse multidrug resistance in a xenograft mouse model.
Materials:
Methodology:
Tumor Monitoring and Analysis:
Mechanistic Analysis (P-gp Expression):
Drug Accumulation Analysis (HPLC):
Diagram 2: Ivermectin mechanism for reversing multidrug resistance via the EGFR pathway.
The exploration of natural product chemical space extends beyond terrestrial plants to marine organisms. Marine cyanobacteria, for instance, are prolific sources of potent anticancer agents [114]. Key developments include:
This expanding pipeline, which also includes SERCA inhibitors and mitochondrial cytotoxins, underscores the richness of marine natural products for discovering agents with novel mechanisms to overcome drug resistance [114].
Table 3: Key Research Reagent Solutions for Investigating Natural Product Anticancer Agents
| Reagent / Material | Function in Research | Specific Examples / Notes |
|---|---|---|
| Cell Line Panels | In vitro screening for cytotoxicity, mechanism studies, and resistance modeling. | MCF-7 (breast), A549 (lung), HCT-8 (colorectal), KKU214 (cholangiocarcinoma), and their drug-resistant variants (e.g., MCF-7/ADR, HCT-8/VCR) [113] [111]. |
| Xenograft Mouse Models | In vivo evaluation of efficacy, toxicity, and drug resistance reversal. | Nude mice for solid tumors; NOD/SCID mice for leukemias [113]. |
| Antibodies for Western Blot / IHC | Mechanistic analysis of signaling pathways and target expression. | Antibodies against P-gp, p-EGFR, p-Akt, p-ERK, NF-κB, PAK1, YAP1, and cleaved caspases [112] [113]. |
| Flow Cytometry Reagents | Analysis of cell cycle, apoptosis, and surface markers. | Propidium Iodide (PI), Annexin V-FITC, commercial kits (e.g., FxCycle PI/RNase) [113] [111]. |
| Cytotoxicity Assay Kits | High-throughput assessment of cell viability and proliferation. | MTT, Sulforhodamine B (SRB), and Cell Counting Kit-8 (CCK-8) [111]. |
| HPLC Systems | Quantifying drug concentrations in biological samples (pharmacokinetics) and analyzing compound purity. | Used to measure chemotherapeutic drug accumulation in cells and tissues [113]. |
Artemisinin, ivermectin, and marine-derived cytotoxins exemplify the immense potential of natural products in anticancer drug discovery. The evidence supports their multi-target mechanisms, which include inducing various forms of cell death, overcoming multidrug resistance, and modulating the tumor microenvironment. Future efforts should focus on integrating advanced methodologies such as artificial intelligence, high-throughput screening, and chemical biology to explore novel NP targets and accelerate development [92]. The successful clinical translation of these repurposed drugs and novel natural products will depend on robust, well-designed clinical trials that validate preclinical findings, ultimately increasing the accessibility and affordability of cancer therapies globally [108].
The exploration of chemical space for novel drug leads represents a fundamental challenge in modern drug discovery. This whitepaper provides a comparative analysis of two primary approaches: screening libraries of natural products (NPs) and those comprising synthetic compounds. Natural products, chemical entities produced by living organisms, are pre-validated by nature and have been the historical source of a majority of novel drug classes and essential medicines [17] [49]. In contrast, synthetic libraries, constructed using methodologies like combinatorial chemistry and diversity-oriented synthesis (DOS), offer advantages in terms of scalability and modularity [115] [36]. Framed within the broader thesis of exploring natural product chemical space for drug discovery, this analysis examines the structural diversity, hit-rate performance, and practical applications of these distinct yet complementary strategies, providing drug development professionals with a data-driven guide for library selection and design.
Computational analyses reveal that natural products and synthetic compounds derived from medicinal chemistry efforts occupy notably different regions of biologically relevant chemical space.
Table 1: Chemical Property and Structural Feature Comparison
| Characteristic | Natural Products (NPs) | Synthetic/Bioactive Medicinal Chemistry Compounds |
|---|---|---|
| General Rigidity | Structurally more rigid [49] | Generally more flexible [49] |
| Aromaticity | Lower degree of aromaticity [49] | Higher degree of aromaticity [49] |
| Structural Complexity | Higher structural complexity and more stereocenters [116] [36] | Typically less complex [36] |
| Adherence to Ro5 | ~60% have no Lipinski's Rule of 5 (Ro5) violations; many remain bioavailable despite violations [49] | Designed for Ro5 compliance to ensure oral bioavailability [36] |
| Coverage of Uniqueness | Populate unique, sparsely explored regions of chemical space [49] [24] | Often cluster in over-sampled regions of chemical space [49] |
Tools like ChemGPS-NP have mapped these differences, showing that NPs cover regions that lack representation in typical medicinal chemistry libraries, such as the World Drug Index (WOMBAT) database [49]. This unique occupancy is attributed to evolutionary selection, which optimizes NPs for interactions with biological macromolecules, rendering them a valuable component for any screening library aimed at discovering novel bioactive compounds [49] [24].
Scaffold diversity is a critical metric for assessing the potential of a compound library to yield novel hits. Analyses using frameworks like the Murcko framework and Scaffold Tree hierarchies provide quantitative measures.
Table 2: Scaffold Diversity of Standardized Compound Libraries (41,071 compounds each)
| Compound Library | Number of Unique Murcko Frameworks | Number of Unique Level 1 Scaffolds | Relative Structural Diversity |
|---|---|---|---|
| TCMCD (Natural Product Library) | 4,289 | 5,134 | Highest Complexity |
| Chembridge | 5,268 | 6,441 | High |
| Mucle | 4,953 | 6,123 | High |
| VitasM | 4,866 | 5,978 | High |
| ChemicalBlock | 4,911 | 6,032 | High |
| Enamine | 4,522 | 5,654 | Medium |
| Maybridge | 3,987 | 4,956 | Medium |
The Traditional Chinese Medicine Compound Database (TCMCD), a representative NP library, demonstrates the highest structural complexity among the libraries studied [116]. However, its scaffolds are more conserved, resulting in fewer unique frameworks and Level 1 scaffolds compared to some highly diverse synthetic libraries like Chembridge and Mucle [116]. This suggests that while NPs introduce high-value, complex scaffolds, synthetic libraries can offer a greater raw number of distinct core structures, highlighting a key trade-off.
The ultimate validation of a screening library lies in its ability to produce viable hits and successful drugs. The historical and contemporary data strongly favor natural products in this regard.
The Build/Couple/Pair (B/C/P) strategy in DOS is a powerful method for generating skeletally diverse synthetic libraries with NP-like complexity [115].
Figure 1: The Build/Couple/Pair (B/C/P) workflow in Diversity-Oriented Synthesis (DOS) for generating skeletally diverse compound libraries, such as lactams, from commercially available building blocks [115].
A computational-aided DOS workflow can be implemented using platforms like KNIME to generate a library of lactams, a privileged scaffold in drug discovery [115]. The process involves:
For natural products, a major challenge is the heterologous expression of their often large and complex biosynthetic gene clusters. Modern synthetic biology addresses this through combinatorial DNA assembly and refactoring.
Figure 2: Refactoring the C. jejuni Pgl pathway using combinatorial DNA assembly to optimize heterologous production in E. coli [117].
A seminal application of this approach is the refactoring of the Campylobacter jejuni N-glycosylation (pgl) pathway in E. coli [117]. The experimental protocol is as follows:
Table 3: Key Research Reagent Solutions for Glycoengineering and Library Screening
| Reagent / Tool | Source / Example | Function / Application |
|---|---|---|
| Oligosaccharyltransferase PglB | Campylobacter jejuni | Key enzyme for in vivo N-linked protein glycosylation in bacterial glycoengineering [117] [118] [119]. |
| PNGase F & PNGase A | New England Biolabs | Amidases for enzymatic deglycosylation; gold standard for validating N-glycosylation status of proteins from mammalian (F) and plant/insect (A) systems [120]. |
| ChemGPS-NP | Public Web Resource | Chemical space navigation tool for comparing and visualizing the location of compounds in a property-based reference space [49] [24]. |
| Build/Couple/Pair (B/C/P) | Nielsen and Schreiber, 2007 | A systematic DOS strategy for generating skeletally diverse small molecule libraries from simple building blocks [115]. |
| Combinatorial DNA Assembly (Start-Stop) | Taylor et al., 2019 | Scarless modular DNA assembly system for constructing combinatorial libraries of multigene expression constructs [117]. |
| KNIME Analytics Platform | Open Source | Platform for designing and executing computational workflows, e.g., for generating virtual libraries of lactams [115]. |
| Traditional Chinese Medicine Compound Database (TCMCD) | Academic Source | A curated database of NPs used for analysis of structural complexity and scaffold diversity in comparative studies [116]. |
The comparative analysis between natural product and synthetic libraries reveals a landscape of compelling synergies rather than simple superiority. Natural products provide access to evolutionarily pre-validated, complex chemotypes that occupy unique and biologically relevant regions of chemical space, leading to historically high success rates in delivering novel drugs. Synthetic libraries, particularly those designed using DOS principles, offer unparalleled capacity for exploring vast regions of chemical space and generating high counts of unique, often lead-like scaffolds in a controlled and scalable manner. The future of productive drug discovery lies in the strategic integration of both paradigms. This can be achieved by using computational tools like ChemGPS-NP to identify sparsely populated, biologically relevant areas of chemical space and then employing advanced synthetic biology to refactor NP pathways or sophisticated DOS to populate these regions with novel, synthetically tractable compounds. This integrated approach maximizes the chances of discovering novel, effective, and developable small-molecule therapeutics.
Antibody-Drug Conjugates (ADCs) represent a transformative class of targeted cancer therapeutics that combine the specificity of monoclonal antibodies with the potent cytotoxicity of small-molecule payloads. The structural architecture of ADCs comprises three fundamental components: a monoclonal antibody for target recognition, a chemical linker ensuring stability, and a cytotoxic payload responsible for ultimate tumor cell eradication [121] [122]. Within this sophisticated framework, natural products (NPs) and their derivatives have emerged as indispensable payload sources, contributing significantly to the clinical success of ADC technology.
Natural products have served as historic cornerstones in oncology drug discovery, with over half of approved small-molecule drugs originating directly or indirectly from NP origins [12]. This dominance extends powerfully into the ADC landscape, where NP-derived payloads constitute the majority of currently approved conjugates. The inherent biological compatibility, structural complexity, and potent mechanisms of action exhibited by natural products make them ideal candidates for ADC payload development [92] [121]. These compounds often demonstrate exquisite targeting of fundamental cellular processes, including microtubule dynamics and DNA integrity, with potencies 100 to 1000-fold greater than conventional chemotherapeutics [121].
The exploration of NP chemical space continues to yield valuable insights for ADC development. Current databases document over 1.1 million natural products displaying remarkable structural diversity and complexity, frequently featuring glycosylation and halogenation patterns [12]. NPs occupy broader chemical spaces than synthetic compounds and exhibit distinct characteristics based on their origins, with marine-derived NPs often displaying larger molecular weights and greater hydrophobicity than their terrestrial counterparts [12]. This review examines the rising role of NP-derived payloads within ADC development, framed within the broader context of exploring natural product chemical space for drug discovery research.
The ADC clinical landscape is predominantly populated by natural product-derived payloads, which can be broadly categorized into several mechanistic classes. Table 1 summarizes the key characteristics of NP-derived payload classes used in approved ADCs.
Table 1: Natural Product-Derived Payload Classes in Approved ADCs
| Payload Class | Representative Payloads | Natural Product Origin | Mechanism of Action | Approved ADC Examples |
|---|---|---|---|---|
| Tubulin Inhibitors | DM1, DM4 (maytansinoids) | Maytenus serrata (African shrub) | Inhibits microtubule assembly, disrupting cell division | Trastuzumab emtansine (Kadcyla), Mirvetuximab soravtansine (ELAHERE) |
| Tubulin Inhibitors | Monomethyl auristatin E (MMAE), Monomethyl auristatin F (MMAF) | Dolastatin 10 (marine peptide from Dolabella auricularia) | Inhibits tubulin polymerization, preventing mitosis | Brentuximab vedotin (Adcetris), Enfortumab vedotin (Padcev) |
| DNA-Damaging Agents | Calicheamicin | Micromonospora echinospora (bacterial source) | DNA double-strand breaks via enediyne core | Gemtuzumab ozogamicin (Mylotarg), Inotuzumab ozogamicin (Besponsa) |
| Topoisomerase I Inhibitors | Exatecan (DXd derivative), SN-38 | Camptothecin (Camptotheca acuminata tree) | Stabilizes topoisomerase I-DNA cleavage complexes | Trastuzumab deruxtecan (Enhertu), Sacituzumab govitecan (Trodelvy) |
| DNA Alkylators | SG3199 (PBD dimer) | Pyrrolobenzodiazepines (streptomyces species) | DNA minor groove cross-linking | Loncastuximab tesirine (Zynlonta) |
The market dominance of NP-derived payloads is evident in commercial ADC therapeutics. As of 2024, twelve ADCs have received FDA approval, with eight achieving this milestone in the last five years alone, signaling a maturation of the field [123]. Monomethyl auristatin E (MMAE) represents one of the most successful NP-derived payloads, capturing 41% of the current ADC payload market share [124]. The auristatins originate from the marine peptide dolastatin 10, isolated from the sea hare Dolabella auricularia, demonstrating how exploration of diverse ecological niches yields valuable therapeutic compounds [121].
The commercial impact of NP-derived payloads extends across target indications and therapeutic areas. Table 2 quantifies the market distribution of ADC payloads and their clinical applications based on 2024 market data.
Table 2: Market Distribution of ADC Payloads and Applications (2024)
| Parameter | Category | Market Share (%) | Projected CAGR (%) | Key NP-Derived Payloads |
|---|---|---|---|---|
| Payload Type | Monomethyl Auristatin E (MMAE) | 41 | - | Marine-derived tubulin inhibitor |
| Camptothecin derivatives | 18 | - | Plant-derived topoisomerase I inhibitors | |
| DM1/DM4 (Maytansinoids) | 12 | - | Plant-derived tubulin inhibitors | |
| Target Indication | Breast Cancer | 44.7 | - | T-DM1, T-DXd |
| Lung Cancer | - | 31.6 | T-DXd, Sacituzumab govitecan | |
| Therapeutic Area | Solid Tumors | 71 | 76 (2035 projection) | Various NP-derived payloads |
| Hematological Cancers | 29 | 24 (2035 projection) | Auristatins, Calicheamicin |
The global ADC market, valued at $12.30 billion in 2024, is projected to reach $28.41 billion by 2035, representing a compound annual growth rate (CAGR) of 6.4% [124]. This growth is substantially fueled by the continued innovation in NP-derived payloads, particularly as applications expand beyond hematological malignancies to dominate solid tumor therapeutics, which currently account for 71% of the ADC market share [124].
NP-derived payloads exert their cytotoxic effects through targeting essential cellular processes, with two primary mechanisms dominating the clinical landscape: microtubule disruption and DNA damage.
Microtubule inhibitors, including auristatins (MMAE, MMAF) and maytansinoids (DM1, DM4), bind to tubulin and prevent polymerization into microtubules, disrupting mitotic spindle formation and arresting cell division during mitosis [121]. These compounds demonstrate exceptional potency, with IC50 values in the picomolar to nanomolar range against susceptible tumor cells.
DNA-damaging agents encompass structurally diverse NP-derived compounds including calicheamicins, duocarmycins, and pyrrolobenzodiazepines (PBDs). Calicheamicin, an enediyne antibiotic, binds to the DNA minor groove and generates double-strand breaks via a radical-mediated mechanism [123]. PBD dimers, such as SG3199 in loncastuximab tesirine, form covalent cross-links between opposing strands of DNA, preventing strand separation and essential processes like transcription and replication [123].
The following diagram illustrates the sequential mechanism of action of ADCs from cellular binding to payload-mediated cytotoxicity:
Figure 1: ADC Mechanism of Action from Cellular Binding to Payload-Mediated Cytotoxicity
Despite their potency, the therapeutic efficacy of NP-derived payloads is often limited by the emergence of resistance. A primary mechanism involves drug efflux mediated by ATP-binding cassette (ABC) transporters, particularly P-glycoprotein (P-gp) [122]. These transmembrane proteins recognize and actively export payloads from tumor cells, reducing intracellular accumulation and diminishing cytotoxicity. Multiple NP-derived payloads, including MMAE, DM1, DM4, and calicheamicin, have been identified as P-gp substrates [122].
Additional resistance mechanisms include:
The following diagram illustrates the key resistance mechanisms that impair ADC efficacy:
Figure 2: Key Resistance Mechanisms Limiting ADC Efficacy
Evaluating the cytotoxicity of NP-derived payloads requires sophisticated bioanalytical approaches due to their exceptional potency. The standard workflow involves multiple complementary techniques:
In vitro cell viability assays form the cornerstone of payload potency assessment. These include:
Advanced mechanistic studies provide deeper insights into payload activity:
The exceptional potency of NP-derived payloads necessitates highly sensitive detection methods. "Free payload concentrations are typically low, requiring more sensitive, innovative methodologies. Simple sample preparations such as protein precipitation often need to be replaced with more elaborate ones such as liquid-liquid extraction or solid phase extraction, in combination with the use of the most sensitive triple quad mass spectrometers" [125].
Accurate measurement of NP-derived payloads presents unique technical challenges that require specialized methodologies:
Chromatographic interference must be carefully managed. "Large molecule ADCs, with their much bigger contact surfaces, will have much more retention and generally will not interfere on a reversed-phase liquid chromatography (LC) system. This is regardless of whether or not the molecule is left intact during sample preparation" [125].
Payload stability considerations are critical for accurate quantification. "ADC concentrations can be orders of magnitude higher than payload concentrations. Due to the excess of ADC in study samples, even the smallest amount of ADC degradation will result in huge payload biases over time" [125].
Sample preparation optimization is essential for reliable results. "Incorporating a high organic flush phase in the mobile phase gradient is necessary to prevent column contamination or interference from slow moving large molecules. This flush phase will wash off the ADC and any other high retentive matrix constituents after each injection" [125].
A revolutionary advancement in ADC technology involves the incorporation of two distinct NP-derived payloads within a single conjugate, designed to overcome resistance mechanisms and enhance antitumor efficacy. The field has recently "exploded" with at least 15 dual-payload ADCs disclosed in preclinical presentations as of 2025 [126].
Table 3 highlights promising dual-payload ADC candidates in development:
Table 3: Emerging Dual-Payload ADCs in Development
| ADC Candidate | Company | Target | Payload Combination | Development Stage |
|---|---|---|---|---|
| KH815 | Chengdu Kanghong | TROP2 | Topo1 inhibitor + RNA pol 2 inhibitor | Phase 1 (first in human) |
| DXC018 | Hangzhou Dac | HER2 x HER2 | Topo1 inhibitor + antimetabolite inhibitor | Preclinical |
| Unnamed | Sutro | Undisclosed | Topo1 inhibitor + PARP inhibitor | Preclinical |
| JSKN021 | Jiangsu Alphamab | EGFR x HER3 | Topo1 inhibitor + MMAE | Preclinical |
| IMD2113 | Affinity Biopharmaceutical | EGFR x TROP2 | Undisclosed dual mechanism | Preclinical |
The rationale for dual-payload strategies centers on overcoming resistance: "patients treated with an ADC can relapse not only through loss of the target antigen, but also by developing resistance to the payload that an ADC uses" [126]. However, significant challenges remain in optimizing linker technology specifically for dual-payload configurations and managing potential increased toxicity profiles [126].
Future innovation in NP-derived payloads depends on accessing novel chemical scaffolds from underexplored biological sources. Several promising approaches include:
Marine and extremophile natural products: "Marine NPs are larger and more hydrophobic than terrestrial counterparts, while deep-sea and extremophile-derived NPs show novel scaffolds and bioactivities" [12]. These environments represent rich reservoirs for discovering payloads with unique mechanisms.
AI-enabled NP discovery: "Integrating advanced methodologies, such as artificial intelligence (AI), high-throughput screening, chemical biology, bioinformatics, gene regulation, the highly accurate non-labeling chemical proteomics approach to explore novel NPs targets" [92] will accelerate payload identification.
Addressing NP availability: "The limited availability of NPs (only ~10% purchasable) and the redundancy in known scaffolds pose major challenges in NP research" [12]. Future strategies highlight integrating multidimensional databases and exploring untapped species and extreme environments to uncover unique bioactive compounds [12].
Successful development of ADCs with NP-derived payloads requires specialized reagents and technical capabilities. The following table details essential research tools and their applications:
Table 4: Essential Research Reagents and Resources for ADC Payload Development
| Reagent/Resource | Function/Application | Key Considerations |
|---|---|---|
| High-Potency Payload Standards (MMAE, DM1, Calicheamicin) | ADC assembly and analytical reference standards | Require specialized containment facilities; typically >95% purity [127] |
| Cleavable Linkers (Valine-Citrulline, GGFG) | Enable intracellular payload release | Account for ~70% of ADC market; provide plasma stability with tumor-specific cleavage [128] |
| Site-Specific Conjugation Technologies (Engineered cysteines, unnatural amino acids) | Generate homogeneous ADC products with defined DAR | Growing at >30% CAGR; reduce heterogeneity-related toxicity [128] |
| Triple Quadrupole Mass Spectrometers | Quantify free payload concentrations in plasma | Essential for detecting low payload levels; newest models offer 3-4x sensitivity improvements [125] |
| Specialized Chromatography (Reversed-phase with high aqueous mobile phases) | Separate payloads from ADC molecules | Requires high organic flush phases to prevent column contamination [125] |
| Cell-Based Potency Assays (CellTiter-Glo, MTT) | Determine payload and ADC cytotoxicity | Must include resistant cell lines to assess P-gp mediated efflux [127] [122] |
| Natural Product Databases (Super Natural II, Dictionary of Natural Products) | Source structural and bioactivity data for NP discovery | Contain >1.1 million compounds; only ~10% are readily purchasable [12] |
Natural product-derived payloads continue to dominate the ADC landscape, driven by their unparalleled potency, diverse mechanisms of action, and proven clinical efficacy. The integration of NP chemical space exploration with advanced ADC engineering approachesâincluding site-specific conjugation, novel linker technologies, and emerging dual-payload strategiesâpromises to address current limitations in resistance and therapeutic index. Future directions will increasingly leverage AI-enabled NP discovery, exploration of underexplored biological sources, and innovative bioanalytical methods to quantify payload dynamics with unprecedented precision. As the ADC field continues to mature, NP-derived payloads will remain indispensable components in the targeted therapy arsenal, offering renewed hope for addressing challenging malignancies through their exquisite targeting of fundamental biological processes.
Antimicrobial resistance (AMR) represents one of the most severe global health threats of the 21st century, directly challenging the efficacy of modern medicine. The unchecked use and abuse of traditional antibiotics have precipitated this crisis, leading to increased treatment failures and mortality rates [129]. In response, the World Health Organization (WHO) has prioritized the research and development of new antimicrobial agents. Natural Products (NPs), with their vast and evolutionarily refined chemical diversity, have emerged as a beacon of hope. This whitepaper details how the systematic exploration of natural product chemical space provides powerful, innovative strategies to reconstruct the antibiotic pipeline and combat AMR [129] [30]. We outline the most promising NP-inspired approaches, provide detailed experimental methodologies, and visualize the critical pathways and workflows, offering a technical guide for researchers and drug development professionals.
The AMR crisis is exacerbated by the rapid development of resistance mechanisms in bacteria, including drug efflux pumps, modification of antibiotic targets, and enzymatic degradation of the drugs themselves [130]. The COVID-19 pandemic has further intensified this threat, leading to constrained antibiotic treatment options and surging resistance rates [131].
To focus global research efforts, the WHO has established a Bacterial Priority Pathogens List, categorized by urgency level [131]:
Critical Priority
High Priority
Medium Priority
This list serves as a crucial guide for targeting research on novel antimicrobial agents, with NPs showing significant activity against these resilient pathogens [131].
The chemical space of NPs is inherently "biologically relevant," as these molecules have evolved through natural selection to interact with biological macromolecules [132]. This makes them ideal starting points for drug discovery. Several sophisticated strategies have been developed to systematically explore this space, moving beyond simply isolating novel compounds from nature.
Biology-Oriented Synthesis (BIOS) uses the structural scaffolds of known NPs as inspiration. The core principle is the systematic simplification of complex NP scaffolds into core structures that retain biological relevance but are synthetically more accessible [133] [30].
Diversity-Oriented Synthesis (DOS) aims to rapidly generate libraries of complex and structurally diverse small molecules that populate broad regions of chemical space, mimicking the structural complexity of NPs [133] [30].
Complexity-to-Diversity (CtD) leverages readily available NPs as complex starting materials and uses chemoselective reactions to dramatically rearrange their core structures, generating unprecedented scaffolds [133].
The following diagram illustrates the logical relationship between the NP chemical space and these core exploration strategies.
This section provides detailed methodologies for key experiments cited in this review, serving as a technical reference for researchers aiming to replicate or build upon these findings.
Objective: To identify and evaluate the efficacy of natural products against antibiotic-resistant "priority pathogens" as defined by the WHO [131].
Methodology:
("natural product*" OR "natural compound*") AND (antibacteri* OR antimicrobial*) AND (MDR OR "multi-drug resistant *").Objective: To discover novel bioactive molecules by synthesizing and screening libraries based on simplified NP scaffolds [133] [30].
Methodology:
Objective: To assess the direct antibacterial and anti-virulence properties of NPs and their derivatives [134].
Methodology:
The following table details key reagents, materials, and computational tools essential for research in NP-based antimicrobial discovery.
| Research Reagent / Solution | Function / Application |
|---|---|
| Ethanol, Methanol, Ethyl Acetate | Common solvents for the extraction of bioactive compounds (e.g., alkaloids, flavonoids, terpenoids) from plant materials [131]. |
| Broth Microdilution Plates | High-throughput platform for determining Minimum Inhibitory Concentrations (MICs) against bacterial and fungal pathogens [131] [134]. |
| Crystal Violet Stain | Dye used to quantify total biofilm biomass formed by bacteria on abiotic surfaces after treatment with test compounds [134]. |
| Biotinylated Analogues | Chemical probes derived from hit compounds; used for target identification via pull-down assays and immunoenrichment [133]. |
| SCONP / Scaffold Hunter | Computational algorithms for the systematic analysis and simplification of natural product scaffolds to guide BIOS library design [133]. |
| PHASE (Schrödinger) | Software module used for pharmacophore modeling and 3D-QSAR studies to identify crucial structural features for activity and guide molecular design [135] [136]. |
| Solid-Phase Synthesis Resin | Polymeric support (e.g., silyl-polystyrene) used for DOS and other library synthesis, enabling rapid purification and automation [133] [30]. |
Understanding the biological pathways targeted by NPs and the mechanisms of antibiotic resistance is crucial for rational drug design. The diagram below illustrates a key signaling pathway that can be modulated by NP-inspired molecules and the primary mechanisms bacteria use to resist antibiotics.
The exploration of natural product chemical space represents a paradigm shift in the fight against antimicrobial resistance. Strategies such as Biology-Oriented Synthesis, Diversity-Oriented Synthesis, and Complexity-to-Diversity provide systematic, rational frameworks to discover novel bioactive molecules that bypass conventional resistance mechanisms [129] [133] [30]. The continued integration of these approaches with advances in synthetic chemistry, computational biology, and chemical genomics is paramount. By leveraging NPs as both inspiration and starting points, the scientific community can reconstruct the antibiotic pipeline, transforming this beacon of hope into a new arsenal of effective therapies to safeguard global health for future generations.
The exploration of natural products for drug discovery represents a journey through an expansive chemical multiverse, a term introduced to describe the comprehensive analysis of compound datasets through several distinct chemical spaces, each defined by a different set of chemical representations [137]. Unlike a single, unified chemical space, the chemical multiverse acknowledges that a given set of molecules represented with different descriptors leads to distinct chemical universes, each providing complementary insights into molecular structure and properties [137]. This conceptual framework is particularly relevant to the study of multi-compound extracts from medicinal plants, which are complex, adaptive systems whose therapeutic efficacy often emerges from synergistic interactions between their numerous bioactive constituents [138]. The intrinsic complexity of these extracts means that their overall activity cannot be predicted from the activity of isolated compounds alone, as they contain hundreds or even thousands of individual bioactive molecules in varying abundances [138].
Within this chemical multiverse, synergistic interactions between compounds become a vital part of therapeutic efficacy [138]. Synergy occurs when the combined effect of compounds is greater than the sum of their individual effects, potentially arising through multiple mechanisms including multi-target effects, enhanced bioavailability, or protection from toxicity [138] [139]. This review explores the theoretical foundations, experimental methodologies, and practical applications of synergistic effects in multi-compound natural extracts, framing this exploration within the broader context of navigating natural product chemical space for drug discovery research.
The interactions between multiple bioactive compounds in natural extracts can be systematically classified based on the observed effect relative to the expected effect from individual components. These classifications provide the vocabulary for describing combination effects:
Table 1: Classification and Definitions of Combination Effects
| Interaction Type | Mathematical Relationship | Therapeutic Implication |
|---|---|---|
| Synergy | Combined effect > Sum of individual effects | Enables lower doses, reduces adverse effects, enhances efficacy |
| Additivity | Combined effect = Sum of individual effects | Straightforward dose combination without interaction |
| Antagonism | Combined effect < Sum of individual effects | May reduce toxicity or undesirable effects |
The superior therapeutic performance of multi-compound extracts compared to isolated constituents can be explained by several fundamental mechanisms through which synergy manifests:
Multi-Target Effects (Pharmacodynamic Synergism): Different compounds in a mixture simultaneously engage multiple therapeutic targets or pathways relevant to a disease state [139]. This network-level intervention is particularly valuable for complex, multifactorial diseases where single-target approaches often prove insufficient [138].
Enhanced Bioavailability (Pharmacokinetic Synergism): Certain compounds may improve the absorption, distribution, or metabolic stability of other active compounds in the mixture, thereby increasing their bioavailability and therapeutic concentration [139]. Some natural compounds, while not possessing direct effects themselves, may increase the solubility or inhibit the metabolism of co-administered active compounds [139].
Attenuation of Adverse Effects: Compounds within an extract may interact to reduce the toxicity or side effects associated with individual constituents while maintaining therapeutic efficacy [138]. This protective synergy allows for the use of potentially toxic but highly effective compounds with a improved safety profile [138].
Diagram 1: Fundamental mechanisms of therapeutic synergy in multi-compound extracts
Rigorous experimental design is essential for accurately identifying and quantifying synergistic interactions in natural product research. Several well-established methodologies enable researchers to distinguish true synergy from simple additive effects:
Isobolographic Analysis: This classical graphical method involves constructing an "isobole" - a line connecting the doses of two individual compounds that each produce the same specified effect level (typically the ED50) [140]. The combined doses that fall below this line indicate synergy, while points above the line indicate antagonism [140]. The method is based on the concept of dose equivalence, where the drug B-equivalent of dose a is calculated as aB/A, leading to the fundamental isobole equation: a/A + b/B = 1 [140].
Combination Index (CI) Method: This quantitative approach calculates a combination index to determine the nature of drug interactions [139]. The CI is defined by the equation: CI = dâ/Dxâ + dâ/Dxâ, where dâ and dâ are the respective combination doses of drug one and drug two that produce an effect x, and Dxâ and Dxâ are the corresponding single doses for drug one and drug two that result in the same effect x [139]. CI values < 1 indicate synergy, CI = 1 indicates additivity, and CI > 1 indicates antagonism [139].
Universal Surface Response Analysis: This statistical method provides a comprehensive estimate of differentiation between synergy, additivity, and antagonism across a range of concentration combinations, offering a more complete interaction profile than single-point measurements [139].
Table 2: Comparison of Major Methodologies for Synergy Detection
| Method | Key Principle | Output | Advantages | Limitations |
|---|---|---|---|---|
| Isobolographic Analysis [140] | Dose equivalence for specified effect level | Graphical representation (isobole) | Intuitive visualization; Clear interpretation | Limited to two compounds at a time; Fixed effect level |
| Combination Index (CI) [139] | Summation of fractional doses | Numerical index (CI < 1, =1, >1) | Quantitative results; Applicable to multiple compounds | Requires full dose-response curves for each agent |
| Universal Surface Response [139] | Statistical modeling of response surface | Three-dimensional interaction profile | Comprehensive across concentration ranges | Complex experimental design and analysis |
A systematic approach to screening synergistic interactions in natural product extracts ensures comprehensive and reproducible results. The following workflow outlines key stages in this process:
Diagram 2: Systematic workflow for screening synergistic interactions
Table 3: Essential Research Reagents and Materials for Synergy Studies
| Reagent/Material | Function in Synergy Research | Application Notes |
|---|---|---|
| Standardized Natural Extracts | Provides consistent, reproducible starting material for combination studies | Chemical fingerprinting recommended; Source and batch documentation critical [138] |
| Purified Bioactive Compounds | Enables controlled combination studies with known constituents | High purity (>95%) essential for accurate dose-response characterization [138] |
| Cell-Based Assay Systems | Initial screening platform for combination effects | Include relevant cell lines; Multiple assay endpoints recommended [138] [139] |
| Reference Standards (e.g., controls, calibration standards) | Ensures analytical validity and experimental consistency | Include positive/negative controls for biological assays [140] |
| Analytical Grade Solvents | Extraction, purification, and solubilization of compounds | Low UV cutoff for HPLC applications; Mass spectrometry compatibility [138] |
The therapeutic advantage of multi-compound extracts often derives from their ability to simultaneously modulate multiple interconnected signaling pathways, creating a network response that is difficult to achieve with single compounds. The following diagram illustrates key pathways frequently engaged by synergistic natural product combinations:
Diagram 3: Key signaling pathways modulated by synergistic natural product combinations
Research has demonstrated that medicinal plant extracts target biological systems through the combined action of structurally and functionally diverse active compounds that modulate complex cellular networks [138]. This multi-target approach is particularly evident in antimicrobial applications, where combinations of bioactive compounds can simultaneously disrupt cell membrane integrity, inhibit essential enzymes, and impair cellular energy production, resulting in enhanced efficacy and reduced potential for resistance development [138]. The polyvalent nature of these extracts denotes an improved and cooperative effect that cannot be easily attributed to single mechanisms [138].
Natural product combinations show significant promise in addressing the growing challenge of antimicrobial resistance. Studies have demonstrated that whole plant preparations are frequently more effective than isolated compounds due to synergistic interactions between constituents within them [138]. These combinations can produce enhanced antimicrobial effects through several mechanisms:
Research on medicinal plant extracts has revealed that disease resistance is less likely to occur against a combination of bioactive compounds than against single active molecules, highlighting the strategic advantage of multi-component antimicrobial approaches [138].
Combinations of marine and plant-derived bioactive compounds have demonstrated significant potential for managing chronic non-communicable diseases, including metabolic, inflammatory, and age-related conditions [139]. These combinations work through several synergistic mechanisms:
The development of marine-based functional foods and nutraceuticals represents a particularly promising application, with research showing that combinations of marine bioactive compounds can produce synergistic effects that enhance their preventive and therapeutic potential against chronic diseases [139].
The study of synergistic effects in multi-compound natural extracts represents a paradigm shift in natural product-based drug discovery, moving from reductionist single-compponent approaches to a more holistic systems-level understanding. The chemical multiverse concept provides a valuable framework for navigating the complex chemical space of natural products, acknowledging that comprehensive assessment requires multiple complementary representations of chemical structure and properties [137]. This approach aligns with the inherent complexity of natural extracts, whose therapeutic advantages frequently emerge from synergistic interactions between their numerous constituents [138].
Future research in this field should prioritize several key areas: First, developing more sophisticated computational models to predict synergistic interactions based on chemical structures and known biological activities. Second, advancing analytical techniques, particularly metabolomics approaches, to better characterize complex mixtures and identify interaction networks [138]. Third, establishing standardized methodologies and reporting guidelines for synergy research to improve reproducibility and comparability across studies [140] [139]. Finally, increasing efforts to translate in vitro synergy findings to validated clinical outcomes, particularly for complex chronic conditions where multi-target approaches offer distinct advantages over monotherapies [138] [139].
As drug discovery faces increasing challenges with single-target approaches, the strategic exploration of synergistic multi-compound extracts within the natural product chemical multiverse offers a promising path forward. By embracing the complexity of natural extracts rather than attempting to reduce it, researchers can harness the full therapeutic potential of these evolved chemical systems, potentially leading to more effective, safer, and more resistance-resistant therapeutic interventions.
The exploration of the natural product chemical space remains a cornerstone of innovative drug discovery, uniquely positioned to deliver the structural diversity and biological relevance needed to tackle complex diseases. The integration of foundational concepts with advanced methodologiesâfrom AI and high-throughput screening to synthetic biologyâis successfully overcoming historical challenges of supply and characterization. The proven track record of NPs, particularly in generating leads for antimicrobial and anticancer agents, validates their continued strategic importance. Future success hinges on a collaborative, multidisciplinary approach that fully embraces technological revolutions, adheres to ethical and regulatory frameworks, and systematically populates the underexplored regions of the biologically relevant chemical space to usher in a new era of therapeutics.