Fact-checked by Grok 2 weeks ago

Bioconjugation

Bioconjugation is the chemical process of covalently linking a , such as a protein, , , or , to another molecule—often a synthetic compound, label, or surface—to create hybrid structures with enhanced or novel properties while preserving the biomolecule's . This technique relies on selective reactions under mild, aqueous conditions to avoid disrupting native structures, enabling precise modifications that combine the specificity of with the versatility of . The field has evolved from early nonspecific couplings in the mid-20th century to modern site-specific methods, driven by advances in bioorthogonal chemistry that allow reactions orthogonal to endogenous cellular components. Key approaches include thiol-maleimide chemistry, which targets cysteine residues for stable thioether bonds; amide bond formation via native chemical ligation or enzymatic methods; and click chemistry, such as copper-catalyzed azide-alkyne cycloaddition (CuAAC) for efficient triazole linkages. Other prominent techniques encompass oxime and hydrazone formations from aldehyde/ketone and amine/aminooxy groups, as well as strain-promoted azide-alkyne cycloaddition (SPAAC) for metal-free conjugation. These methods prioritize chemoselectivity (targeting specific functional groups) and site-selectivity (focusing on particular residues), often using genetically engineered proteins with unnatural amino acids to achieve homogeneity in conjugates. Bioconjugation plays a pivotal role across biotechnology and medicine, facilitating applications like antibody-drug conjugates (ADCs) for targeted cancer therapy, where toxins are precisely attached to monoclonal antibodies; biomolecular imaging with fluorescent or radiolabeled probes for diagnostics; and drug delivery systems that improve pharmacokinetics and bioavailability. In materials science, it enables the functionalization of nanoparticles, hydrogels, and biosensors for tissue engineering and point-of-care detection. Ongoing innovations, such as photo- or enzyme-mediated conjugations, continue to expand its utility in proteomics, vaccine development, and sustainable biocatalysis.

Introduction

Definition and Scope

Bioconjugation refers to the covalent attachment of synthetic or biological entities to , such as proteins, nucleic acids, or carbohydrates, to form stable hybrid constructs. This process typically involves linking functional groups, labels, or therapeutic agents to specific sites on the biomolecule, enabling the creation of multifunctional materials while leveraging the inherent biological properties of the native component. The scope of bioconjugation spans diverse applications in and , including , , and the development of advanced biomaterials. For instance, in , bioconjugation facilitates the precise tethering of cytotoxic payloads to targeting moieties, enhancing therapeutic efficacy and reducing off-target effects. In imaging, it allows the attachment of fluorescent or radioactive labels to biomolecules for visualization . Biomaterials benefit from bioconjugation by incorporating bioactive elements that promote or tissue regeneration. Unlike non-covalent interactions, such as the high-affinity biotin-streptavidin (with a dissociation constant of approximately 10^{-15} M), bioconjugation forms irreversible covalent bonds that ensure long-term stability under physiological conditions. Central to bioconjugation are reactive handles—intrinsic or engineered chemical groups on biomolecules (e.g., or residues on proteins)—that selectively react with complementary functionalities on the conjugate partner. These reactions must be designed to minimize disruption to the biomolecule's structure and function, preserving its native activity, such as enzymatic or antigen recognition. A prominent example is antibody-drug conjugates (ADCs), where monoclonal antibodies are covalently linked to potent drugs via linkers, enabling targeted cancer therapy; as of 2025, over 15 ADCs have received FDA regulatory approval, demonstrating the clinical impact of this approach. , which employs mutually reactive groups inert to biological milieus, further expands the scope by allowing site-specific conjugation without interfering with endogenous processes.

Historical Development

The field of bioconjugation originated in the mid-20th century with early efforts to modify proteins for radiolabeling and structural studies. In the 1960s, iodination of residues emerged as a primary technique for introducing radioactive iodine (e.g., I-125 or I-131) into proteins, enabling tracking in biological systems. This method, developed by Hunter and Greenwood in 1962 using as an oxidizing agent, allowed high-specific-activity labeling of proteins like human growth hormone while preserving biological activity. Concurrently, acylation gained traction for radiolabeling, with Means and Feeney introducing reductive in 1968, which involved reacting ε-amino groups with and to attach labels without disrupting protein charge. These approaches, though non-specific and often leading to heterogeneous modifications, laid the groundwork for protein derivatization in diagnostics and research. The and marked a shift toward more targeted and therapeutic applications, driven by advances in and chemistry. Thiol-maleimide conjugation rose prominently for residues, building on earlier uses from the but gaining widespread adoption in the for its selectivity under mild conditions; the between thiols and maleimides formed stable thiosuccinimide linkages, facilitating antibody labeling and drug attachment. Parallel to this, —covalent attachment of () to proteins—emerged as a strategy to enhance and reduce . Pioneered by Abuchowski et al. in 1977 using to link PEG to amines on , this technique saw therapeutic breakthroughs in the , with the first FDA-approved PEGylated protein, Adagen (pegademase bovine), launched in 1990 for . Entering the 2000s, bioconjugation evolved toward precision and bioorthogonality, spurred by the advent of . The concept of click reactions, coined by Sharpless in , emphasized modular, high-yield ligations; this culminated in with independent reports from Meldal and Sharpless on copper-catalyzed azide-alkyne cycloaddition (CuAAC), enabling efficient formation between azides and terminal alkynes for site-specific protein modifications. Bioorthogonal ligations, such as strain-promoted azide-alkyne cycloadditions developed by Bertozzi in the early 2000s, further expanded copper-free options for applications. Key milestones included the 2000 FDA approval of , the first antibody-drug conjugate () utilizing a linker for delivery in , demonstrating bioconjugation's clinical potential. Additionally, genetic code expansion enabled incorporation of unnatural bearing bioorthogonal handles, with Wang et al. achieving amber suppression in E. coli for analogs in , paving the way for precise conjugations in the 2000s. These innovations transitioned bioconjugation from rudimentary labeling to sophisticated therapeutic tools. These advancements were recognized with the 2022 awarded to Bertozzi, Meldal, and Sharpless for their contributions to click and .

Fundamental Principles

Chemical Reactivity in Biomolecules

Biomolecules exhibit inherent chemical reactivity through specific functional groups that enable selective modifications in bioconjugation processes. In proteins, side chains serve as primary reactive sites, categorized by their nucleophilic or electrophilic nature. groups predominate, including the ε-amine on residues (pKa ≈ 10.5), which acts as a strong when deprotonated, and the on residues (pKa ≈ 8.3), which is particularly reactive under near-physiological conditions due to its low pKa allowing partial . The phenolic hydroxyl of (pKa ≈ 10.1) provides moderate nucleophilicity upon deprotonation, while polar hydroxyl groups on serine (pKa ≈ 13) are less reactive and infrequently targeted owing to their high pKa values that limit deprotonation in aqueous environments. These properties stem from the electronic characteristics of the side chains, making them susceptible to electrophilic conjugating agents without requiring additional activation. The reactivity of these functional groups is modulated by the structural context of the . In proteins, secondary structures such as α-helices, β-sheets, and random coils influence accessibility, which directly impacts conjugation efficiency. For example, s in compact β-sheets display approximately 10% solvent accessibility, burying nucleophilic sites like those on lysines and cysteines, whereas residues in flexible random coils or turns exhibit up to 30% accessibility, enhancing their exposure to reagents. This solvent-accessible surface area (ASA) variation arises from hydrogen bonding and hydrophobic packing in secondary elements, restricting reactivity in buried regions while favoring surface-exposed residues. In nucleic acids, reactivity arises from nucleobases, where exocyclic amines on and serve as nucleophiles, and the backbone acts as an , particularly at 5'- or 3'-termini, allowing site-specific attachments under mild conditions. For carbohydrates, reactivity primarily involves multiple hydroxyl groups, which are weakly nucleophilic and often require activation (e.g., via oxidation to generate aldehydes for / formation) or enzymatic guidance for selectivity; amines in amino sugars like provide additional nucleophilic sites. In lipids, conjugation typically targets polar head groups, such as primary amines in (pKa ≈ 9-10) or carboxyl groups in fatty acids, which act as nucleophiles or electrophiles respectively, though amphiphilic nature influences solubility and reaction conditions in aqueous media. Stability considerations are paramount to preserve biomolecular integrity during reactivity exploitation. Conjugation reactions must operate under conditions that avoid denaturation, such as neutral and aqueous solvents that mimic physiological environments. The influences states—for instance, at pH 7.4, cysteines are more nucleophilic than lysines, which remain largely until pH >9—while solvent polarity affects of charged groups, with promoting native folding but potentially slowing reactions compared to mixed solvents. Deviations in pH or harsh solvents can disrupt secondary structures, reducing accessibility and leading to off-target modifications or loss of function.

Site-Selectivity Strategies

Bioconjugation methods can be broadly classified into random and site-specific approaches. Random conjugation, which targets abundant reactive residues such as lysines or cysteines in proteins, often results in heterogeneous products with variable conjugation sites and stoichiometries, leading to polydisperse mixtures that complicate downstream applications and reduce reproducibility. In contrast, site-specific strategies enable precise modification at predefined locations, yielding homogeneous conjugates with defined structures, which is essential for maintaining protein function and achieving consistent biological properties. These approaches mitigate the heterogeneity inherent in natural reactions, such as those involving multiple lysines or cysteines, by introducing unique reactive handles. One prominent site-specific strategy involves the genetic incorporation of unnatural amino acids (UAAs) bearing orthogonal reactive groups, such as azides, alkynes, or ketones, into proteins during biosynthesis. This is achieved through genetic code expansion, where the UAA is site-specifically inserted at an amber (TAG) stop codon using an orthogonal aminoacyl-tRNA synthetase (aaRS)/tRNA pair that does not cross-react with endogenous machinery. A seminal example is the incorporation of p-acetyl-L-phenylalanine (pAcPhe), a ketone-containing UAA, into proteins in Escherichia coli with high fidelity (>99% suppression efficiency) and yields up to 3.6 mg/L. The ketone group of pAcPhe enables selective bioconjugation via hydrazone or oxime formation with high efficiency (80-90%), allowing attachment of labels or payloads without affecting native residues. Another key strategy is protein engineering to introduce unique reactive residues, such as a single unpaired cysteine, by mutating the protein sequence to eliminate competing sites while preserving structure and function. This approach, exemplified by THIOMAB technology, positions the cysteine at solvent-exposed locations on antibodies, enabling quantitative, site-specific thiol-based conjugation and producing conjugates with uniform drug-to-protein ratios. Amber suppression is a cornerstone tool for UAA incorporation, relying on engineered orthogonal tRNA/aaRS pairs derived from archaeal or sources to decode the amber codon specifically in the presence of the UAA, bypassing competition. This method has been optimized for efficiency in various expression systems, including and mammalian cells, achieving suppression efficiencies up to 50-70% at permissive sites. For specificity, proximity labeling strategies exploit spatial constraints to direct conjugation to nearby residues, enhancing in complex environments. These include proximity-induced (PIC), such as using engineered cysteine-lysine pairs to form stable cyclic linkages via bis-electrophiles like dichloro butenediamides, with site-selectivity levels exceeding 90% in proteins like . Such methods enable targeted modifications in cellular contexts by leveraging local microenvironments or templated assembly. Site-specific strategies significantly improve conjugate homogeneity, often achieving >90% monodisperse products compared to <50% for random methods, as measured by metrics like the homogeneity index (ratio of desired to undesired species via mass spectrometry). This uniformity positively impacts pharmacokinetics, with site-specific conjugates exhibiting prolonged half-lives (e.g., 2-3 fold improvements in circulation time) and reduced clearance due to minimized aggregation and immunogenicity risks. In engineered cysteine systems, such homogeneity correlates with enhanced stability and predictable biodistribution, underscoring the advantages over polydisperse random conjugates.

Bioconjugation Reactions on Natural Amino Acids

Lysine-Targeted Reactions

Lysine residues, featuring a nucleophilic ε-amine group with a pKa around 10.5, are common targets in bioconjugation due to their abundance, typically comprising 5-7% of amino acids in eukaryotic proteins. This prevalence enables straightforward modification but complicates site-selectivity, as multiple lysines can react, resulting in heterogeneous products that may impair protein function or uniformity in applications like antibody-drug conjugates. Strategies to mitigate this often draw from broader site-selectivity principles, such as pH control or engineered variants, though lysine reactions remain widely used for their simplicity and compatibility with aqueous conditions. The most established lysine-targeted reaction is amidation with N-hydroxysuccinimide (NHS) esters, introduced by Anderson et al. in 1964 as an active ester for efficient peptide bond formation. In this process, the deprotonated ε-amine of lysine acts as a nucleophile, attacking the carbonyl carbon of the NHS-activated carboxylic acid in an acyl substitution reaction; this displaces the NHS leaving group, forming a stable amide linkage under mild, near-physiological conditions (pH 7-8.5). The reaction proceeds rapidly, often completing in minutes to hours, with yields exceeding 80% for many proteins. A representative equation is: \ce{R-C(O)-O-NHS + H2N-Lys -> R-C(O)-NH-Lys + HO-NHS} where R represents the conjugating moiety, such as a fluorophore or drug linker; sulfo-NHS variants enhance water solubility for better reactivity in biological media. Despite these advantages, the method's non-specificity can lead to over-modification, altering protein charge and potentially causing aggregation, necessitating optimization for each target protein. Alternative approaches include isothiocyanate-mediated conjugation, which forms thiourea bonds via nucleophilic addition of the lysine amine to the electrophilic carbon of the isothiocyanate (R-N=C=S), yielding R-NH-C(S)-NH-Lys without requiring activation steps. This reaction is favored for its stability and has been employed since the early 20th century in protein labeling, though it proceeds more slowly than NHS chemistry (hours to days) and can be sensitive to pH. Another method is reductive amination, where an aldehyde (R-CHO) condenses with the lysine amine to form a reversible Schiff base imine, which is then reduced by NaBH₄ or similar agents to a stable secondary amine (R-CH₂-NH-Lys); this approach preserves positive charge on unmodified lysines but requires careful control to avoid over-reduction or side reactions with other nucleophiles. Both methods leverage lysine's abundance for high loading efficiency but share the selectivity challenges inherent to amine reactivity.

Cysteine-Targeted Reactions

Cysteine residues in proteins are prime targets for bioconjugation due to the nucleophilic (-SH) group in their side chains, which exhibits high reactivity under physiological conditions. This reactivity stems from the low of the (approximately 8.3), enabling to form a thiolate that acts as a strong . Unlike more abundant residues like , cysteines are typically fewer in number and often involved in bonds, allowing for site-selective modifications when free thiols are available or engineered. The primary and most widely adopted cysteine-targeted reaction is the thiol-maleimide Michael addition, first demonstrated for protein cross-linking in 1956. In this reaction, the thiolate adds across the electron-deficient of the maleimide, forming a stable thiosuccinimide linkage. The proceeds via nucleophilic attack by the thiolate on the β-carbon of the maleimide, followed by proton transfer and ring closure to yield the adduct. This conjugate addition is highly efficient, with second-order rate constants typically ranging from 10² to 10⁴ M⁻¹ s⁻¹ at neutral pH, making it suitable for mild, aqueous conditions. The reaction can be represented as: \text{R-Maleimide} + \text{HS-Cys} \rightarrow \text{R-thioether-Cys} where R denotes the maleimide substituent and the product is a thioether-linked conjugate. This method's advantages include rapid kinetics and compatibility with biomolecules, but the thiosuccinimide can undergo retro-Michael elimination or , particularly under reducing conditions, leading to instability. Additionally, thiols are prone to oxidation, forming disulfides that reduce conjugation efficiency. Reversible variants, such as those incorporating cleavable linkers, mitigate some limitations by allowing controlled release. Alternative cysteine-targeted approaches include , a classic where the thiolate displaces iodide from , yielding a carboxamidomethylated . This method, routinely used since the mid-20th century in protein chemistry, provides irreversible modification with high specificity but slower rates than maleimide chemistry and potential over- if excess is employed. offers a reversible option, involving nucleophilic attack by a protein on an activated (e.g., pyridyl ), exchanging the thiol partners and forming a new bond. This equilibrium-driven process is useful for dynamic conjugations, though it requires optimization to favor the desired product. To enhance site-selectivity, free cysteines are often introduced via , replacing non-essential residues with while mutating native s to avoid heterogeneity. This engineering strategy enables precise placement of conjugation sites, improving homogeneity in applications like antibody-drug conjugates. For instance, THIOMAB technology uses such mutations to generate defined variants for targeted modifications.

Tyrosine-Targeted Reactions

Tyrosine residues, with their phenolic side chains, offer unique opportunities for bioconjugation through or oxidative activation, enabling the attachment of diverse functional groups to proteins and peptides. These reactions primarily target the electron-rich aromatic ring of , distinguishing them from nucleophilic modifications of other . Unlike more abundant residues, tyrosines are often surface-exposed, facilitating selective labeling in native biomolecular contexts. A primary method involves diazonium coupling, where aryldiazonium salts (Ar-N_2^+) act as electrophiles in an azo-coupling reaction with the phenol. This proceeds via at the ortho or para position relative to the hydroxyl group, forming stable azo linkages (Ar-N=N-Tyr). The reaction is rapid under mildly acidic aqueous conditions and has been applied to label surface tyrosines on bacteriophages and peptides for diagnostic purposes. For instance, diazonium reagents bearing fluorescent dyes enable site-specific visualization of tyrosine reactivity in proteomes. However, the acidic and potential for over-modification can disrupt , limiting . Enzymatic approaches, such as tyrosinase-mediated oxidation, convert exposed tyrosines to reactive o-quinones, which then undergo with thiols, amines, or for conjugation. , a copper-dependent , catalyzes the sequential and oxidation of to dopaquinone intermediates, enabling mild, site-selective modifications on protein N- or C-termini tagged with . This method has been used to couple peptides or fluorophores to antibodies, preserving bioactivity due to physiological conditions ( 6-7, ambient temperature). Despite advantages in specificity, the enzyme's bulkiness may hinder access to buried residues. Metal-catalyzed C-H arylation provides another route, typically using complexes to functionalize the ortho position with aryl halides via directed C-H activation. This installs diverse aryl groups, expanding applications in peptide stapling or drug conjugation, and can be extended from broader -mediated strategies. While offering high , these reactions often require organic co-solvents or ligands that may denature sensitive proteins. Overall, -targeted methods excel in labeling surface residues for or therapeutics but demand optimization to mitigate structural perturbations.

Terminal Residue Reactions

Terminal residue reactions in bioconjugation target the N- or C-termini of proteins and peptides, providing a strategy for site-specific modification with minimal disruption to the biomolecule's overall structure and function. These approaches leverage the unique reactivity of the α-amine at the or the at the , which are present as a single site per polypeptide chain in unmodified proteins. This specificity arises from the distinct chemical environments of terminal residues compared to internal side chains, enabling controlled conjugation under mild aqueous conditions. At the , the primary α-amine (pKa ~7–9) reacts with to form reversible , which are subsequently reduced to stable secondary amines via . Common reagents include benzaldehydes or 2-pyridinecarboxaldehydes, with (NaBH₃CN) serving as the reducing agent to selectively trap the imine without over-reduction. This method achieves high selectivity at mildly acidic (e.g., 6–7.5), where the N-terminal amine is more nucleophilic than ε-amines (pKa ~10.5). Yields can range from 10–90% depending on the , protein, and conditions, for example 10% for RNase A and 46% for , as demonstrated in modifications of model proteins. The reaction proceeds as follows: \text{R-NH}_2 + \text{R'-CHO} \rightleftharpoons \text{R-N=CH-R'} + \text{H}_2\text{O} followed by reduction: \text{R-N=CH-R'} + \text{NaBH}_3\text{CN} \rightarrow \text{R-NH-CH}_2\text{-R'} where R represents the protein N-terminus and R' the conjugating moiety. For the C-terminus, the carboxylic acid is activated using carbodiimide chemistry, typically with 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) and N-hydroxysuccinimide (NHS), to form a reactive NHS ester intermediate. This ester then couples with a primary amine (e.g., on a payload molecule) to yield a stable amide bond. The activation step generates an O-acylisourea intermediate from EDC, which NHS stabilizes against hydrolysis, enabling efficient conjugation in aqueous buffers at pH 5–7. This approach has been widely applied to peptides and proteins, such as immobilizing C-terminal peptides on surfaces or linking to nanomaterials. The overall C-terminal coupling reaction is: \text{R-COOH} + \text{H}_2\text{N-R'} \xrightarrow{\text{EDC/NHS}} \text{R-CONH-R'} + \text{byproducts} where R is the protein C-terminus and R' the amine-containing conjugate. A key advantage of terminal residue reactions is the availability of only one reactive site per chain, facilitating homogeneous bioconjugates without the heterogeneity issues of side-chain targeting. This is particularly beneficial for maintaining protein folding and activity, as modifications occur away from the core structure. However, these methods require proteins or peptides with unprotected, accessible termini, often necessitating recombinant expression without affinity tags or post-expression processing to expose free ends. In and , terminal residue reactions enable the assembly of larger constructs by coupling C-terminal carboxylic acids to N-terminal amines of complementary segments, forming native-like bonds essential for total of proteins. For instance, EDC/NHS-mediated couplings have been used to ligate peptide fragments in the synthesis of bioactive sequences, while N-terminal supports sequential modifications in multi-step assemblies.

Bioorthogonal and Strain-Promoted Reactions

Azide-Based Cycloadditions

Azide-based cycloadditions represent a cornerstone of in bioconjugation, enabling the selective linkage of biomolecules under physiological conditions without interference from native cellular components. These reactions primarily involve the 1,3-dipolar cycloaddition between azides and alkynes to form stable 1,2,3-triazole linkages, which mimic bonds in stability and provide a versatile handle for further modifications. Introduced in the early , these methods have revolutionized protein labeling, conjugation, and material applications by offering high specificity and efficiency. The (I)-catalyzed - cycloaddition (CuAAC), developed independently by the Meldal and Sharpless groups in , accelerates the otherwise slow thermal Huisgen cycloaddition to proceed rapidly in aqueous media at . In this process, Cu(I) coordinates to the terminal , forming a copper acetylide intermediate that reacts stepwise with the : first via nucleophilic attack on the coordinated , followed by cyclization and to yield the 1,4-regioisomer of the product. The reaction is highly efficient, often achieving near-quantitative yields in minutes, and tolerates a wide range of functional groups, making it ideal for bioconjugation. The general equation for CuAAC is: \mathrm{R-N_3 + R'-C \equiv CH \xrightarrow{Cu(I)}} \mathrm{1(R)-4(R')-1,2,3-triazole} where the triazole ring provides a rigid, bioorthogonal linker. However, the need for copper catalysis limits its use in vivo due to potential toxicity, though ligand-stabilized variants mitigate this for certain applications. To address copper toxicity, strain-promoted azide-alkyne cycloaddition (SPAAC) was introduced by the Bertozzi group in 2004, utilizing cyclooctynes bearing ring strain to drive the reaction without catalysts. The strained triple bond in cyclooctynes, such as difluorobenzocyclooctyne (DIBO), lowers the activation energy for the [3+2] cycloaddition with azides, proceeding via a concerted mechanism involving distortion of the cyclooctyne ring and azide approach to form the triazole. SPAAC reactions occur selectively in biological environments, with second-order rate constants around 1 M⁻¹ s⁻¹, enabling live-cell imaging and in vivo labeling. Examples include DIBO derivatives, which enhance reactivity through electron-withdrawing substituents, allowing efficient conjugation of azide-modified glycans or proteins. Azides for these cycloadditions are commonly introduced into biomolecules via genetic encoding of unnatural , such as p-azidophenylalanine (pAzF), which can be site-specifically incorporated into proteins using orthogonal tRNA/synthetase pairs in or eukaryotic systems. This amber suppression strategy, first demonstrated in 2002, allows precise placement of the at targeted residues, facilitating subsequent CuAAC or SPAAC with alkyne-functionalized probes like fluorophores or drugs. The bioorthogonality of azide-alkyne pairs ensures minimal off-target reactions, with applications spanning targeted therapeutics to nanoscale assemblies.

Ketone and Aldehyde Modifications

Ketone and aldehyde modifications in bioconjugation primarily involve the formation of oximes and hydrazones through the reaction of carbonyl groups with aminooxy or hydrazine nucleophiles, respectively. These reactions proceed via a condensation mechanism where the nucleophile attacks the carbonyl carbon, forming a tetrahedral hemiaminal intermediate, followed by proton-catalyzed dehydration to yield the stable C=N linkage. The general reaction for oxime formation is represented as: \text{R-CHO} + \text{H}_2\text{N-OR'} \rightarrow \text{R-CH=N-OR'} + \text{H}_2\text{O} where R is the biomolecule-linked aldehyde and R' is typically a methyl or functionalized group on the aminooxy reagent. Hydrazone formation follows an analogous pathway using hydrazines (H₂N-NHR'), resulting in R-CH=NNHR' products. These modifications are bioorthogonal, occurring selectively in biological environments due to the scarcity of free aldehydes and ketones in native biomolecules, which minimizes off-target reactions. Oximes exhibit greater hydrolytic stability compared to hydrazones, with half-lives often exceeding 180 hours at physiological pH 7.4, while hydrazones can hydrolyze more readily under acidic conditions (e.g., half-life ~4 hours at pH 5), enabling tunable release profiles for drug delivery applications. Reaction rates are pH-dependent, accelerating at mildly acidic to neutral pH (optimal around 4-7) due to proton catalysis of the dehydration step, though uncatalyzed rates can be slow (second-order rate constants ~0.1-1 M⁻¹ min⁻¹ at pH 7). Carbonyl groups for these reactions are incorporated site-specifically into biomolecules, such as proteins or peptides. Ketones are commonly introduced via , which can be appended to protein termini or side chains using enzymatic (e.g., sortase-mediated) or during solid-phase , providing a ketone handle for selective modification. Aldehydes are generated through conversion of residues to formylglycine using the formylglycine-generating (FGE), enabling precise placement at genetically encoded sites without disrupting . This approach has been applied to conjugate fluorophores or drugs to antibodies, achieving high yields (>90%) under mild aqueous conditions. To address the inherently slow of these ligations, especially at neutral , catalyzed variants have been developed. acts as a nucleophilic catalyst by forming a reactive intermediate with the carbonyl, accelerating and formation up to 400-fold (e.g., rate constants increasing from 0.01 M⁻¹ min⁻¹ to 4 M⁻¹ min⁻¹ at 4.5). More advanced bifunctional catalysts, such as m-phenylenediamine derivatives, further enhance rates at physiological (up to 6-fold improvement), broadening applicability . Aldehydes can also arise from oxidation of N-terminal serines or threonines in proteins, providing a entry point for these modifications.

Staudinger Ligation

The Staudinger ligation is a bioorthogonal that enables the formation of stable bonds between an azide-functionalized and a phosphine-bearing acyl donor under physiological conditions. Developed in 2000 by Carolyn R. Bertozzi and colleagues, it adapts the classic Staudinger reduction—originally described by in 1919—to create covalent links without interfering with native biological processes. This method has become a cornerstone of for site-specific labeling and conjugation. The reaction mechanism begins with the nucleophilic attack of a on the terminal of the (R-N₃), forming a phosphazide intermediate that rapidly extrudes N₂ to yield an iminophosphorane (R-N=PPh₃). In the standard Staudinger , this iminophosphorane then undergoes in aqueous media to generate an (R-NH₂) and oxide (Ph₃P=O); however, for , the phosphine is engineered with an adjacent or group that facilitates intramolecular acyl transfer from the iminophosphorane to form the bond (R-NH-C(O)-R'). This process ensures , as azides and phosphines are absent in natural systems. A key variant is the traceless Staudinger ligation, introduced concurrently in 2000 by Ronald T. Raines and coworkers, which employs a phosphinothioester (R'-C(O)-S-PPh₂) to enable direct formation without incorporating phosphine-derived residues into the product. Here, the iminophosphorane attacks the thioester carbonyl, forming a tetrahedral that collapses to release the native (R-NH-C(O)-R') and (Ph₃P=O), mimicking a . The overall simplified reaction is represented as: \text{R-N}_3 + \text{R'-C(O)-X-PPh}_2 + \text{H}_2\text{O} \rightarrow \text{R-NH-C(O)-R'} + \text{Ph}_3\text{P=O} + \text{XH} where X is S or O. This variant proceeds with second-order rate constants around 10⁻³ M⁻¹ s⁻¹, suitable for biological applications. The primary advantages of the Staudinger ligation include its metal-free nature, , and ability to produce native-like linkages, which are stable and hydrolytically resistant . Unlike reductive processes, it avoids byproducts that could disrupt cellular function, making it ideal for selective bioconjugation. Azides for this reaction are often introduced metabolically into glycans or proteins. In applications, the Staudinger ligation has enabled in vivo protein labeling, such as tagging cell-surface sialic acids in living mice by administering azido-modified sugars (e.g., Ac₄ManNAz) followed by probes, allowing visualization of metabolic pathways without toxicity. It has also facilitated the synthesis of antibody-drug conjugates and protein by uniting fragments.

Transition Metal-Catalyzed Reactions

Palladium-Mediated Arylation

Palladium-mediated arylation represents a powerful class of transition metal-catalyzed reactions for installing aryl groups onto biomolecules through carbon-carbon bond formation, particularly targeting residues in peptides, proteins, and nucleic acids. These methods leverage classic cross-coupling chemistries adapted for aqueous, biocompatible conditions to enable site-specific modifications that introduce functional handles such as fluorophores, affinity tags, or therapeutic moieties. By extending traditional to biological contexts, facilitates the creation of complex bioconjugates with enhanced properties for applications in imaging and . The Suzuki-Miyaura reaction stands as the most widely adopted palladium-mediated arylation strategy in bioconjugation, involving the coupling of an aryl or heteroaryl boronic acid with a halogenated biomolecule, typically a tyrosine derivative bearing an iodine or bromine substituent at the ortho position of the phenolic ring. This reaction proceeds under mild aqueous conditions using Pd(OAc)2 as the catalyst precursor and ligands such as 2-amino-4,6-dihydroxypyrimidine to enhance solubility and activity. In unprotected peptides, iodinated tyrosine or phenylalanine residues undergo efficient arylation with arylboronic acids in water at room temperature, achieving yields up to 90% without protecting groups. Similarly, Davis and coworkers in 2009 applied this chemistry to site-specifically modify a cysteine-mutated protein (subtilisin Bacillus lentus) via a genetically incorporated iodotyrosine analog, coupling diverse arylboronic acids in >95% conversion within 30 minutes at 37°C and pH 8.0. The general reaction can be represented as: \text{Ar-B(OH)}_2 + \text{Tyr-X} \xrightarrow{\text{Pd catalyst, base, H}_2\text{O}} \text{Ar-Tyr} + \text{HX} + \text{B(OH)}_3 where Ar-B(OH)2 is an aryl or heteroaryl boronic acid and X is a halide substituent (typically iodine or bromine) on the tyrosine residue. These approaches build on prior tyrosine-targeted halogenation strategies to enable selective modification of natural or genetically encoded residues. The Heck coupling, another palladium-catalyzed process, facilitates arylation through the reaction of an aryl halide with an alkene-functionalized biomolecule, typically yielding a substituted alkene product via syn addition and β-hydride elimination. Adapted for bioconjugation, this method has been employed to modify proteins containing alkene-bearing unnatural amino acids, such as styryl-modified lysozyme. In 2011, Myers and coworkers developed storable arylpalladium(II) reagents that enable Heck-type couplings in aqueous media at room temperature, achieving approximately 75% conversion on the lysozyme substrate without significant protein denaturation. This variant uses preformed Pd(II)-aryl complexes to bypass oxidative addition steps, enhancing compatibility with sensitive biomolecules. The mechanistic cycle of palladium-mediated arylations, common to both Suzuki-Miyaura and Heck reactions, begins with oxidative addition of the aryl halide to Pd(0), forming an aryl-Pd(II)-halide intermediate. This is followed by transmetalation—in the case of Suzuki-Miyaura, with the boronic acid partner facilitated by a base—or coordination and insertion of the alkene in Heck coupling. The cycle concludes with reductive elimination to release the arylated product and regenerate Pd(0). These steps occur under ligand modulation to maintain catalytic turnover in aqueous environments, as elucidated in foundational studies on palladium catalysis. A key advantage of palladium-mediated arylation lies in its ability to introduce structurally diverse and complex aryl groups, including heterocycles and functional tags, that are challenging to access via nucleophilic additions or other bioconjugation methods. This enables precise tuning of properties, such as or binding affinity, in contexts ranging from natural residues (post-halogenation) to unnatural incorporated via . However, a primary disadvantage is the inherent of species, which can interfere with and necessitate post-reaction purification strategies like chelation resins or to remove residual metal below cytotoxic thresholds (typically <1 μM). Despite these challenges, ongoing ligand and catalyst innovations continue to mitigate toxicity while preserving efficiency.

Copper-Free and Metal-Free Alternatives

To address the toxicity concerns associated with copper in azide-alkyne cycloaddition (CuAAC) reactions, refinements have focused on ligand-stabilized copper catalysts that enhance reaction efficiency while minimizing oxidative damage to biomolecules. The ligand tris(3-hydroxypropyltriazolylmethyl)amine (THPTA) stabilizes Cu(I) species, accelerates the cycloaddition by up to 100-fold compared to uncatalyzed rates, and acts as a sacrificial reductant to intercept reactive oxygen species generated during the reaction, thereby reducing cytotoxicity in cellular environments. Similarly, the water-soluble ligand THETA has been developed to bind Cu(I) effectively under aqueous conditions, enabling efficient bioconjugation of peptides and proteins with minimal metal residue and toxicity, as demonstrated in the synthesis of triazole-linked conjugates at neutral pH. These ligand systems allow CuAAC to proceed at low copper concentrations (e.g., 0.1 mM), making them more biocompatible for in vitro protein labeling and nucleic acid modifications compared to traditional setups. Metal-free alternatives to transition metal-catalyzed arylation have emerged, particularly photo- or strain-promoted methods that enable selective without catalyst toxicity, suitable for in vivo applications. For instance, 2-sulfonylpyrimidines facilitate metal-free S-arylation of cysteine residues under aqueous, neutral pH conditions, forming stable thioether linkages with reaction rates exceeding 1 M⁻¹ s⁻¹ and high selectivity in full proteins, avoiding the need for metal removal post-reaction. Photoinduced energy transfer enables metal-free amino(hetero)arylation of alkenes, allowing precise installation of aryl groups on biomolecules via iminyl radical intermediates, with yields up to 90% in biological media and no observable off-target reactivity. These approaches contrast with copper-based cycloadditions by eliminating metal involvement entirely, enhancing biocompatibility for therapeutic conjugates, as evidenced by their use in labeling cell-surface proteins without cellular disruption. Ruthenium complexes offer specific advantages in photolabile bioconjugation strategies, where light activation releases functional groups with spatiotemporal control. Ru(II) polypyridyl complexes, when conjugated to biomolecules via amide or click linkages, serve as photocages for drugs or probes, releasing payloads upon visible light irradiation (λ > 400 ) due to photodissociation, with quantum yields around 0.01–0.1 and minimal dark in cellular assays. For example, Ru(II)- bioconjugates enable targeted phototherapy, where UV-Vis light (450–550 ) cleaves photolabile ligands to activate anticancer agents selectively in tumor cells, improving efficacy over non-caged systems by 5–10 fold . Post-2010 developments have emphasized recycling to further reduce metal exposure in bioconjugation workflows. Copper-based heterogeneous s, such as Cu supported on or polymers, allow facile separation and reuse (up to 5–10 cycles) in CuAAC reactions, maintaining >95% yield while minimizing leaching to below 1 ppm, as shown in large-scale protein modifications. These recyclable systems enhance and , enabling in vivo-compatible conjugations like antibody-drug linkages with reduced environmental and toxicological footprints.

Enzymatic and Emerging Bioconjugation Methods

Enzyme-Mediated Ligation

Enzyme-mediated ligation employs biocatalysts such as transpeptidases to form covalent bonds between biomolecules under mild, physiological conditions, enabling site-specific modifications with minimal disruption to and function. This approach leverages the natural specificity of enzymes to target particular sequences or motifs, distinguishing it from purely chemical methods by avoiding harsh reagents and reducing off-target reactions. Key enzymes in this category include sortase A and transglutaminases, which catalyze transpeptidation or formation, respectively, facilitating applications in and therapeutic conjugate assembly. Sortase A (SrtA), derived from , is a calcium-dependent transpeptidase that recognizes the C-terminal LPXTG motif (where X is any ) on one and a nucleophilic N-terminal on another, cleaving the threonine- bond to form a new linkage. The reaction proceeds via a catalytic involving an active-site residue, resulting in the product and release of . This can be represented as: \text{LPXTG} + \text{R-NH}_2 \rightarrow \text{LPXT-R} + \text{NH}_2\text{-Gly} The specificity for the allows precise attachment of payloads, such as fluorophores or drugs, to recombinant proteins engineered with this , often at N- or C-termini for terminal residue conjugations. Microbial transglutaminases (mTGs), particularly from mobaraensis, offer an alternative by catalyzing the formation of isopeptide bonds between and residues without requiring a free , enabling crosslinking in native or partially denatured proteins. Unlike sortases, mTGs do not release a and can operate in the absence of calcium, broadening their utility for glutamine-targeted modifications in antibodies and peptides. The primary advantages of these enzyme-mediated methods lie in their compatibility with aqueous buffers at neutral and ambient temperatures, preserving the bioactivity of sensitive conjugates, and their inherent site-selectivity, which minimizes heterogeneity in product mixtures compared to chemical labeling. For instance, sortase A reactions achieve near-quantitative yields in hours under optimized conditions, supporting scalable bioconjugation for therapeutics.49004-1/fulltext) Since the , and rational engineering have expanded substrate scopes; variants like penta-mutant SrtA (e.g., P94R/D160N/D165A/K190E/K196T) enhance reaction rates by up to 140-fold and reduce calcium dependence, while reprogrammed versions accept non-canonical motifs such as LPXTA or LPXTS for greater versatility. Similarly, engineered mTGs with altered glutamine specificity have improved efficiency for site-specific antibody-drug conjugates, addressing limitations in wild-type selectivity. These advancements have solidified enzyme-mediated as a cornerstone for precise biomolecular assembly in .

Photoclick and Light-Induced Methods

Photoclick and light-induced methods in bioconjugation leverage photochemical reactions to achieve precise spatial and temporal control over biomolecular linkages, enabling on-demand activation under mild conditions without catalysts or enzymes. These approaches typically involve UV, visible, or near-infrared (NIR) light to trigger bond formation or deprotection, minimizing off-target reactions and enhancing biocompatibility in complex biological environments. Key advantages include the ability to direct conjugation to specific sites, such as within living cells or tissues, by focusing light irradiation, which reduces nonspecific interactions compared to constant reactivity in traditional bioorthogonal methods. A prominent strategy employs photocaged groups, where reactive functionalities are masked and released upon light exposure to initiate conjugation. For instance, o-nitrobenzyl (oNB) groups are commonly used to cage , protecting residues or thiol-containing biomolecules until uncaging. The mechanism proceeds via UV light (around 350 nm) absorption by the nitro , leading to intramolecular hydrogen abstraction, formation of an aci-nitro intermediate, cyclization to a benzisoxazol, and subsequent to liberate the free . This can be represented as: \text{Photocaged-R-S-oNB} + h\nu \rightarrow \text{R-SH} + \text{o-nitroso benzaldehyde} Quantum yields for thiol release typically range from 0.085 to 0.37, depending on the derivative and conditions, allowing efficient deprotection with minimal byproducts. In bioconjugation, uncaged thiols can then react with maleimides or other electrophiles for site-specific protein labeling, offering spatiotemporal precision in applications like hydrogel formation or enzyme modulation. Light-induced cycloadditions, particularly those involving tetrazines, provide another versatile platform by photoactivating strained alkene partners like for inverse electron-demand Diels-Alder reactions. or oxidation can generate active tetrazines from photocaged dihydrotetrazines, enabling rapid with second-order rate constants exceeding 1 M⁻¹ s⁻¹ under physiological conditions. A seminal example is the red (625 nm) activation of dihydrogen tetrazines, where oxidizes the precursor to the reactive tetrazine, which then undergoes with , facilitating bioorthogonal crosslinking across 1 cm of tissue with high fidelity and low toxicity to encapsulated cells. Post-2015 advancements include NIR variants using cyanine-based photocages, which cleave upon 690 nm via photooxidative bond scission and cyclization, releasing thiols or for conjugation while penetrating deeper tissues (up to several millimeters) with low (1–10 mW/cm² intensity). These methods collectively advance precise assembly and labeling by harnessing 's orthogonality to biological processes.

Applications in Biotechnology

Therapeutic Conjugates

Bioconjugation plays a pivotal role in the development of therapeutic conjugates, particularly antibody-drug conjugates (ADCs) and PEGylated proteins, by enabling the precise attachment of bioactive payloads to biological molecules for targeted delivery and improved . In ADCs, cytotoxic drugs such as auristatins are covalently linked to monoclonal via bioconjugation chemistries, allowing selective targeting of cancer cells while minimizing off-target toxicity. Common attachment strategies involve maleimide-thiol reactions with residues on the antibody, which are exposed after reduction of interchain bonds, resulting in a drug-antibody ratio (DAR) typically ranging from 2 to 8 to balance and . Controlling the DAR is crucial, as heterogeneous conjugation can lead to variable and increased aggregation, whereas site-specific methods—such as engineered mutants or enzymatic conjugation—enhance uniformity and by achieving defined DAR values, often around 2-4, which correlate with improved and reduced toxicity in preclinical models. A prominent example is , an approved by the FDA in 2011 for relapsed and anaplastic large cell , where the anti-CD30 is conjugated to the auristatin derivative (MMAE) via a protease-cleavable valine-citrulline linker attached to cysteines, yielding an average DAR of 4. This conjugation enables intracellular payload release by lysosomal enzymes, contributing to its clinical efficacy, with response rates exceeding 75% in CD30-positive tumors. A more recent example is datopotamab deruxtecan (Datroway), approved by the FDA in January 2025 for certain non-small cell lung cancers, utilizing a I inhibitor payload linked via a tetrapeptide-based cleavable linker to enhance targeted delivery. Though challenges persist in optimizing linker stability to prevent premature drug release and associated systemic . Metrics from clinical trials highlight that site-specific ADCs like next-generation variants demonstrate up to 10-fold lower off-target compared to conventional cysteine-linked ones, underscoring the impact of precise bioconjugation on therapeutic outcomes. Beyond ADCs, represents a foundational bioconjugation approach for extending the of protein therapeutics by attaching () chains, typically via amine-reactive chemistries to residues or the , which shields the protein from renal clearance and . The pioneering example is Adagen (pegademase bovine), approved in 1990 for (SCID), where multiple 5 kDa chains are conjugated to the residues of using succinimidyl carbonate chemistry, increasing its circulating from minutes to over 30 hours and enabling effective . This modification reduces dosing frequency and , with clinical improvements in immune function, though careful control of PEG attachment sites is essential to preserve enzymatic activity and avoid over-conjugation-induced loss of function. Overall, these bioconjugation strategies in therapeutic conjugates have transformed protein-based drugs, with ongoing refinements focusing on release mechanisms and conjugation specificity to further enhance efficacy-to-toxicity ratios.

Diagnostic and Imaging Tools

Bioconjugation plays a pivotal role in diagnostic and tools by enabling the attachment of probes to biomolecules such as antibodies, peptides, and nanoparticles, facilitating their in cellular and contexts. These modifications allow for targeted detection of disease biomarkers, with common strategies involving covalent linkages that preserve biomolecular function while enhancing signal specificity. Fluorophores and radiometals are among the most widely used probes, conjugated via site-specific chemistries to minimize off-target effects and improve resolution. Fluorophore attachment is a cornerstone of optical imaging techniques, particularly for fluorescence microscopy, where dyes like are linked to biomolecules through thiol-maleimide reactions or -alkyne . Thiol-based conjugation targets residues, offering high efficiency under mild aqueous conditions, while handles enable bioorthogonal labeling for live-cell imaging without disrupting native biology. For instance, dyes, spanning near-UV to near-IR spectra, provide bright, photostable signals with reduced self-quenching when conjugated to proteins, supporting applications in confocal and . These methods ensure selective labeling, with reaction yields often exceeding 90% in optimized buffers. Radiolabeling via chelation is essential for positron emission tomography (PET), where macrocyclic chelators like DOTA are conjugated to lysine residues on antibodies or peptides to stably bind radiometals such as ^{64}Cu or ^{89}Zr. This approach allows non-invasive, whole-body imaging of tumor targets, with DOTA's tetracarboxylate arms forming thermodynamically stable complexes (log K > 20) that prevent metal dissociation in vivo. Seminal work has demonstrated that site-specific DOTA conjugation maintains antibody affinity while enabling high-specific-activity labeling, crucial for detecting low-abundance antigens in clinical settings. Antibody-fluorophore conjugates exemplify practical applications, such as trastuzumab-Alexa Fluor 647 for HER2-positive , where conjugation yields uniform labeling for intraoperative fluorescence-guided surgery. often leverages bioorthogonal handles, like tetrazines on antibodies reacting with trans-cyclooctene-modified probes, to enable tracking of cellular processes with minimal . These conjugates have shown tumor-to-background ratios up to 10:1 in preclinical models, highlighting their diagnostic precision. Advances in the include multimodal probes combining optical and MRI capabilities, such as gadolinium-chelating fluorophores bioconjugated to antibodies for hybrid imaging that merges high-resolution cellular detail with deep-tissue penetration. Nanoparticle integrations, like gold nanorods conjugated via linkers to targeting ligands, enhance signal amplification and , with recent reviews noting improved clearance profiles through zwitterionic coatings that extend circulation half-lives beyond 24 hours. These developments support theranostic platforms, where diagnostic imaging informs subsequent interventions. Key considerations in bioconjugation for include avoiding , often mitigated by asymmetrical distribution on protein surfaces to prevent π-stacking, and optimizing clearance rates to reduce non-specific uptake. choice impacts ; for example, hydrophilic dyes like sulfo-Cyanine minimize liver accumulation, achieving renal clearance rates of 50-70% within hours, thereby improving signal-to-noise ratios in longitudinal studies. These factors ensure probes maintain diagnostic utility without compromising host physiology.

Challenges and Future Directions

Stability and Specificity Issues

One major challenge in bioconjugation is the instability of linkers, particularly the retro-Michael reaction in maleimide-thiol conjugates, which leads to and premature payload release. This instability arises from the reversible addition of thiols to the maleimide double bond, resulting in ring opening under physiological conditions, with half-lives as short as hours in . Post-modification aggregation further compromises , as chemical alterations to proteins can expose hydrophobic regions, promoting oligomerization and loss of bioactivity, especially in antibody-drug conjugates (ADCs). Specificity issues often stem from cross-reactivity, such as unintended thiol-disulfide exchange reactions that disrupt native protein structures or lead to heterogeneous conjugates. For instance, free thiols introduced for conjugation can react with endogenous disulfides, reducing site-selectivity and yielding mixtures with variable drug-to-antibody ratios. To mitigate this, excess quenching agents like are employed to cap unreacted thiols, improving conjugate homogeneity, though this adds complexity to purification. In vivo, bioconjugates face serum instability from enzymatic degradation and immunogenicity risks, where linker hydrolysis or foreign payloads trigger immune responses, shortening circulation half-lives to minutes in some cases. Half-life assays, such as pharmacokinetic studies in animal models, reveal that unstable conjugates exhibit rapid clearance, with effective half-lives often below 24 hours without stabilization strategies. Conjugation efficiency metrics, targeting yields above 90%, are critical for scalability, yet off-target reactions frequently reduce this to 70-80% without optimized conditions. A notable case study is the early ADC gemtuzumab ozogamicin (Mylotarg), where the acid-labile hydrazone linker failed to maintain stability in clinical trials, causing premature calicheamicin release, elevated toxicity, and limited efficacy, leading to its initial market withdrawal in 2010. Similarly, first-generation ADCs with cleavable linkers showed high failure rates in phase II/III trials due to systemic payload exposure, underscoring the need for robust linker design. These examples highlight how instability and poor specificity can derail therapeutic translation, with immunogenicity assays confirming anti-drug antibody formation in up to 20% of patients.

Advances in Multifunctional Conjugates

Recent innovations in bioconjugation have enabled the of multifunctional conjugates that integrate multiple therapeutic or diagnostic payloads into a single construct, enhancing in complex biological environments such as heterogeneous tumors. Post-2020 advancements emphasize dual-drug antibody-drug conjugates (ADCs) and theranostic agents, which combine with for real-time monitoring and personalized dosing. These constructs leverage site-specific conjugation to achieve uniform drug-to-antibody ratios (DARs) and minimize off-target effects, as demonstrated in clinical trials where multifunctional ADCs showed improved overall response rates (ORRs) compared to single-payload predecessors. Dual-drug ADCs represent a key advance, incorporating two distinct cytotoxic payloads to address tumor heterogeneity and . For instance, KH815, a TROP2-targeted with I and inhibitors, entered Phase I trials in 2025, exhibiting synergistic cytotoxicity in preclinical models. Similarly, zanidatamab zovodotin (ZW49), a bispecific HER2-targeted , achieved a 28-31% ORR in Phase I studies for solid tumors, including gastric cancer, by enhancing and immunogenic . BL-B01D1, targeting and HER3, reported a 34% ORR across 174 patients in Phase I trials, highlighting the potential of bispecific designs to overcome resistance mechanisms. These developments rely on heterotrifunctional linkers and site-specific engineering to attach dual payloads without compromising stability. Theranostic bioconjugates integrate therapeutic and diagnostic moieties, such as radionuclides or near-infrared dyes, to enable simultaneous treatment and . sarotalocan, approved in 2020 for head and neck (HNSCC), uses a non-internalizing conjugated to the IR700 via a cleavable linker, activating via near-infrared photoimmunotherapy (NIR-PIT) to induce systemic antitumor immunity with reduced toxicity. ESG401, a TROP2-targeted , demonstrated promising in Phase I trials for solid tumors, allowing dosimetry-guided dosing. As of 2025, over 15 ADCs and a handful of antibody-radionuclide conjugates (ARCs) have gained approval worldwide, with ongoing trials emphasizing bioorthogonal linkers for modular payload exchange. These agents improve in personalized by correlating signals with therapeutic response. Sequential bioorthogonal conjugations facilitate the stepwise attachment of multiple functionalities, enabling precise control over conjugate architecture. The nitrile-aminothiol conjugation (NATC) strategy, advanced in 2024, allows dual and sequential labeling of proteins with high , as shown in the site-specific modification of antibodies for bifunctional ADCs. This method outperforms traditional copper-catalyzed azide-alkyne cycloadditions by avoiding metal catalysts, thus preserving protein integrity. In practice, tetrazine-norbornene cycloadditions have been used to sequentially install imaging and therapeutic groups on nanoparticles, yielding multifunctional probes with DARs up to 4 without . Such techniques support the creation of immunoconjugates that elicit both direct and immune . DNA-templated assembly provides spatial precision for multifunctional bioconjugates, guiding the organization of proteins, nanoparticles, and drugs via hybridization. A approach using templates positioned multiple DNA-functionalized gold nanoparticles for enhanced assembly in biosensing applications. This method has been applied to create programmable nanocomposites that respond to biochemical cues, such as pH changes in tumors, for controlled drug release. By leveraging Watson-Crick base pairing, DNA-templated systems achieve nanoscale assemblies with defined stoichiometries, outperforming random conjugation in yield and functionality. Proteolysis-targeting chimeras (PROTACs) conjugated via bioconjugation emerged in the as multifunctional degraders, linking E3 ligase recruiters to target proteins for ubiquitin-mediated . Antibody-PROTAC conjugates (Ab-PROTACs), developed in 2020, use site-specific bioconjugation to attach PROTAC warheads to antibodies, enabling cell-surface protein in tumors while sparing healthy tissue. For example, HER2-targeted Ab-PROTACs degraded receptor tyrosine kinases with values below 1 nM in preclinical models, offering advantages over small-molecule PROTACs in . Aptamer-PROTAC conjugates further extend this to targets, demonstrating selective in cancer cells. These constructs exemplify bioconjugation's role in expanding PROTAC applications beyond intracellular targets. Nanoscale assemblies via bioconjugation have advanced for multifunctional , such as polymer-coated gold conjugated to antibodies using . In 2025, NIR-luminescent gold were sequentially conjugated to polymers and targeting ligands, forming assemblies that enable deep-tissue and photothermal with enhanced stability. DNA-programmed assemblies, reported in 2025, integrate multiple bioconjugates for stimuli-responsive release, achieving up to 90% in hypoxic tumor models. These structures support theranostic functions by combining , targeting, and in sub-100 nm scales. Sustainability trends in bioconjugate synthesis prioritize eco-friendly methods, such as enzyme-mediated conjugations and metal-free click reactions, reducing use and waste. The ACS GCI Pharmaceutical Roundtable promotes green bioconjugation for ADCs through and synthesis efforts. These approaches align with broader efforts in conjugates, where bioorthogonal strategies minimize hazardous reagents. In , bioconjugates are tailored to patient-specific biomarkers, as seen in custom ADCs designed via genomic profiling for rare mutations. Developments in 2025 include patient-derived antibody conjugates for neoantigen targeting, improving response rates in precision oncology trials by 20-30%. These enable adaptive therapies, such as switchable linkers responsive to individual tumor microenvironments. Future directions include AI-optimized linkers, where models like Linker-GPT generate novel linkers with predicted stability and cleavage rates, accelerating design cycles by 50%. CRISPR-enabled tagging integrates bioorthogonal handles into genomes for conjugation, as in 2023 protocols for endogenous protein labeling, facilitating real-time multifunctional conjugate assembly in . These innovations promise scalable, patient-centric bioconjugates for complex diseases.

References

  1. [1]
    Advances in Bioconjugation - PMC - PubMed Central
    This review focuses on modern methods for bioconjugation, and delineates both imperatives and means for making useful bioconjugates.2. Motivation For... · 3. Bioconjugation Linkages · 3.2. Linkages Containing...Missing: definition | Show results with:definition
  2. [2]
    and site-selectivity aspects in the chemical conjugation of proteins
    Jan 26, 2022 · Bioconjugation can be defined as the covalent coupling between a biomolecule—in this case, a protein—and any other molecule, including another ...
  3. [3]
    Bioconjugation - an overview | ScienceDirect Topics
    Bioconjugation is defined as the process of forming structures composed of metal substances that are adsorbed or chemically adhered to biomacromolecules.
  4. [4]
    [PDF] Bioconjugation and crosslinking technical handbook
    Bioconjugation is the process of chemically joining two or more molecules by a covalent bond where at least one molecule is a biomolecule.
  5. [5]
    Bioconjugation - an overview | ScienceDirect Topics
    Bioconjugation is a chemical process of attaching a biomolecule to another molecule by forming a stable covalent link between them.
  6. [6]
    Nanoscale bioconjugates: A review of the structural attributes ... - NIH
    Nanobioconjugates are nanoscale drug delivery vehicles that have been conjugated to or decorated with biologically active targeting ligands.
  7. [7]
    Bioconjugation in Drug Delivery: Practical Perspectives and Future ...
    Aug 6, 2025 · 1−6 With a wide range of substrates, protein bioconjugation has been used for drug discovery and delivery, 5, 7, 8 protein imaging, 2,7,9 ...
  8. [8]
    A brief review of biomaterials used in drug delivery, vaccine ...
    In this review, the state-of-the-art studies on the applications of biomaterials in biotherapy, including drug delivery, vaccine development, gene therapy, and ...
  9. [9]
    Avidin-Biotin Interaction | Thermo Fisher Scientific - US
    Advantages of using Avidin-biotin systems. The Avidin-biotin complex is the strongest known non-covalent interaction (Kd = 10-15M) between a protein and ligand.Missing: bioconjugation | Show results with:bioconjugation
  10. [10]
    Achieving Controlled Biomolecule–Biomaterial Conjugation
    Jul 24, 2018 · In this review, we focus on the chemistry of biomolecule conjugation and provide a comprehensive overview of the key strategies for achieving controlled ...
  11. [11]
    Basic Bioconjugation Chemistry of Reactive Groups in Biomolecules
    Apr 13, 2022 · In most cases, the selected reaction conditions must preserve the biomolecules' three-dimensional structures to be conjugated or cross-linked.Missing: handles | Show results with:handles
  12. [12]
    A review of conjugation technologies for antibody drug ... - NIH
    Abstract. Antibody–drug conjugates (ADCs) have gained significant attention in biotherapeutics after several years of steady development.
  13. [13]
    The Evolving Landscape of Antibody–Drug Conjugates: In Depth ...
    Oct 11, 2023 · This report provides a detailed overview of the recent advances in antibody–drug conjugate research and considers future directions and ...
  14. [14]
  15. [15]
  16. [16]
    Effect of covalent attachment of polyethylene glycol on ... - PubMed
    1977 Jun 10;252(11):3582-6. Authors. A Abuchowski, J R McCoy, N C Palczuk, T van Es, F F Davis ... PEG-1900-catalase and PEG-5000-catalase exhibited enhanced circulating lives in the ...
  17. [17]
  18. [18]
    A summary of the measured pK values of the ionizable groups ... - NIH
    The average pK values are Asp 3.5 ± 1.2 (139); Glu 4.2 ± 0.9 (153); His 6.6 ± 1.0 (131); Cys 6.8 ± 2.7 (25); Tyr 10.3 ± 1.2 (20); Lys 10.5 ± 1.1 (35); C- ...
  19. [19]
  20. [20]
    Analysis of accessible surface of residues in proteins - Lins - 2003
    Jan 1, 2009 · The different secondary structures correspond to different accessibility of residues. Random coils, turns, and β-structures (outside β-sheets) ...
  21. [21]
    Addition of the keto functional group to the genetic code of Escherichia coli | PNAS
    ### Key Facts on Incorporation of p-Acetylphenylalanine and Its Use for Bioconjugation
  22. [22]
    None
    Nothing is retrieved...<|separator|>
  23. [23]
    Unnatural amino acid incorporation in E. coli: current and future ...
    Apr 1, 2014 · Unnatural amino acid (UAA) incorporation by amber codon suppression offers scientists a powerful tool to modify the properties of proteins at will.
  24. [24]
    Advances in Proximity-Assisted Bioconjugation - ACS Publications
    Sep 25, 2025 · By harnessing the latent reactivity of molecules, proximity techniques can improve upon traditional bioconjugation strategies, simplify ...
  25. [25]
    From molecules to medicine: thiol selective bioconjugation in ...
    Moore and Ward pioneered the initial exploration of the maleimide-thiol-based reaction and elucidated its potential for cross-linking thiol-containing proteins ...
  26. [26]
  27. [27]
    Tunable Degradation of Maleimide–Thiol Adducts in Reducing ...
    Addition chemistries are widely used in preparing biological conjugates, and in particular, maleimide–thiol adducts have been widely employed.
  28. [28]
    Fast Cysteine Bioconjugation Chemistry - PMC - NIH
    Cysteine bioconjugation refers to the use of cysteine as a reactive linker to conjugate proteins with synthetic functional groups under physiological conditions ...
  29. [29]
    Systematic Evaluation of Protein Reduction and Alkylation Reveals ...
    In this study, we compared common reduction reagents (dithiothreitol, tris-(2-carboxyethyl)-phosphine, and β-mercaptoethanol) and alkylation reagents.Missing: seminal | Show results with:seminal
  30. [30]
    Bioconjugation Using Thiols: Old Chemistry Rediscovered to ...
    Dec 14, 2012 · Copolymers were synthesized via thiol-maleimide coupling reaction, by conjugation of semitelechelic poly(HPMA)-SH with maleimide-modified β- ...
  31. [31]
    Cysteine-specific protein multi-functionalization and disulfide ...
    Feb 21, 2020 · The addition of cysteine to 3Br-5MPs generates conjugates that are amenable to secondary addition by another thiol or cysteine, making 3Br-5MPs ...
  32. [32]
    A Platform for the Generation of Site-Specific Antibody–Drug ...
    Jul 22, 2020 · THIOMAB antibody technol. utilizes cysteine residues engineered onto an antibody to allow for site-specific conjugation. The technol. has ...
  33. [33]
    Research progress on chemical modifications of tyrosine residues in ...
    Dec 11, 2023 · This review highlights methods for chemically modifying tyrosine (Tyr) residues, which are suitable for selective modifications, and are used ...
  34. [34]
    [PDF] Utilizing Tyrosine: Advancing Site-Specific Bioconjugation ...
    For example, palladium-catalyzed cross- coupling reactions enable the attachment of a wide range of functional groups to tyrosine residues, expanding the scope ...
  35. [35]
    Tyrosine bioconjugation – an emergent alternative - PMC - NIH
    Another group of compounds used to modify tyrosine residues are diazonium reagents 6, which were used to modify tyrosine residues on the MS2 bacteriophage17 and ...
  36. [36]
    Radiometal-Containing Aryl Diazonium Salts for Chemoselective ...
    Dec 10, 2019 · The described azo coupling chemistry represents an innovative and versatile bioconjugation strategy for selective targeting of tyrosine residues ...
  37. [37]
    [PDF] Tyrosine conjugation methods for protein labelling - HAL-Inserm
    Nov 16, 2020 · Figure12 Mechanism of the diazo-coupling reaction. The potential of Y modification by diazonium coupling was shown by the modification of the ...
  38. [38]
    Site-Specific Bioconjugation through Enzyme-Catalyzed Tyrosine ...
    Aug 21, 2020 · We show that the enzyme tyrosinase is capable of oxidizing exposed tyrosine residues into o-quinones that react rapidly with cysteine residues on target ...
  39. [39]
    Tyrosine bioconjugation – an emergent alternative - RSC Publishing
    Oct 28, 2020 · Tyrosine bioconjugation is a less explored, emergent alternative to traditional protein bioconjugation, becoming a complement or stand-alone ...<|separator|>
  40. [40]
    Arylation Chemistry for Bioconjugation - PMC - PubMed Central
    Here we outline and review sulfur, nitrogen, selenium, oxygen, and carbon arylative bioconjugation strategies and their applications to modify peptides, ...
  41. [41]
    C–H Functionalization in the Synthesis of Amino Acids and Peptides
    Aug 21, 2014 · 2.3.​​ The formation of similar ortho-arylated tyrosines through rhodium-catalyzed C–H activation has been reported by Bedford et al. (42) and is ...
  42. [42]
    Exploiting Protein N-Terminus for Site-Specific Bioconjugation - NIH
    Jun 9, 2021 · Direct approaches provide N-terminal protein modification in one step, taking advantage ... disadvantages, allow for the decoration of protein ...
  43. [43]
    Selectivity and stability of N-terminal targeting protein modification ...
    Nov 17, 2022 · The most classical selective reactions rely on benzaldehyde (BA) handles that can reversibly form imines at protein N-termini, and be trapped ...
  44. [44]
  45. [45]
    Characterization of covalent crosslinking strategies for synthesizing ...
    Jul 10, 2019 · It has been used in a variety of conjugation techniques to couple carboxyl groups to primary amines [1,2,3]. EDC first forms an active ester ...
  46. [46]
    Carbodiimide Crosslinker Chemistry | Thermo Fisher Scientific - US
    EDC couples NHS to carboxyls, forming an NHS ester that is considerably more stable than the O-acylisourea intermediate while allowing for efficient conjugation ...
  47. [47]
  48. [48]
    [PDF] Click Chemistry and Bioorthogonal Chemistry - Nobel Prize
    Oct 5, 2022 · The Royal Swedish Academy of Sciences has decided to award Carolyn R. Bertozzi,. Morten Meldal, and K. Barry Sharpless the Nobel Prize in ...
  49. [49]
    Cu-Catalyzed Azide−Alkyne Cycloaddition | Chemical Reviews
    This review will focus mainly on the Cu(1) catalysis in the Huisgen reaction, broadly known as the azide/alkyne-“click”-reaction or CuAAC-reaction.
  50. [50]
    From Mechanism to Mouse: A Tale of Two Bioorthogonal Reactions
    Aug 15, 2011 · Bioorthogonal reactions are chemical reactions that neither interact with nor interfere with a biological system.
  51. [51]
    Bioorthogonal chemistry: strategies and recent development - PMC
    ... (SPAAC), also known as Cu-free click chemistry. The first example of SPAAC was reported by Bertozzi and co-workers in 2004 when they used cyclooctyne (OCT) ...Missing: seminal paper
  52. [52]
    Oximes and Hydrazones in Bioconjugation: Mechanism and Catalysis
    Jun 22, 2017 · This review will highlight the most recent strategies to improve oxime/hydrazone bioconjugation reactions with a particular emphasis on ...
  53. [53]
  54. [54]
  55. [55]
  56. [56]
    Cell Surface Engineering by a Modified Staudinger Reaction
    ### Summary of Key Content from https://www.science.org/doi/10.1126/science.287.5460.2007
  57. [57]
    Staudinger Ligation and Reactions – From Bioorthogonal Labeling ...
    Nov 27, 2022 · In this review, we highlight groundbreaking discoveries and applications of Staudinger reactions in the molecular life sciences.Abstract · Introduction · Summary & Future Objective · References
  58. [58]
    Staudinger Reaction - Organic Chemistry Portal
    Mechanism of the Staudinger Reaction. Triphenylphosphine reacts with the azide to generate a phosphazide, which loses N2 to form an iminophosphorane. Aqueous ...
  59. [59]
    A “Traceless” Staudinger Ligation for the Chemoselective Synthesis ...
    Here we report a novel modification of our previously reported “Staudinger ligation” that generates an amide bond from an azide and a specifically ...Author Information · References
  60. [60]
    [PDF] Reaction Mechanism and Kinetics of the Traceless Staudinger ...
    Mechanism of the Staudinger Ligation. Our putative mechanism for the Staudinger ligation is shown in Scheme 1.10. The nitrogen atom of an iminophosphorane ...
  61. [61]
    Protein Engineering with the Traceless Staudinger Ligation - NIH
    Here, we present the traceless Staudinger ligation as a strategy for the convergent chemical synthesis of proteins. This reaction unites a phosphinothioester ...<|control11|><|separator|>
  62. [62]
  63. [63]
    THETA as an Efficient Cu-Binding Ligand for Manual and Automated ...
    Jul 15, 2024 · We present efficient CuAAC protocols for the synthesis of rufinamide and its precursor, methyl 1-[(2,6-difluorophenyl)methyl]-1H-1,2,3-triazole-4-carboxylate.
  64. [64]
  65. [65]
    Structure–Reactivity Studies of 2-Sulfonylpyrimidines Allow ...
    Sep 1, 2023 · Arylation by 2-SPs is metal-free, operates under benign aqueous conditions, at neutral pH, and forms highly stable conjugates. In full protein ...
  66. [66]
    Designing photolabile ruthenium polypyridyl crosslinkers for ...
    Feb 8, 2019 · Here we describe spectrally tunable (λmax = 420–530 nm) ruthenium polypyridyl complexes functionalized with two photolabile nitrile ligands that ...Missing: bioconjugation | Show results with:bioconjugation
  67. [67]
    Enzymatic Bioconjugation: A Perspective from the Pharmaceutical ...
    (7) This chemistry is generally robust and leads to high conversion during the process, but it comes with drawbacks. First, proteins of interest need to contain ...
  68. [68]
    Enzymatic Bioconjugation: A Perspective from the Pharmaceutical ...
    In this Perspective, we seek to illuminate applications of enzymatic bioconjugation in the face of an expanding palette of new drug modalities.2. Antibody Drug Conjugates · 3. Radioligand Therapies · 6. Oligonucleotide Based...
  69. [69]
    Site-Specific Conjugation of Native Antibody: Transglutaminase ...
    This review focuses on the versatility of transglutaminases in the field of bioconjugation and covers recent developments in utilizing mTG for generating ...Abstract · Scheme 2 · Scheme 3
  70. [70]
    Sortase A as a Novel Molecular “Stapler” for Sequence-Specific ...
    The Sortase family of transpeptidase enzymes catalyzes sequence-specific ligation of proteins to the cell wall of Gram-positive bacteria.Missing: seminal | Show results with:seminal
  71. [71]
    Reprogramming the specificity of sortase enzymes - PNAS
    Sep 3, 2014 · Here we present a system for the directed evolution of reprogrammed SrtA variants that accept proteins with altered sorting motifs.
  72. [72]
    Improved variants of SrtA for site-specific conjugation on antibodies ...
    Aug 18, 2016 · Sortase mediated ligation is a highly specific platform for conjugation that relies on the specificity of the transpeptidase Sortase A ...Missing: seminal | Show results with:seminal
  73. [73]
    Recent advances in sortase-catalyzed ligation methodology
    Sortase-catalyzed ligation is a powerful strategy for protein modification. Engineered sortases improve reaction rates and eliminate Ca 2+ dependency.
  74. [74]
    Light-Triggered Click Chemistry
    ### Summary of Light-Triggered Click Chemistry for Bioconjugation
  75. [75]
    Photoclick Chemistry: A Bright Idea
    ### Summary of Photoclick Reactions in Bioconjugation (Post-2015)
  76. [76]
    Photoremovable Protecting Groups in Chemistry and Biology
    Dec 21, 2012 · Nitrobenzyl, nitrophenethyl compounds, and their dimethoxy derivatives (nitroveratryl) (section 3) are by far the most commonly used PPGs. The ...
  77. [77]
    Page not found | Nature
    Insufficient relevant content.
  78. [78]
    Red Light Activation of Tetrazine–Norbornene Conjugation for ...
    Apr 5, 2017 · Red Light Activation of Tetrazine–Norbornene Conjugation for Bioorthogonal Polymer Cross-Linking across Tissue. Click to copy article link ...
  79. [79]
    A Near-IR Uncaging Strategy Based on Cyanine Photochemistry
    Here we report a near-IR light-initiated uncaging reaction sequence based on readily synthesized C4′-dialkylamine-substituted heptamethine cyanines.
  80. [80]
    Antibody drug conjugate: the “biological missile” for targeted cancer ...
    Mar 22, 2022 · Antibody–drug conjugate (ADC) is typically composed of a monoclonal antibody (mAbs) covalently attached to a cytotoxic drug via a chemical linker.
  81. [81]
    Site-Specific Antibody–Drug Conjugates: The Nexus of ... - NIH
    Antibody–drug conjugates (ADCs) combine the specificity of antibodies with the potency of small molecules to create targeted drugs.I. Introduction · Figure 2 · Figure 3
  82. [82]
    Strategic and Chemical Advances in Antibody–Drug Conjugates
    Sep 5, 2025 · This review critically examines established and emerging conjugation strategies, including lysine- and cysteine-based chemistries, enzymatic ...
  83. [83]
    Definition of brentuximab vedotin - NCI Drug Dictionary
    An antibody-drug conjugate (ADC) directed against the tumor necrosis factor (TNF) receptor CD30 with potential antineoplastic activity.
  84. [84]
    Brentuximab vedotin, an antibody–drug conjugate, in patients ... - NIH
    Brentuximab vedotin (ADCETRIS®) is an antibody–drug conjugate (ADC), consisting of a CD30‐directed antibody, cAC10, conjugated by a protease‐cleavable ...
  85. [85]
    PEGylated therapeutics in the clinic - PMC - PubMed Central
    Sep 22, 2023 · The first two PEGylated products to receive FDA approval were Adagen™ (1990) ... approved products use the amine aldehyde conjugation to ...
  86. [86]
    Pegademase - an overview | ScienceDirect Topics
    ADAGEN is the first PEGylated drug approved by the FDA (March 1990) for clinical use in cancer treatment [128]. There are about 20 PEGylated drugs under ...
  87. [87]
    Bioorthogonal Chemistry and Its Applications - ACS Publications
    Nov 30, 2021 · Bioorthogonal chemistry is a set of methods using the chemistry of non-native functional groups to explore and understand biology in living organisms.
  88. [88]
    Biological Probes Built from Fluoresceins and Rhodamines
    Apr 7, 2017 · Fluorescent bioconjugates are used in a variety of applications: Fluorophore- labeled antibodies are crucial elements of immunofluorescence ...
  89. [89]
    The Effect of Fluorophore Conjugation on Antibody Affinity and ... - NIH
    In general, conjugation of fluorophores to antibodies can be carried out via amino, thiol, and carbohydrate groups (3, 4). Coupling of dyes to ε-amino groups ...Labeling Of Cells With... · Flow Cytometry · Results
  90. [90]
    An Overview of PET Radiochemistry, Part 2: Radiometals
    Oct 1, 2018 · This review gives a short overview of the most frequently used radiometals for PET, their physical properties, suitable chelators, and examples of applications.
  91. [91]
    State of the Art in Radiolabeling of Antibodies with Common and ...
    This review presents a state of the art overview of the practical aspects of radiolabeling of antibodies, ranging from fast kinetic affibodies and nanobodies ...Missing: seminal | Show results with:seminal
  92. [92]
    Zirconium 89 and Copper 64 for ImmunoPET: From Antibody ... - MDPI
    In PET imaging, the chelating agent DOTA is considered the gold standard for 64Cu and 68Ga. It is characterized by 4 oxygens and acetate groups that act as ...Missing: seminal | Show results with:seminal
  93. [93]
    Monoclonal antibody–fluorescent probe conjugates for in vivo target ...
    Examples include ICG-labeled daclizumab, trastuzumab or panitumumab. These conjugates demonstrated minimal fluorescence in solution, but rapidly became ...
  94. [94]
    Tetrazine bioorthogonal chemistry derived in vivo imaging - PMC - NIH
    Bioorthogonal chemistry represents plenty of highly efficient and biocompatible reactions that proceed selectively and rapidly in biological situations ...
  95. [95]
    Bioorthogonal Chemistry and Bioconjugation: Synergistic Tools for ...
    Jul 29, 2021 · Bioorthogonal chem. can be used for the selective modification of biomols. without interfering with any other functionality present in the cell.
  96. [96]
    Implication of Bimodal Magnetic Resonance and Fluorescence ...
    Jul 29, 2025 · The probes combine fluorescence for precise cellular imaging with MRI for in-depth tissue visualization, providing synergistic benefits that ...<|separator|>
  97. [97]
    Advances in multimodal imaging techniques in nanomedicine
    Jul 30, 2025 · This review introduces current imaging techniques such as optical imaging, computed tomography (CT), magnetic resonance imaging (MRI), nuclear ...Missing: 2020s | Show results with:2020s
  98. [98]
    The Current Status and Future Directions on Nanoparticles for ... - NIH
    Sep 14, 2024 · Probes based on nanomaterials have demonstrated promising imaging potential and have garnered significant attention. Within the realm of ...
  99. [99]
    Minimization of self-quenching fluorescence on dyes conjugated to ...
    Asymmetrical distribution prevents the chromophores from π-stacking thus minimizing the energy transfer and fluorescence quenching. The conjugation of the dye ...Missing: avoidance clearance
  100. [100]
    Not so innocent: Impact of fluorophore chemistry on the in vivo ...
    A number of studies have found that fluorophores can dramatically impact the pharmacokinetic and tumor-targeting properties of the bioconjugates they are meant ...<|separator|>
  101. [101]
    assessing the impact of chemical modifications on antibody ... - Nature
    Oct 28, 2025 · This review examines in vivo imaging studies that compare “parental” and “modified” mAbs imaged under identical conditions to assess the effects ...
  102. [102]
    Addressing the retro-Michael instability of maleimide bioconjugates
    We provide an overview on methods that attempt to increase the safety and efficacy of ADCs via “self-hydrolysing maleimides” or by improving the stability of ...
  103. [103]
    [PDF] Long-Term Stabilization of Maleimide−Thiol Conjugates - ProLynx Inc
    Dec 12, 2014 · ABSTRACT: Michael-addition of a thiol to a maleimide is commonly used for bioconjugation of drugs to macromolecules.
  104. [104]
    Stabilization challenges and aggregation in protein-based ... - NIH
    Dec 11, 2023 · In this review, we have discussed some features of protein aggregation during production, formulation and storage as well as stabilization strategies.
  105. [105]
    The role of thiols and disulfides in protein chemical and physical ...
    Thiol groups and disulfides have important roles in the stability and solubility of proteins. Thiols and disulfides are inherently very reactive.2. Disulfide Bond Formation · Figure 3 · 6. Thiol Reaction And...<|control11|><|separator|>
  106. [106]
    Quantification of Thiols and Disulfides - PMC - PubMed Central - NIH
    Here, free thiols can be quenched with a small reagent, such as NEM, and, after removal of excess maleimide, the disulfide bonds are reduced so that the ...
  107. [107]
    Predictable and tunable half-life extension of therapeutic agents by ...
    Stable conjugates suffer several limitations: loss of drug potency due to conjugation, confining the drug to the extracellular space, and the requirement for a ...
  108. [108]
    Conjugation Efficiency of Laser-Based Bioconjugation of Gold ...
    Oct 22, 2009 · Low HS-ssO:NP ratios of up to 40 seem to enable conjugation efficiencies of more than 90% (Figure 3). Here, the binding seems to be limited by ...
  109. [109]
    Effects of antibody, drug and linker on the preclinical and clinical ...
    There are many examples of ADCs that have shown early preclinical promise and that entered clinical trials briefly, but have not progressed or have been ...
  110. [110]
    Immunogenicity of Antibody Drug Conjugates: Bioanalytical ... - NIH
    Immunogenicity risk assessment, testing strategies, and bioanalytical methods can be adapted to reflect the complexity of ADC therapeutics.
  111. [111]
    The next frontier in antibody-drug conjugates - PubMed Central - NIH
    Jul 3, 2025 · Antibody-Drug Conjugates (ADCs) have achieved significant success in cancer therapy by combining the targeting specificity of monoclonal ...
  112. [112]
    Antibody–Drug Conjugates (ADCs): current and future ...
    Apr 30, 2025 · Notable examples include trastuzumab deruxtecan and sacituzumab govitecan, which achieve DAR values of 7.8 and 7.6, respectively. This ...
  113. [113]
    Theranostic antibody conjugates: A review of recent trends and ...
    Currently, more than 15 ADCs and ARCs have been approved for theranostic purposes, with numerous candidates in various stages of preliminary and clinical ...Missing: bioconjugates | Show results with:bioconjugates
  114. [114]
    Theranostic antibody conjugates: A review of recent trends and ...
    Aug 25, 2025 · We discuss key strategies for targeting cancer cells as well as recent advances in conjugation techniques, payload selection, and targeting ...Missing: bioconjugates | Show results with:bioconjugates
  115. [115]
    Advancing Nitrile-Aminothiol Strategy for Dual and Sequential ...
    Aug 19, 2024 · Nitrile-aminothiol conjugation (NATC) stands out as a promising biocompatible ligation technique due to its high chemo-selectivity.Missing: multifunctional | Show results with:multifunctional
  116. [116]
    Orthogonal bioconjugation targeting cysteine-containing peptides ...
    Aug 12, 2024 · Crucially, multifaceted bioconjugation is achieved through clickable handles to incorporate structurally diverse functional molecules. This “two ...Missing: multifunctional post-<|separator|>
  117. [117]
    Exploring the Addressability of DNA Decorated Multifunctional Gold ...
    Jul 16, 2018 · To assess the effectiveness of multiple conjugated sequences of DNA strands, a DNA origami template was used to study the hybridization ...
  118. [118]
    Advancing Biosensing and Imaging with DNA-Templated Metal ...
    This review highlights the remarkable versatility and latest advancements of DNA-templated MNCs, particularly in biosensing and imaging.
  119. [119]
    Novel PROTAC Technologies: Advancing Targeted Protein ...
    Sep 21, 2025 · This blog examines three innovative PROTAC modalities: aptamer-PROTAC conjugates (APCs), antibody-PROTAC conjugates (Ab-PROTACs), and ...
  120. [120]
    Unconventional PROTACs for Targeted Protein Degradation in ...
    Jun 10, 2025 · These unconventional PROTACs show great promise for targeted protein degradation in cancer therapy. This review summarizes recent advances in ...2.2 Peptide-Based Ligands · 4 Pro-Protacs · 4.1 Caged E3 Ligase Ligand
  121. [121]
    Bioorthogonal conjugation of NIR luminescent gold nanoclusters ...
    Aug 27, 2025 · The insights gained from this work provide valuable guidance for an efficient bioorthogonal coupling between multifunctional AuNCs and polymers ...Missing: post- | Show results with:post-
  122. [122]
    DNA-programmed nanomaterials: advancing biosensing ...
    May 13, 2025 · DNA-programmed nanomaterials hold immense potential in biosensing, drug delivery, and disease treatment, offering new possibilities for next-generation ...
  123. [123]
  124. [124]
    Recent developments in bioconjugation: From strategies to design ...
    One effective strategy in bioconjugation involves modifying existing functional groups in biomolecules to increase their reactivity or utility. This compensates ...
  125. [125]
    How bioconjugation is unlocking new drug development
    Mar 27, 2025 · Bioconjugation presents an new frontier for pharmaceutical development, providing an option for improved drug efficacy across modalities.
  126. [126]
    Linker-GPT: design of Antibody-drug conjugates linkers ... - Nature
    Jul 1, 2025 · In this study, we introduce Linker-GPT, a Transformer-based deep learning framework leveraging self-attention mechanisms to generate novel ADC linkers.
  127. [127]
    AI-driven innovation in antibody-drug conjugate design - Frontiers
    AI/ML plays a significant role in designing linker architectures for ADCs, moving beyond static structure prediction to proactive optimization of linker ...
  128. [128]
    Efficient tagging of endogenous proteins in human cell lines ... - PNAS
    Jul 24, 2023 · We present a versatile CRISPR/Cas strategy to insert affinity tags to endogenous proteins in human cell lines and demonstrated its efficiency.Results · Materials And Methods · Plasmid Yifancheng (pyc)...<|separator|>