Abstract
Building on recent advances in peptide science, medicinal chemists have developed a hybrid class of bioconjugates, called peptide-drug conjugates, that demonstrate improved efficacy compared to peptides and small molecules independently. In this perspective, we discuss how the conjugation of synergistic peptides and small molecules can be used to overcome complex disease states and resistance mechanisms that have eluded contemporary therapies because of their multi-component activity. We highlight how peptide-drug conjugates display a similar multi-factor therapeutic mechanism to antibody-drug conjugates but also demonstrate improved therapeutic properties such as less-severe off-target effects and conjugation strategies with greater site-specificity. The many considerations that go into peptide-drug conjugate design and optimization, such as peptide/small-molecule pairing and chemo-selective chemistries, are discussed. We also examine several peptide-drug conjugate series that demonstrate notable activity towards complex disease states such as neurodegenerative disorders, and inflammation, as well as viral and bacterial targets with established resistance mechanisms.
Graphical Abstract

1. INTRODUCTION
The evolution of peptide therapeutics and their clinical applications.
Developments in protein chemistry have been instrumental in the identification of several protein- and peptide-based therapeutics that have entered the small molecule-dominated drug market. Protein-based therapeutics represent attractive medicinal chemistry leads, as these macromolecules have been evolutionarily optimized for activity under physiological conditions and possess inherently favorable immunogenic and metabolic properties. These attractive aspects are demonstrated by the sizable increase in approved biologics that target a broad range of disease states, including cancer and metabolic disorders.1 Despite their successes, there are still some limitations of therapeutic proteins, including the need for intravenous, intramuscular, or subcutaneous injections. Because of the recent advances made to develop orally bioavailable peptide therapeutics, there is renewed interest in peptides as a therapeutic modality.2,3 The aim of these peptide-based strategies is to develop a therapeutic with improved drug-like properties that also retains the desired biological activity of the parent protein.
First-generation peptide drugs such as insulin and adrenocorticotropic hormone are examples of endogenous hormones that found success as drugs when applied exogenously.4 In this era of drug discovery, peptides with established physiological roles were isolated from natural sources, purified, and given to patients. Hormone-based peptides were clinically successful in the mid- to late 1900s; however, few examples existed outside this class. During this time, it became apparent that poor peptide plasma half-lives and bioavailability were major hurdles to their clinical application.5 Natural peptides lack the intramolecular interactions necessary to replicate the higher-order structure of their parent proteins; thus, they circulate in the bloodstream as linear poly-peptide chains. These linearized peptides are efficiently digested by peptidases because they are freely recognized by enzymatic active sites. Additionally, peptides are also efficiently excreted by the same homeostatic mechanisms that regulate plasma-hormone concentrations.
The development of peptide synthesis strategies and technologies contributed greatly to the next generation of peptide therapeutics that address the major shortcomings. The introduction of automated peptide synthesizers has enabled a higher-throughput strategy for the development of highly effective and selective peptide analogs. Using this technology, peptide chemists have found that the incorporation of unnatural amino acids has had a significant effect on increasing the plasma half-life of peptides.5 Modifications of the peptide backbone via incorporation of amide bioisosteres yielded similar results.6 These two strategies have been leveraged by peptide chemists to confer secondary structure to peptides through additional non-native covalent bonds, effectively locking them into the desired conformation.7,8 These chemically modified peptides display improved half-lives because they are less susceptible to recognition and degradation by proteolytic enzymes.
Another improvement to peptide properties is through the development of chemo-selective chemistries that permit labeling of specific amino acids. By leveraging the inherent nucleophilicity of some amino acids, particularly cysteine,9 chemists have designed peptide conjugates with improved solubilities and selectivities. The incorporation of unnatural amino acids that are amenable to click chemistry is also exploited for the generation of peptide conjugates.10,11 These strategies are even more powerful when coupled with peptide synthesis as they enable site-specific labeling of peptides, which is critical for the retention of biological activity. The potential for peptide bioconjugates is exponential and includes the incorporation of biomolecules, such as fatty acids and sugars,12,13 as well as compounds such as polyethylene glycols (PEGs).14 Peptide conjugates with increased molecular weight have also been shown to be resistant to clearance, thus staying in circulation longer.15 These chemically modified peptides have found great clinical success in the past decade and are considered the second-generation of peptide therapeutics.
The improved stability and solubility of chemically modified peptides have paved the way for novel therapeutic strategies. One therapeutic avenue that has demonstrated considerable success is the parallel application of synergistic small molecule drugs and peptides to combat complex disease states.16,17 This strategy has led to the development of conjugates where small molecule drugs are linked to peptides, overcoming the challenge of co-administration of different compounds. Peptide-drug conjugates (PDCs) are an emergent class of therapeutics that have already found success in clinical trials (e.g. NCT03613181, NCT03486730, and NCT01767155), indicating that they will be a part of the next-generation of peptide therapeutics. In this Perspective, we will compare PDCs to other well documented bioconjugate therapies, discuss their design and optimization, as well as highlight some exciting examples in the pipeline. It is our hope that this review will give the reader insight into the current state of the field and provide inspiration for the design of their own peptide-drug conjugates.
Peptide-drug conjugates as an alternative to antibody-drug conjugates.
Before embarking on a discussion of PDCs, it is important to consider another well-studied example of bioconjugates: antibody-drug conjugates (ADCs). This class of compounds combines the highly specific target recognition activity of antibodies with the desired therapeutic effect of a small-molecule partner. These conjugates have received significant attention in the realm of cancer therapeutics where antibodies that recognize cancer-specific antigens are coupled to a cytotoxic payload.18,19 In the case of anti-cancer ADCs, the antibody confers target-specificity to the cytotoxic small molecule with the goal of limiting off-target effects and improving the overall efficacy of non-specific cancer therapies. To date there are more than 10 ADCs approved by the U.S Food and Drug Administration (FDA), with dozens of pharmaceutical companies having disclosed the development of additional ADC therapies.19 The success of ADCs might suggest to the reader that the development of alternative bioconjugate therapeutics is redundant; however, ADC-based therapies come with a range of limitations. In the following section we will discuss some pitfalls of traditional ADC therapeutics and highlight ways that PDCs can overcome these challenges.
One of the major advantages of ADCs over traditional small-molecule therapeutics is their capacity for targeted delivery owing to antibodies that bind antigens with high affinity and specificity. This targeted delivery of ADCs represents a significant therapeutic advancement; however, the large size and molecular weight of antibodies limits the application of ADCs. As a result of these physical properties, antibodies are unable to freely diffuse through the plasma membrane with few (if any) membrane proteins capable of transporting these macromolecules into the intracellular space; therefore, ADCs are restricted to antibodies that recognize proteins that present an antigen to the extracellular space.20 This therapeutic strategy requires ADCs to be endocytosed for effective delivery of the small-molecule payload to elicit the desired therapeutic effect.21,22 In contrast, peptides are much smaller than antibodies and several membrane transport proteins exist that traffic peptides into the intracellular space;23 thus, peptides that recognize intracellular proteins can be used in PDC design, which greatly increases the number of target proteins accessible to PDCs. PDCs can be applied to a broader range of protein targets, although peptides generally recognize binding partners with a much weaker affinity than antibodies.
The highly specific antigen recognition of ADCs has encouraged the development of bioconjugates that possess cytotoxic payloads. This therapeutic strategy is centered on the idea that ADCs elicit target-specific cell death with greatly reduced off-target cytotoxicity. These bioconjugates are particularly appealing in the area of cancer therapeutics because an antibody that recognizes a cancer-specific antigen can be leveraged for targeted killing of a cancerous mass, but the antigen-specific binding of ADCs also presents challenges as it requires that a disease-related antigen is almost exclusively present on diseased cells;22 thus, the identification of a disease-related antigen and the subsequent generation of an antibody capable of recognizing the antigen are significant components of the ADC-development process24. If these conditions are not met–that is to say, if an antibody displays even slight promiscuity or the selected antigen is present on healthy cells–off-target cytotoxicity can have serious consequences for a patient. Notably, these challenges are less significant in the PDC space because peptides can be conjugated to a variety of non-cytotoxic small molecules to elicit a desired therapeutic outcome. A famous example of anti-cancer PDCs are the proteolysis targeting chimeras (PROTACs) that combine a peptide that recognizes a target protein and a small molecule that sequesters an E3 ubiquitin ligase.25 These hybrid PROTACs hijack the ubiquitin-proteasome system, which results in the targeted degradation of a protein.25 Compared to the off-target effects of ADCs, the directed degradation of a protein target in a healthy cell is much less severe. Unlike cytotoxic ADCs that rely upon cell death as their therapeutic mechanism, PDCs can be tailored for a wider range of therapies with less severe consequences.
Another limitation of ADC development is the narrow range of conjugation chemistries that can be leveraged to join an antibody to a small-molecule payload. As previously mentioned, peptide chemists have developed a toolbox of chemo-selective chemistries that permit labeling of specific amino acids.26 These powerful conjugation reactions allow for the labeling of complex biological material beyond peptides; however, site-specific conjugation of full-length proteins is particularly challenging, as the incidence of a reactive amino acid in a large protein sequence is usually greater than one. For this reason ADCs are commonly decorated with several small molecules, and ADC samples are heterogeneous as it is impossible to control the antibody-to-payload ratio.27,28 The variability in antibody to payload ratio, as well as the distribution of the small molecule payload(s), can have significant consequences for ADC therapeutics because they impact factors such as activity and pharmacokinetic properties.27,29 These obstacles are less of a concern with peptide-based conjugates because of their smaller size, as well as the speed and ease of peptide synthesis using contemporary equipment. Manual peptide synthesis has enabled peptide chemists to incorporate unnatural amino acids into peptides which can be used for site-specific labeling.10,11 Additionally, chemo-selective labeling of peptides is more easily tolerated as unwanted reactive amino acids can be modified during peptide synthesis to avoid unwanted labeling while also retaining the physicochemical properties of the native peptide.
Despite the limitations outlined in this section, ADCs are powerful therapeutic tools that mark a significant improvement over traditional chemotherapies. In addition, the consortium of researchers involved in ADC development have invested considerable time and resources into overcoming these challenges with substantial success;30–32 however, these solutions are outside the scope of this review. This section highlighted ways that PDC development differs from ADCs and how PDCs can overcome some of the challenges associated with working with large biologics. In the section that follows, we will outline some of the considerations that go into PDC design and optimization.
2. PEPTIDE-DRUG CONJUGATE DESIGN AND OPTIMIZATION
A PDC is a class of targeted therapeutics composed of a peptide and a small molecule drug joined through a covalent linkage. Although the name is unambiguous, it provides little information about the complex considerations that go into PDC development. Designing PDCs is rarely straightforward as researchers must consider the effects and activities of several small molecules and peptides, as well as their limitations. One must also reflect upon the architecture of both the peptide and drug to determine where to join the independent molecules, in addition to the chemistry necessary to form the covalent linkage.
Designing a PDC development campaign begins with the selection of a peptide and small molecule with complementary activities. The identification of small molecules and/or peptides with demonstrated activity towards a particular target or disease state is a logical starting point. The merger of a small molecule and peptide with parallel targets and activities may lead to an ultra-potent PDC; however, conjugation may also be used to redeem once-promising molecules that were ultimately considered undrug-like. For example, a small molecule that exhibits potent anti-cancer activity but demonstrates non-specific cytotoxicity would be a strong candidate for conjugation to peptides that specifically interact with cancer markers to generate a PDC for site-specific delivery. There are countless cases of small molecules identified in high-throughput screens or through iterative rounds of structure-activity relationship (SAR) studies that display a desired activity but fall short of entering clinical trials due to unrelated imperfections. These molecules are strong candidates for adoption into PDC campaigns because their shortcomings may be addressed by a peptide partner. Peptides can be used to combat a range of undesirable effects, from inefficient cell penetration33 to high promiscuity and off-target effects.34 Consequently, a thorough characterization of a small molecule candidate is necessary for selecting the most appropriate peptide for PDC development. This strategy may be applied in reverse, where a peptide is chosen as a starting point for PDC design, and small molecules are selected to enhance activity or overcome limitations of the selected peptide.
Rudimentary PDCs consists of a small molecule covalently bound to a peptide without the introduction of a linker. These PDCs are most effective when the small molecule and peptide act on the same target, and the separate binding sites are near one another; however, linker-less PDC strategies come with limitations as they are structurally rigid, a property that can significantly restrict the peptide or drug from accessing its target in a preferred orientation. The short distance between the peptide and small molecule is another shortcoming of bipartite PDCs as the proximity of these two components may result in poor target binding due to steric hindrance. For these reasons, tripartite PDCs composed of a peptide, drug and chemical linker are preferred as they allow for a greater degree of flexibility and decrease the likelihood of steric hindrance. Chemical linkers range widely in length, chemical composition, and rotational freedom based on their identity and level of unsaturation. Linker selection is another important consideration in bioconjugate design, and we encourage the reader to consult the excellent reviews that exist on this topic.35,36
Potentially the most important factor in PDC design is determining the optimal sites and chemistries for conjugation. Ideally, structural information about small molecule and/or peptide binding would be available to determine the preferred molecular orientation; however, this is not always the case. Unfortunately, it is impossible to predict optimal conditions a priori; thus, trial and error is often necessary. Given the inherent stability of the peptide backbone and the relatively limited reactive functionality of amino acid side chains, identifying the peptide’s reactive centers is a good starting point for designing conjugation strategies. Bioconjugation reactions developed for cysteine thiols, as well as lysine and N-terminal free amines, are highly robust and have been optimized over the course of decades.26 Newer bioconjugation chemistries have also been developed for amino acids with less nucleophilic side-chains, such as tyrosine.26,37 The restricted reactivity of amino acids was once a major limitation for site-specific labeling; however, the incorporation of unnatural amino acids amenable to bioorthogonal chemistry has allowed researchers to precisely modify peptides while retaining many of their native properties.10 Once a conjugation site has been identified, the chemistry required to modify a particular amino acid can guide small molecule or linker derivatization. SAR data establishing the small molecule pharmacophore should also be used to determine the optimal sites of conjugation.
3. APPLICATIONS OF PEPTIDE-DRUG CONJUGATES
In the sections that follow, we will outline examples of PDCs designed to treat viral infections, bacterial infections, and inflammation, as well as compounds developed for CNS targets. Of note, cancer therapeutics are another rich area of PDC development; however, we will not touch on these compounds as several excellent reviews already exist on this topic.38,39 These sections are not meant to be a comprehensive list of PDCs; rather, our aim is to showcase these projects as case studies.
4. ANTI-VIRAL PEPTIDE-DRUG CONJUGATES
The devastating outcomes of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV2) pandemic has highlighted the importance of antiviral therapeutics. These outcomes are compounded by the spread of vaccine-resistant SARS-CoV2 variants, which demonstrated the need for mutant-agnostic anti-viral drugs to complement traditional vaccine campaigns; however, the significant mutation rate of viruses is also a concern for antiviral drug development as drug-resistant viruses emerge at a rapid rate.40 Anti-viral PDCs are an attractive strategy for overcoming this challenge as peptides with a high affinity for viral proteins can be used to deliver drugs to their target. In the case of drug-resistant viruses, high-affinity peptides bring the anti-viral payload near the target protein, increasing the local concentration of the antiviral compound. This apparent increase in concentration may be sufficient to overcome the decreased binding-affinity brought on by mutation.
Notably, analyses of protein structures from genetically distinct viruses revealed significant structural conservation despite a high degree of sequence deviation.41,42 These results suggest that these proteins share a common mechanism of action; thus, antiviral strategies may be repurposed to inhibit homologous proteins across different viral families. This idea is particularly interesting when considering PDCs as highly specific peptides can be designed to tailor therapies for particular viruses. Here, we will discuss PDC strategies that have been developed to treat infections from human immunodeficiency virus (HIV) and Dengue virus (DENV), two viruses that lack effective, mutation-resistant therapies.
Human Immunodeficiency Virus entry-inhibiting PDCs.
HIV infections reached epidemic proportions in 1981 and have claimed millions of lives to date. Several antiretroviral therapies with unique HIV targets have been developed to treat infections43; however, resistance to these therapies develops at a significant rate. An important target for HIV therapies is the envelope protein, gp160, which facilitates virus-cell fusion. Briefly, gp160 on the viral membrane recognizes a receptor protein, CD4, and a coreceptor (CXCR4 or CCR5) on the surface of T-cells, which prompts the envelope protein to undergo a large-scale conformational change that ultimately fuses the two membranes.44,45 To combat this process, therapeutics that inhibit receptor recognition or prevent formation of the post-fusion conformation have been developed to prevent HIV-entry.
The most widely prescribed HIV-entry inhibitor, Enfuvirtide (T-20), is a biomimetic peptide that is used to prevent formation of the post-fusion conformation of the envelope protein.46 Unfortunately, poor plasma half-life and the incidence of resistant mutants47 are significant limitations of the current treatment. The success of T-20 prompted researchers to identify additional peptides with similar binding modes.48,49 Many of these peptides displayed comparable, low nanomolar, half-maximal inhibitory concentration (IC50) values compared to T-20, while also demonstrating efficacy against resistant strains and improved pharmacokinetic properties;48,49 however, these peptides are large (>30 amino acids), which severely limits their clinical application due to poor bioavailability. Notably, shortened peptides with similar binding-modes have significantly reduced potencies,50 and the best small molecule candidates inhibit HIV-entry at micromolar concentrations.51 These findings further demonstrated the need for next-generation cell entry inhibitors with novel modes of action.
Early HIV-entry PDCs were designed to overcome the loss of potency that occurs when working with shorter peptides. In one example, this effect was achieved by substituting a critical gp160 motif, termed WWI, with N-(carboxyphenyl)pyrroles, which are known to bind to the same hydrophobic cavity.52 Previous research into N-(carboxyphenyl)pyrrole derivatives revealed weak HIV-entry inhibition, and SAR analysis highlighted the necessary structural features for inhibition.53 N-(carboxyphenyl)pyrrole derivatives with key inhibitory features were conjugated to the N-terminus of a shortened HIV entry-peptide inhibitor, termed P26, through a β-alanine linker. Notably, this series of PDCs demonstrated a greater than 8-fold increase in potency compared to P26 alone (2540 nM), with the most potent PDC, Aoc-βAla-P26, (1, Figure 1A) reaching an IC50 of 14.9 nM. Compound 1 rivaled T-20, which displayed an IC50 of 10.1 nM. Remarkably, the two most potent PDCs tested in this series also demonstrated inhibition of T-20-resistant HIV strains with IC50 values around 100-200 nM. Pharmacokinetic studies of 1 displayed decreased proteinase-K digestion and an increased in vitro half-life compared to T-20, although the PDC had a 4-fold shorter half-life in vivo.
Figure 1: Chemical Structures of Human Immunodeficiency Virus Cell Entry Inhibitors.

Peptide-drug conjugate structures are displayed such that peptides are in blue, linkers in black and small molecules in red. Cartoon representations of protein targets are displayed on the right with target names below. (A) HIV entry inhibitor peptide P26 is N-terminally conjugated to the N-(carboxyphenyl)pyrrole, Aoc, through a β-alanine linker.52 (B) P26 is conjugated to the antiviral triterpene sapogenin through a chemical linker that contains a triazole moiety covalently attached to the side-chain of a C-terminal lysine and an additional β-alanine residue.54 (C) HIV entry inhibitor peptide SP22 is conjugated to the CCR5 inhibitor, TAK, via a PEG-12 linker covalently attached to the C-terminal lysine side-chain.55
A similar strategy was employed using sapogenin, an antiviral triterpene, conjugated to P26 via a triazole linker generated with click-chemistry.54 In the sapogenin series of inhibitors, the small molecule was connected to P26 at the N-terminus through a modified β-alanine, or the C-terminus via reaction with a modified lysine side-chain. Surprisingly, the C-terminally modified P26 conjugates exhibited the greatest potency, with one of the most potent PDCs, P26-BApc, (2, Figure 1B) inhibiting cell entry at 3.94 nM, which was 3-fold more potent than T-20. Compound 2 also demonstrated inhibition of T-20-resistant HIV strains and displayed improved pharmacokinetic properties compared to T-20, including a longer in vivo half-life.
Recently, alternative HIV cell entry PDC inhibitors targeting the receptor/co-receptor recognition event have shown considerable success.55,56 For example, the gp160 peptide-mimetic, P52, was C-terminally conjugated to CCR5 antagonists that block HIV receptor recognition57–59 to generate a dual-target inhibitor.55 PEG linkers of varying length were selected for this conjugation strategy, and inhibition studies revealed that PEG-4- and PEG-8-conjugated PDCs exhibited inhibition at 1.11 nM and 1.52 nM, respectively, while PEG-12 and PEG-24 linkers resulted in sub-nanomolar inhibition at 0.35 nM and 0.12 nM. Although all of the PDCs mentioned were more potent than T-20 (4.02 nM), these results demonstrate the importance of linker optimization in PDC design. After determining the optimal PEG linker length, P-52 was exchanged for a modified stapled peptide, SP22, with demonstrated anti-HIV entry activity,60 and a C-terminal lysine was introduced for site specific labeling at the side-chain amine. A potent PDC from this series, SP12T (3), is displayed in Figure 1C. Remarkably, this series of PEG-12-linked PDCs was found to inhibit HIV strains that recognize the alternative co-receptor, CXCR4, with greater potency than T-20 against all strains tested. This approach demonstrates the many ways that PDCs can be designed to address therapeutic challenges from different angles. There are even examples of HIV entry PDC inhibitors that use peptide mimetics of the HIV receptor CD4 with reported success.61
Dengue Virus protease-inhibiting PDCs.
The World Health Organization reports that Dengue virus (DENV) infections have grown rapidly over the past decades from 500,000 infections in 2000 to a staggering 5.2 million infections in 2019. Additionally, these infections were once localized to tropical and subtropical regions but have recently been reported in non-tropical climates.62 The rapid spread of DENV has prompted some to consider it a global health concern, as severe infections may be fatal. Worryingly, no therapies are available to treat DENV infections, although drug targets for DENV therapies have been characterized. An appealing target for anti-DENV therapies is inhibition of the viral protease activity required to digest the large DENV polyprotein into its active components.63 This proteolytic activity of DENV is performed by a catalytic complex composed of the multi-domain protein NS3, and the cofactor protein NS2B.64
Although the NS3-NS2B complex demonstrates serine protease activity, known serine protease inhibitors are largely ineffective at inhibiting DENV protease activity.65 This gap prompted researchers to test tetrapeptide libraries for inhibitors, which revealed a preference for peptides complementary to the di-basic recognition motif of NS3-NS2B.66 Following identification of effective tetrapeptide inhibitors, electrophilic warheads were introduced at the C-terminus to improve potency, as demonstrated for other serine protease inhibitors.67 This tetrapeptide-warhead strategy proved effective, with the most potent inhibitor, Bz-Nle-Lys-Arg-Arg-B(OH)2 (4, Figure 2A), achieving an inhibition constant of 43 nM; however, the intrinsic reactivity of these warhead-peptides and the poor pharmacokinetic properties associated with the dibasic motif limit the clinical application of these inhibitors.
Figure 2: Chemical Structures of Dengue Virus Protease Inhibitors.

Peptide-drug conjugate structures are displayed such that peptides are in blue and small molecules in red. Cartoon representations of protein targets are displayed on the right with target names below. (A) A tribasic tetrapeptide complementary to the DENV protease recognition motif is covalently linked to a C-terminal boronic acid moiety.67 (B) A dibasic retro-tripeptide complementary to the DENV protease recognition motif was covalently attached to an arylcyanoacrylamide at the N-terminus.70 (C) The N-benzoyl capped dibasic tripeptide contains an unnatural 4-benzyloxy-D-phenylglycine residue at the C-terminus with a β-lactam ring covalently attached to the terminal nitrogen in the S configuration.74
Building on the success of the warhead-based inhibitors, researchers sought to identify small-molecules with affinity for the DENV NS3-NS2B complex. These efforts resulted in the identification of arylcyanoacrylamide,68 rhodanine and thiazolidinedione69 scaffolds with modest micromolar affinity for the DENV protease. Having identified inhibitory peptides and small molecules, a series of DENV PDCs were developed which situated arylcyanoacrylamide derivatives as an N-terminal cap of various peptide sequences.70 From this series of PDCs, the N-capped retro-tripeptide Arg-Lys-Nle-NH2 (5, Figure 2B) exhibited the best potency, with an inhibition constant of 4.9 µM, as well as high specificity relative to other serine proteases, including the closely related West-Nile virus protease. Despite the in vitro success of the arylcyanoacrylamide PDCs, these compounds demonstrated weak anti-viral activity in cell culture, prompting researchers to explore rhodanine and thiazolidinedione as peptide capping moieties.70 Cyanoacrylamide, rhodanine and thiazolidinedione scaffolds are challenging medicinal chemistry leads due to their high promiscuity;69–72 nonetheless, these anti-DENV PDCs demonstrated low micromolar inhibitory concentrations with a site-specific binding mode as demonstrated by competition assays.71 Interestingly, rhodanine-capped peptides with hydrophobic substituents demonstrated considerable membrane permeability. This trend was also noted in cell-based assays where peptides capped with hydrophobic rhodanines demonstrated greater antiviral activity. Recently, N-sulfonyl capped peptides have emerged as novel anti-DENV PDCs, with the most potent derivatives achieving double-digit nanomolar inhibition constants.73 Remarkably, these compounds have demonstrated high DENV protease selectivity, limited cytotoxicity and significant metabolic stability, indicating that they may represent strong candidates for clinical applications.
A final DENV PDC series must also be mentioned that revisits the covalent inhibition strategy. This PDC series (6, Figure 2C) incorporated β-lactam warheads at the C-terminus of anti-DENV peptides with the aim of developing PDCs with ‘soft’ reactivity74 (i.e., reactivity with “soft” nucleophiles like the thiol group of cysteine). These PDCs demonstrated considerable success with many derivatives achieving single digit micromolar IC50 values. Many of these PDCs also exhibited specificity for the NS3-NS2B complex compared to endogenous human serine proteases. Interestingly, these compounds displayed a complex inhibition mechanism where binding could result in the desired covalent inhibition of the NS3-NS2B serine, or cleavage of an internal peptide bond within the PDC. This report provides an interesting starting point for the development of covalent DENV protease inhibitors with greater specificity and lower reactivity; however, further optimization is required to mitigate inhibitor degradation.
5. ANTIBACTERIAL PEPTIDE-DRUG CONJUGATES
Since their discovery, antibiotics have been the workhorse of medical practitioners with billions of prescriptions being filled on an annual basis worldwide. In 2021, the CDC reported that 211.1 million antibiotic prescriptions were filled in the United States alone. This rise in antibiotic usage is directly proportional to the increase in antibiotic-resistant bacteria, as resistance mechanisms develop at an alarming rate due to prolonged and repeated exposure. Disturbingly, the increase in antibiotic drug resistance is accompanied by an increase in multi-drug-resistant bacteria, further emphasizing the need for novel antibiotic therapeutics. To this end, antibiotic PDC strategies have been developed that either combine synergistic antibiotic activities75,76 or modulate existing antibiotics to escape resistance mechanisms.77,78 Antibiotic PDCs have also been designed with significant efficacy toward gram-negative bacteria,79 which has been a challenge for traditional small molecule antibiotics. In the following section, we will explore the development of vancomycin-based PDCs and the strategies employed to improve the vancomycin pharmacokinetic profile and overcome resistance. We also encourage the reader to consult the many reviews that discuss antibacterial peptides80,81 and the development of antibiotic PDCs82,83 for further reading.
Vancomycin-based peptide-drug conjugates.
Vancomycin is an antibacterial glycopeptide that was first isolated in the 1950s from the bacterial species Amycola-toposisorientalis.84 This antibacterial compound was found to have activity towards gram-positive bacteria, including species with resistance to β-lactam antibiotics such as penicillin. The novel antibiotic exhibited a unique mechanism of action where bacterial cell wall peptidoglycan synthesis was inhibited by targeting a terminal d-Ala-d-Ala motif of the growing peptidoglycan chain.85 Vancomycin interacts with this motif on peptidoglycan intermediates, preventing the formation of the glycan backbone by peptidoglycan polymerase. Unfortunately, vancomycin-resistant organisms were identified less than 30 years after FDA approval, with resistance brought on by single amino acid alterations to the terminal peptidoglycan intermediate motif.86 Because of this developing resistance, vancomycin is administered only in serious and life-threatening cases of bacterial infections, particularly those with established antibiotic resistance. The rise of resistance to this last-resort treatment has motivated the development of vancomycin-based therapies that overcome these resistance mechanisms. Importantly, several vancomycin-based PDCs have been developed that demonstrate antibiotic activity across a wide range of gram-positive bacteria, including vancomycin-resistant isolates.87,88
A convincing demonstration of vancomycin-based PDCs overcoming vancomycin resistance was reported where vancomycin was C-terminally conjugated to highly basic, lipidated membrane-targeting peptides, generating a series of PDCs called vancapticins.89 This strategy was predicated on the idea that membrane-targeting peptides could be used to increase the concentration of vancomycin at the site of action. The first series of vancapticins contained lipidated peptides that were connected to vancomycin through a disulfide bond. Remarkably, this series of conjugates demonstrated 4- to 10-fold improvement over vancomycin towards methicillin-resistant S. aureus (MRSA), vancomycin-intermediate S. aureus (VISA) and multi-drug resistant (MDR) S. pneumoniae, as well as an impressive 100-fold improvement against vancomycin-resistant enterococci (VRE). Despite the success of the disulfide-linked vancapticin series, the compounds were found to have short half-lives and poor in vitro plasma stability, which prompted researchers to exchange the disulfide linker for covalent attachment through a lysine side-chain89. The most potent PDCs from this series, including 7 (Figure 3A), were significantly more potent than vancomycin towards MRSA, VISA, vancomycin-resistant S. aureus (VRSA) and VRE, and rivaled the potency of contemporary glycopeptides such as oritavancin. The lysine-linked PDCs were found to be stable upon exposure to plasma and glutathione, and metabolically inert when exposed to liver microsomes. These PDCs also exhibited bactericidal activity in mouse models with an improved pharmacokinetic profile compared to vancomycin.
Figure 3: Chemical Structures of Vancomycin Peptide-Drug Conjugates.

Peptide-drug conjugate structures are displayed such that non-vancomycin peptides are in blue, linkers in black, small molecules in red, and additional chemical matter in green. Cartoon representations of protein and membrane targets are displayed on the right with target names below. (A) A compound from the vancapticin PDC series. A four-lysine peptide was covalently linked to vancomycin through the C-terminal lysine side chain and vancomycin carboxylic acid. These PDCspossess hydrophobic moieties at the peptide N-terminus.89 (B) A hexa-arginine peptide was covalently attached to vancomycin through a C-terminal cysteine sidechain linked to the vancomycin secondary amine through a cyclohexane and pyrrolidine-2,5-dione linker.90 (C) A lipopolysaccharide-binding peptide was conjugated to the free carboxylic acid of vancomycin via a linker that contains a pyrrolidine-2,5-dione, 2 PEG units and 2 amide moieties.92
Similar studies of vancomycin-based PDCs tested the effect of polycationic hexa-arginine peptide conjugation at four unique sites on vancomycin.90 All of the PDCs reported in this study demonstrated more potent antibacterial activity compared to vancomycin. Interestingly, the most potent PDC reported, FU002 (8, Figure 3B), had a different conjugation site than the vancapticins. This compound was generated via conjugation at the lone secondary amine in vancomycin and demonstrated a 1000-fold greater antibacterial activity than vancomycin against VRE, as well as significant activity against other vancomycin-resistant strains. The PDC also contained a unique linker that included cyclohexane and pyrrolidine-2,5-dione moieties owing to the bifunctional cross-linker used to facilitate the conjugation chemistries. In vivo studies revealed that 8 was partially excreted by the hepatobiliary route, suggesting this PDC has improved biodistribution over vancomycin, which is primarily excreted by the kidneys. Adding to the success of these PDCs, a separate study reported that poly-arginine vancomycin-based PDCs connected by an aminohexanoic acid linker eradicated MRSA biofilms and persister cells, both in vitro and in vivo.91 Taken together, these independent vancomycin PDC series demonstrate that vancomycin resistance and poor pharmacokinetic profiles can be overcome using PDC strategies.
Vancomycin-based PDCs have also been developed as gram-negative antibacterial agents through careful selection of a peptide partner. In one series of vancomycin PDCs, lipopolysaccharide (LPS)-binding proteins were conjugated to vancomycin at the four sites mentioned previously via CuAAC mediated click-chemistry or maleimide-sulfhydryl addition.92 LPS-binding proteins were selected because LPS constitutes a major portion of the gram-negative bacterial outer membrane (OM); thus, these vancomycin PDCs are expected to target gram-negative bacteria. Notably, the most effective PDCs from this series of antibiotics (9, Figure 3C) displayed single digit micromolar minimum inhibitory concentrations (MICs) for gram-positive bacteria and double digit micromolar MIC values for several gram-negative bacterial species. In a separate study, vancomycin was conjugated to an OM-disrupting peptide, the polymyxin E nonapeptide (PMEN), to generate a PDC series called vancomyxins.93 The vancomyxin series contained PDCs with varying linker lengths and compositions; however, in all cases PMEN was conjugated to vancomycin at the C-terminus or disaccharide moiety via CuAAC click-chemistry. The vancomyxins that contained alkyl chain linkers demonstrated significantly improved antibacterial activity towards gram-negative bacteria compared to vancomycin, whereas this effect was less pronounced with PEG-3 linkers. Unfortunately, the vancomyxins did not display significant antibacterial activity towards vancomycin-resistant gram-positive bacteria. The results from these studies highlight the ways that antibacterial PDCs can be designed to repurpose antibiotics to target gram-negative bacteria with limited effective treatments.
6. PEPTIDE-DRUG CONJUGATES FOR CNS TARGETS
Peptide-Drug Conjugates for Neurodegenerative Diseases.
Neurodegenerative diseases (NDs) are associated with deterioration of the synapse and neural network, as well as the deposition and accumulation of altered protein variants in the brain. These diseases have a broad impact on basic human functioning, impairing basic abilities like speech, movement, stability, and balance, as well as more complex tasks like cognitive abilities. As the global population ages, the prevalence of memory-, cognition-, and movement-impairing diseases will continue to increase.94,95 According to the World Health Organization, NDs require urgent treatment as they are projected to surpass cancer as the second-leading cause of death by 2040.96,97 Alzheimer’s disease (AD) is a leading neurodegenerative disease responsible for most cases of elderly dementia. It causes progressive memory loss, impaired learning, and behavioral decline. The deposition of amyloid plaques in the brain is believed to contribute to the disease, leading to neuronal and synaptic loss.98–100 At present, there is no established cure for AD, and the medications employed in its management primarily focus on addressing cognitive symptoms or other manifestations.98 The blood-brain barrier (BBB) poses a problem for the development of neurodegeneration therapeutics, where most treatments fail due to limited drug permeability. Such poor BBB-permeability hinders the availability of effective options for treating NDs. Additionally, NDs are reported to have many different pathophysiologies,101–104 so it is crucial to prioritize the development of new therapeutic agents that directly target multiple key processes involved in the progression of AD. Several PDC classes have been studied to address these challenges.
Galantamine-peptide derivatives.
Galantamine is an alkaloid natural product that acts as an inhibitor of acetylcholinesterase (AChE),105 which has been studied as a potential target for AD therapeutics. Inhibition of this enzyme causes levels of acetylcholine in the brain to increase to normal levels, which may explain why galantamine treatment is associated with reduced mortality in AD patients and prolonged decline in cognition.106,107 AD has also been linked to the formation and accumulation of amyloid beta (Aβ) deposits.108 Inhibiting Aβ release by γ-secretase is also a widely sought approach against AD. Peptides such as N-(3,4-dichlorophenyl)-d,l-Ala-OH have been shown to inhibit γ-secretase,109 so researchers hypothesized that by conjugating peptides containing this moiety with galantamine, compounds would exhibit joint AChE and γ-secretase inhibition.110 Conjugates were designed to link several di- or tripeptides with the N-(3,4-dichlorophenyl)-D,L-Ala-OH sequence linked to positions 6 or 11 of galantamine through ester or amide bonds (10, Figure 4A).
Figure 4: Chemical Structures of Galantamine-Peptide Conjugates.

Peptide-drug conjugate structures are displayed such that peptides are in blue, linkers in black and small molecules in red. Cartoon representations of protein targets are displayed on the right with target names below. (A) Example of a galantamine-peptide drug conjugate where galantamine (shown in red) is covalently linked through an ester bond to a tripeptide comprising N-(3,4-dichlorophenyl)-d,l-Ala-OH (shown in blue) at position 6.110 (B) Example of a galantamine-peptide drug conjugate with a shortened sequence of OM 99–2 covalently linked at position 11 through an amide bond to an Asp residue, which is also linked to a nicotinic residue.115 (C) Example of a galantamine-peptide drug conjugate with an analog of Leu-Val-Phe-Phe (Aβ17-Aβ20) covalently linked through an ester bond to position 6 of galantamine.118
In vitro testing of the conjugates towards AChE and butyrylcholinesterase (BuCHE) showed that three of the six synthesized conjugates had lower AchE IC50 values than galantamine, and all of them had significantly increased activity as BuChE inhibitors.111 This enzyme is also of interest because BuChE also hydrolyzes acetylcholine,112 meaning that both AchE and BuChE participate in the degradation of this neurotransmitter. All six conjugates were determined to be potent non-competitive inhibitors of BuChE. It is also interesting to note that the less-potent inhibitors against AchE exhibited the highest potency against BuChE, demonstrating that small structural differences in these peptides play a role in selectivity towards the enzymes. γ -Secretase activity was evaluated through cellular and in vitro assays, where one of the conjugates was active in vitro but not in vivo. The results suggest that the lack of activity in vivo might be due to permeability problems and points to a factor to consider when developing these peptide conjugates. All the other conjugates were able to reduce cellular Aβ production in vitro in a dose-dependent manner with similar IC50 values.
Based on the success of these conjugates, studies were continued with an additional interest in inhibition against β-secretase activity. This enzyme is also implicated in the release of the β-amyloid precursor protein.113 Peptides with shortened sequences of Boc-Val-Asn-Leu-Ala-ОН (OM 99-2), which are known β-secretase inhibitors,114 conjugated to galantamine were synthesized towards this goal.115 To additionally optimize the permeability of the compound, which is an essential aspect for drugs that act on the central nervous system (CNS), conjugates were also linked to a nicotinic or an isonicotinic acid moiety (11, Figure 4B). Nicotinic acid has been found to improve permeability of compounds and has increased their passage through the BBB.116,117 These conjugates were expected to exhibit AChE, BuChE, and β-secretase inhibition while ensuring crossing of the BBB. It was determined that there was no sign of toxicity or behavioral changes in male albino mice after a 7-day window following intraperitoneal administration of conjugates. Cellular cytotoxicity assays determined that conjugates had no effect on cellular viability. The conjugates effectively reversed scopolamine-induced memory-impairment in mice and exhibited improved cholinesterase inhibition and antioxidant activity compared to galantamine in ex vivo experiments.
Following these studies, another set of galantamine conjugates were studied.118 This set had galantamine conjugated to residues 17-20 of Aβ (Leu-Val-Phe-Phe; 12, Figure 4C) and analogs of the peptide where Val is replaced by tert-Leu (Tle) or nor-Val (Nva), and Leu is replaced by nor-Leu (Nle). Results showed that all compounds exhibited inhibitory activity against β-secretase, and four of the tested compounds were able to inhibit platelet aggregation. Most notably, this study demonstrated that the alterations that include Nva or Tle in place of Val in the original peptide, along with a free N-terminal carboxyl group, resulted in the most favorable outcomes.
Trolox-conjugated β-amyloid C-terminal peptides.
The most-studied variants of the Aβ peptides are Aβ1-40 and Aβ1-42, although the latter exhibits the higher aggregation propensity because of additional hydrophobic residues at the C-terminus that cause aggregation.119–122 Peptide Aβ1–42 is thus considered to be the most relevant peptide in AD, and inhibition of its self-aggregation has been targeted for the development of novel AD therapeutics. The Aβ peptide has also been linked to the production of reactive oxygen species (ROS) by interacting with molecular oxygen and bioactive metal ions,123–124 promoting cell-membrane perturbation and ultimately resulting in oxidative stress and cell death. Antioxidants like Vitamin E or polyphenols have been studied as a way to counteract the cellular damage caused by oxidative stress in AD.125 A series of peptide conjugates were designed to aid in oxidative injuries and inhibit self-aggregation of Aβ peptides in AD patients’ brains. To this end, the radical-scavenging portion of Vitamin E (trolox) was conjugated to the C-terminus of several Aβ peptide variants.126
Evaluation of the inhibitory activity of the trolox-peptide conjugates (TxAβx–n, where x is the starting residue and n is the final residue of Aβ peptide) on Aβ1–42 aggregation showed that they prevent aggregation of the peptide, while incubation with trolox alone showed no activity. The inhibitory activity of the conjugates increased with the length of the Aβ1–42 C-terminal motif in the TxAβx–42 peptides. Similar inhibitory activities were also observed for TxAβx–43 conjugates. On the other hand, incubation with TxAβx–40 did not show any significant inhibitory activity. This result suggests that the additional residues in the C-termini of TxAβx–42 and TxAβx–43 may play a role in the interruption of Aβ1–42 affinity. No inhibitory activity was reported for Aβ peptides that were not conjugated to trolox, indicating that the length of the peptide, the identity of C-terminal residues, and their conjugation to trolox all play an important role in inhibiting Aβ-peptide aggregation.
Cytotoxicity of the conjugates was evaluated through cell viability assays in human neuroblastoma SH-SY5Y cells.126 Results showed that there was no effect in cell viability after incubating the cells with TxAβx–42 and TxAβx–43, which shows lack of cytotoxicity. To study whether the conjugates could counteract the cytotoxic effects of Aβ1–42 peptide alone, TxAβx–42 and TxAβx–43 were incubated along with Aβ1–42. Results show that cytotoxicity towards the cells is significantly decreased, with cell viability increasing in a dose-dependent manner, and even completely inhibited by one of the conjugates.126 Finally, radical activity of the conjugates was assessed by comparing with controls, and results showed that TxAβ36–42 (13, Figure 5A) was able to completely scavenge ROS induced by H2O2. Additionally, it was able to significantly decrease ROS generation by Aβ1–42.
Figure 5: Chemical Structure of Anti-Neurodegenerative Peptide-Drug Conjugates.

Peptide-drug conjugate structures are displayed such that peptides are in blue, linkers in black, small molecules in red, and additional chemical matter in green. Cartoon representations of protein targets are displayed on the right with target names below. (A) A Trolox-peptide conjugate where trolox is linked through an amide bond to Aβ peptide variant Aβ36–42.126 (B) General structure and an example of a tripartite structure consisting of membrane anchor (raftophile), spacer and pharmacophore (inhibitor). The tripartite structure shown features GL 189 as the BACE1 inhibitor and dihydrocholesterol as the membrane anchor linked through a H-3Gl-OH spacer.127,128 (C) Example of a shuttle-cargo fusion molecule from Hybrid Molecule Subset I. Peptidic carrier TfR-P is conjugated to fallypride directly through an amide bond.131 (D) Example of a shuttle-cargo fusion molecule from Hybrid Molecule Subset II. TfR-P is conjugated to fallypride through a PEG-2 linker.131 (E) Example of a shuttle-cargo fusion molecule from Hybrid Molecule Subset III. Angiopep-2 was conjugated to fallypride through a PEG4 linker.131 (F) Example of of a shuttle-cargo fusion molecule from Hybrid Molecule Subset IV. Carrier peptide TfR-P is conjugated to fallypride through a PEG-3 linker and derivatized with a radiolabeling moiety (SiFA, shown in green), which is linked to the carrier peptide of the compound.131
Tripartite structures of β-secretase inhibitors to raftophilic lipid anchors.
In another approach to target β-secretase activity—specifically, the β-site APP-cleaving enzyme 1 (BACE1)—the concept of “tripartite structures” was proposed.127 These structures consist of a pharmacophore (i.e., a BACE1 inhibitor), a lipid, and a linker to serve as an appropriate connection between them (general structure shown in Figure 5B). Lipids such as cholesterol or dihydrocholesterol partition into lipid rafts on the outer leaflet of the cell membrane. Since BACE1 inhibitors should be anchored to the cell membrane before the compound is endocytosed, coupling inhibitors to these lipids was expected to result in a decrease in Aβ production by tethering the compound to the membrane and facilitating endocytosis.128 A sterol moiety was therefore coupled to the C-terminus of the inhibitory peptide by a polyglycol linker (14, Figure 5B). Commercially available BACE1 inhibitor III (H-Glu-Val-Asn-Sta-Val-Ala-Glu-Phe-NH2) was used as the inhibitor. The conjugates were more potent than the free inhibitor both in vitro and in vivo. As controls, the inhibitor with the polyglycol linker was tested, along with the inhibitor directly linked to the sterol moiety. Both were inactive, which indicated the need for the tripartite structure.
As a continuation to this study, inhibitors GL 189 and a derivative of lysine were coupled to dihydrocholesterol through linkage with a series of spacers.127 The spacers were chosen based on their low toxicity, physiological stability, solubility, and variability in lengths. These new conjugates were tested in cell-based assays, and results showed that at a concentration of 100 nM, tripartite structures reduced the secretion of Aβ40 in all cell lines, while free inhibitors had no effect even at higher concentrations. Because one of the active conjugates had the lysine pharmacophore, results suggested that the tripartite concept also showed efficacy when applied to non-peptidic inhibitors. Conjugates with spacers ranging from 18 to 123 Å were synthesized and tested for their ability to reduce Aβ-peptide secretion to identify optimal length of the spacers. Best results were obtained with spacer lengths ranging from 35 – 89 Å, with a 75–90% reduction in Aβ-peptide secretion. Some flexibility may therefore be allowed in terms of the distance to the membrane. Several other analogs with tripartite structures have also been synthesized to further evaluate the structure-activity relationship.129
Shuttle−cargo fusion molecules of transport peptides and the hD2/3 receptor antagonist fallypride.
Many efforts have been employed to improve blood-brain-barrier permeability of CNS drugs. The most widely studied approach to overcome this limitation is commonly called the molecular Trojan horse approach,130 where a “shuttle” is used to facilitate the crossing of the BBB of a “cargo,” which is the poorly permeable therapeutic; however, the use of large bioactive molecules as shuttles, which, although highly efficient and specific, results in poor pharmacokinetic properties along with other undesirable side effects. A proposed solution is to use small peptides as shuttles that are transported across the BBB by endogenous transporters.131 The use of these small peptides is favorable because they exhibit fewer undesired side effects and better pharmacokinetic properties. They are also easier to synthesize and derivatize with the cargo drug. Several conjugates were synthesized consisting of the hD2/3 receptor-binding-drug fallypride and a peptide carrier. Compound fallypride can be used in vivo with positron emission tomography (PET) imaging and in vitro with hD2/3 receptor binding assays. For the shuttle moiety, peptides with different lengths, structure, polarity, and geometry that enter the brain parenchyma by receptor-mediated transcytosis were chosen. The structural variations of the peptides would give insight into the tolerance of peptide identity while retaining biological activity and receptor selectivity and affinity.
Four different sets of conjugates were designed and synthesized for this first study. The first subset tested the influence of the complexity of the carrier peptide (15, Figure 5C). The second subset was conjugated to a transferrin receptor binding peptide HAIYPRH (TfR-P) by linkers of different lengths to explore the extent to which steric hindrance exerted by the peptide on the drug affects binding affinity of fallypride (16, Figure 5D). The third subset consisted of the conjugation of AngioPep to fallypride through a PEG linker to test the BBB transport capacity of AngioPep in the conjugates (17, Figure 5E). Finally, the fourth subset contained a moiety for radiolabeling with 18F or 68Ga to be applicable in in vivo PET imaging or combined PET/optical imaging of hD2 receptors (18, Figure 5F). The binding affinity for all compounds was in the nanomolar range, which was comparable to that of controls raclopride and chlorpromazine. Results showed that complexity, length, and individual structure of the peptides had no correlation to affinity. Best results were obtained with “medium-sized” linkers, while shorter or longer linkers resulted in lower affinities; however, with the third-subset conjugates, different results were observed: the conjugate with highest binding affinity had no linker at all.
Docking studies based on homology modeling of the receptor more closely relate the influence of the different structural elements of the fusion molecules on hD2 receptor binding. Results showed that the linker, but not the peptide carrier most exposed to solvent, played a role in stabilizing conjugate binding to the receptor. It also confirmed that fallypride can still bind to the receptor without being blocked by the other constituents.
Peptide-Drug Conjugates for Pain Management.
Pain represents a significant burden to healthcare systems as individual treatment plans are often required to manage the frequency and severity of pain in individual cases. Medicinal chemists have addressed this issue with the development of highly potent, small molecule analgesics that typically interact with G protein-coupled receptors (GPCRs) to relieve pain. Despite the drug-likeness of analgesic compounds, these molecules often display target promiscuity and produce harmful off-target effects132. Opioids, such as morphine or fentanyl, are well-known examples of highly potent analgesic compounds with a high potential for abuse and instances of fatal overdose133. According to the CDC, opioid overdose was responsible for over 645,000 deaths between 1999 to 2021. The most potent opioids developed target the GPCR called the μ-opioid receptor (MOR); however, molecules that target this receptor often result in severe and undesirable side effects, including addiction134. To overcome the unfavorable and severe side-effect profiles of MOR targeted ligands, medicinal chemists have designed small molecules that target the κ-opioid receptor (KOR), which represents an alternate target with less significant side-effects. Despite the identification of this parallel target, ligands developed for KOR activation continued to produce unfavorable side effect profiles, albeit with significantly less severe side-effects135. The gradual improvement towards KOR-related non-addictive analgesics has prompted researchers to develop novel KOR ligands with improved side-effect profiles.
Recently, PDCs have been designed to target the KOR to generate compounds with high receptor specificity, while also reducing analgesic side effects136. In this study, the crystal structure of KOR complexed with a ligand, MP1104137, was used to computationally design a series of PDCs that link KOR ligands based on MP1104 to thioether cyclized peptides designed to interact with the extracellular loops of KOR. Before small molecule conjugation, the computationally designed peptides were assessed in cell-based assays which demonstrated that the compounds were unable to displace an orthosteric ligand at concentrations as high as 10 μM. These results suggested that the peptides did not bind the common small molecule KOR binding-pocket, The C-terminal carboxylic acid of the cyclic peptides was then conjugated to the free amine of an MP1104 derivative, β-NalA, to generate this series of PDCs. Remarkably, all four of the PDCs demonstrated low nanomolar affinities, with the best binding PDC, DNCP-β-NalA (19, Figure 6A), producing a Ki of 3.9 nM. This PDC marked a substantial improvement over β-NalA which gave a Ki of 72 nM.
Figure 6: Chemical Structure of Analgesic and Anti-inflammatory Peptide-drug conjugates.

Peptide-drug conjugate structures are displayed such that peptides are in blue, linkers in black, small molecules in red, and additional chemical matter in green. Cartoon representations of protein targets are displayed on the right with target names below. (A) The cyclic peptide DNCP was linked to β-NalA through an l-Thr-d-Phe linker.136 (B) Example of a betulinic acid peptide conjugate where betulinic acid is linked to an azido peptide directly through a C-C bond.142 (C) Example of a Methotrexate-Hyaluronic acid conjugate where methotrexate is linked to hyaluronic acid through a cleavage susceptible peptide and a PEG-3 linker.148 (D) Methotrexate-cIBR conjugate where methotrexate is linked by an amide bond to the N-terminus of cIBR peptide.151 (E) β-carboline peptide conjugate where a Β-carboline alkaloid is linked to ARPAK pentapeptide through an amide bond.162
Pharmacokinetic profiling of 19 revealed full agonism and demonstrated a preference for GPCR binding compared to β-arrestin. This finding is notable as compounds that interact with β-arrestin are associated with a greater incidence of side effects. Mice models also demonstrated the antinocecptive efficacy of 19 while also highlighting a lack of side-effects associated with KOR-specific ligands. Notably, opioid receptor selectivity studies demonstrated that 19 binds KOR and MOR in an equipotent manner; thus, the results of the mouse model studies could be the result of dual KOR/MOR agonism. Another important component of this study was the validation of the binding site of 19 site via cryo-electron microscopy which revealed that β-NalA bound to the common small-molecule binding site of KOR, while the cyclic peptide portion associated with extracellular loops and transmembrane domains of KOR. In summation, this PDC series demonstrates how small molecules with significant side-effects can be leveraged in PDC design to generate potent compounds with reduced off-target effects.
7. ANTI-INFLAMMATORY PEPTIDE-DRUG CONJUGATES
Inflammation is the body’s defense against injury or infection. Acute inflammation is a short-term response wherein the immune system releases white blood cells to defend the area, causing visible redness and swelling. Although inflammation is crucial for fighting infections, chronic inflammation leads to various inflammatory disorders. PDCs have a broad range of applications, including peptides serving as carriers for a drug. Conjugation of anti-inflammatory drugs with peptidic components can result in targeted delivery of drugs to inflammation sites, such as bones or tumor cells.
Betulinic acid-peptide conjugates.
Betulinic acid is a triterpenoid natural product that has been reported to have antiviral, anticancer, anti-bacterial, antimalarial, and anti-inflammatory activities.138–140 Various strategies to increase its solubility and bioavailability have been employed in the past, and integration of amino acids in various positions of the natural product was demonstrated to increase water solubility.141 A series of betulinic acid derivatives with 1,2,3-triazole peptide fragments were developed as peptide-drug conjugates with anti-inflammatory activity (20, Figure 6B).142 The 1,2,3-triazole was introduced as a bioisostere for an amide bond with improved stability in physiological conditions. Anti-inflammatory activity for these conjugates was first tested in vivo through a histamine-induced paw edema model, where analogs conjugated with histidine, alanine, tryptophan and isoleucine residues exhibited anti-inflammatory effects. Comparison between the conjugates’ activity and a synthetic intermediate in their synthesis, a betulinic acid derivative with C-3 position bound to an alkyne group, showed inferior activity to its peptide-conjugated analogues.
Docking studies were employed to evaluate affinity of the peptide conjugates to the Kelch domain of the KEAP1 protein. The broad-complex, tramtrack, bric-à-brac and intervening region domains of KEAP1 are known molecular targets for triterpene scaffolds.143 KEAP1 is involved in antioxidant/anti-inflammatory activity through the NRF2/KEAP1/ARE pathway. Activation of this pathway has been reported to promote the suppression of pro-inflammatory enzymes.144 Although the conjugate’s affinity towards KEAP1’s Kelch domain was less than that of the native ligand, structural analysis revealed that the π-system of the dimethoxyphenyl group plays a role in the interaction with Kelch-domain amino acid residues. The absence of a correlation between the docking studies and the conjugates’ inflammatory activity may suggest that its activity involves a more complex mechanism that gives rise to their anti-inflammatory effects.
Peptide conjugates of the structurally related betulonic acid have also been studied for their anti-inflammatory activities after research showed that 1,2,3-triazole derivatives of the acid exhibited high hepatoprotective and anti-inflammatory activity.145 This prompted the synthesis of betulonic acid-conjugates that were further derivatized with 1,2,3-triazole peptide fragments.146 Six tripeptides were incorporated into the betulonic acid conjugates to increase aqueous solubility and enhance bioactivity. The peptide fragment and betulonic acid structure were linked through the 1,2,3-triazole moiety that was previously conjugated to the acid. Six conjugates were evaluated for their anti-inflammatory activity, but none of them showed promising activity.
Hyaluronic acid – methotrexate conjugates.
Arthritis is a chronic autoimmune disease that affects approximately 0.5% to 1% of the worldwide population.147 It leads to joint stiffness, deformity, and damage. Arthritis is characterized by recurring episodes of joint swelling, pain, and resultant disability, which significantly impact physical health, quality of life, employment, and expenses. Hyaluronic acid (HA) and methotrexate (MTX) conjugates have been studied as potential treatments for osteoarthritis (OA), based on HA’s pain-eliminating effect and MTX’s anti-inflammatory effect.148 These conjugates were proposed because of HA’s reported use as a carrier for various other medicinal agents and because of the need for MTX derivatives that retain anti-inflammatory activity but lack harmful adverse effects.149,150
Conjugation of the two clinically evaluated agents was done through a peptide chain sensitive to lysosomal enzymes. Upon cell uptake and peptide chain degradation, the conjugate would release the active form of MTX into the cell. Linkage of this peptide was studied using PEG-13 to eliminate potential steric hindrance related to interaction with proteases. This linker would further separate the HA backbone from the peptide chain, allowing the peptides to be accessible for cleavage in the lysosome. Conjugates with and without the peptide chain were synthesized to validate this reasoning (21, Figure 6C). The anti-proliferative effect of the conjugates was evaluated using human fibroblast-like synoviocytes (HFLS) stimulated by TNF-ɑ, and results showed that conjugates with the peptide chain inhibited proliferation, while linker-only conjugates had no anti-proliferative activity. The anti-inflammatory effects of these conjugates were also tested in vivo in an antigen-induced-osteoarthritis rat model. Two peptidic conjugates with different amino acid composition were tested alongside HA in this model. Neither intra-articularly injected MTX or HA had any effect on swelling. On the other hand, one of the conjugates showed significant reduction of swelling, and the other exhibited inhibition of swelling. This suggests that the composition of peptide chains plays a role in its lysosomal cleavage.
This compound was then further modified starting with the composition of its peptide chain.151 Peptide sequences were designed based on recognition sites for cathepsins B, D, or L, because they are the most abundant lysosomal enzymes in synovial tissues. Fragmentation resulting from cathepsin cleavage was studied through mass spectrometry. Only peptides with α-connected compounds were recognized and cleaved by cathepsins and released free MTX. A Phe-Phe sequence was deemed to be important in cathepsin recognition and cleavage because of the tendency of cathepsin D to hydrolyze inside the sequence and cathepsin B and L to hydrolyze after the sequence. Linker optimization was done by comparing different linker lengths, and although it was hypothesized that the most important factor was separation of MTX compound and HA, hydrophilicity turned out to be the leading factor in linker efficacy. In vitro, the three best linkers were PEG-8, PEG-5, and Alkyl2, which had the lowest Log P values. This correlated with in vivo efficacy of the conjugates and thus Alkyl2 was selected as the optimal linker because of its efficacy in both in vitro and in vivo studies and its low molecular weight. Molecular weight was also studied as a potential determining factor for conjugate activity. Low molecular-weight HA yielded low or no efficacy in vivo. On the other hand, conjugates with HA with higher molecular weight exhibited the same effects as free HA.
Methotrexate – cIBR conjugates.
Another approach on the development of MTX-peptide conjugates was explored through its conjugation to (cyclo(1,12)PenPRGGSVLVTGC) peptide (cIBR).152 Peptide cIBR mimics Domain-1 of the intracellular adhesion molecule-1 (ICAM-1), a protein that interacts with leukocyte function-associated antigen-1 (LFA-1) on T-cells.153 This interaction has been identified as a target for rheumatoid arthritis, because of its role in the formation of inflammatory cellular aggregation in synovial tissues, which would make the delivery of MTX more targeted to leukocytes and avoid side effects.154 cIBR was linked to MTX through an amide bond between the γ-carboxylic acid group of MTX and the N-terminus of the peptide (22, Figure 6D). In vivo efficacy of this conjugate was evaluated using the rat adjuvant arthritis model. Arthritic rats and normal control rats were treated with injections of different doses of MTX-cIBR and were monitored for ankle diameter, paw weight, inflammation, and bone resorption. Treatment showed reduced body weight and suppressed the progress of arthritis, with moderate synovium and no bone resorption at the physis. On the other hand, arthritic rats treated with vehicle exhibited severe periarticular inflammation, synovitis, and bone resorption. Uptake of MTX-cIBR by LFA-1 was evaluated in Molt-3 T cells by evaluating how anti-LFA-1 monoclonal antibody (mAb) and CIBR peptide would modulate the toxicity of the conjugate on the cells. Results showed that increasing anti-LFA-1 mAb and cIBR peptide concentrations increased cell viability, which suggests that MTX-cIBR uptake is mediated by LFA-1. Stability of the conjugate was studied at pH 1, 4, 5, 6, 7, 8, 10, and 12. Maximal stability was observed at pH 6, with the conjugate exhibiting a half life (t1/2) of 770 min, which suggests good stability and longer half-lives close to physiological pH (7.2). In vitro stability of MTX-cIBR was evaluated in rat plasma and homogenized rat heart tissue and was found to be 43.8 and 38.1 min, respectively.
β-Carboline-peptide conjugates.
A new class of β-carboline-peptide conjugates was studied for the treatment of ischemia/reperfusion (I/R) injuries, a condition caused by low/blocked blood-flow to tissues, leading to tissue and organ death.155,156 I/R injuries have been associated with an increase in ROS, leading to attack on cell-membrane constituents resulting in lipid peroxidation, DNA damage, release of proinflammatory substances, and increased formation of ROS, which leads to cell death.157,158 These events activate the inflammatory cascade, and ultimately result in organ failure and death. Existing therapies focus on anti-inflammatory, anti-leukocyte, and antioxidant agents. β-Carbolines are naturally occurring alkaloids that have been studied for their antioxidant properties, and some have been reported to be even more potent than ascorbic acid.158–161 Short peptide sequences such as Ala-Arg-Pro-Ala-Lys (ARPAK), Gly-Arg-Pro-Ala-Lys (GRPAK) and Gln-Arg-Pro-Ala-Lys (QRPAK), have thrombolytic activity and, in combination with β-carbolines, it was hypothesized that their combination would exhibit both anti-oxidant and thrombolytic activities that may have therapeutic potential in treating I/R injury.
β-Carboline alkaloids were first synthesized along with the unconjugated pentapeptides and were then coupled through an amide bond to produce nine conjugates.162 In vivo thrombolytic activity of the β-carboline-peptide conjugates was evaluated through comparison of thrombus mass reduction. Conjugates exhibited the same significant activity in reducing thrombus mass. The conjugates’ ability for free radical-scavenging was studied in vitro through a cell survival assay in rat pheochromocytoma (PC 12) cells, which are particularly sensitive to oxidative stress. The scavenging capacity of the conjugates compared to their parent species was comparable in increasing cell survival after exposure of cells to NO, H2O2, and HO−. Of the nine conjugates, only one (23, Figure 6E) was studied for its effect on lipid peroxidation-malondialdehyde (MDA), glutathione (GSH), and glutathione disulfide (GSSG) levels in the cell. 23 increased MDA in muscle tissues, which indicates its role in suppressing the lipid peroxidation cascade that results in inflammatory response. It was also found to reverse decreased GSH levels in tissue and blood by increasing the GSH/GSSG ratio, which demonstrates it lowers oxidative stress.
8. CONCLUSIONS
The rapid advancements in peptide chemistry have enabled the development of the next generation of peptide therapeutics that incorporate the potency of small molecules with the specificity of peptides in a class of compounds called PDCs. These multi-component molecules have had success overcoming drug-resistance mechanisms and treating complex disease states that present significant challenges to both peptides and small molecules independently. Unlike other bioconjugate therapeutics, such as ADCs, PDCs can be tailored to address a wide range of therapeutic challenges because of their smaller size and their ability to accommodate a wide array of small molecules. This perspective highlights the many considerations that go into PDC design and optimization, while also emphasizing several PDC series that demonstrate significant effectiveness against infections and disorders that lack adequate treatment. As drug resistance mechanisms develop proportionally with the increased prescription of modern medicines, new drugs will be needed to address these clinical challenges. It is our belief that PDCs represent an important part of the next generation of peptide therapeutics that will be required to overcome the rise in drug resistance and treat complex disease states.
SIGNIFICANCE.
The significance, impact, and innovation of this article:
Juxtaposition of peptide-drug conjugates and antibody-drug conjugates with a focus on challenges that can be overcome with peptide-drug conjugate strategies.
Considerations for the design and optimization of peptide-drug conjugates.
Analysis of several peptide-drug conjugate series that demonstrate effectiveness towards drug resistant infections and complex disease states.
ACKNOWLEDGMENTS
This work was supported by the National Institute of Allergy and Infectious Diseases (1 R01 AI162961), the National Institutes of General Medical Sciences (K12 GM139186), the American Cancer Society (RSG-21-037-01-CDD), and the University of Illinois Chicago Graduate School Access to Excellence Fellowship. The table of contents graphic and cartoon diagrams in Figures were generated using Biorender.com. The authors take this opportunity to congratulate the MIKIW Meeting-in-Miniature on its 60th anniversary.
ABBREVIATIONS USED
- Aβ
amyloid beta
- AChE
acetylcholinesterase
- AD
Alzheimer’s disease
- ADC
antibody-drug conjugate
- BACE1
β-site APP-cleaving enzyme 1
- BBB
blood-brain barrier
- BuChE
butyrylcholinesterase
- CNS
central nervous system
- DENV
dengue virus
- FDA
U.S Food and Drug Administration
- GK
glucokinase
- GSH
glutathione
- HA
hyaluronic acid
- HFLS
human fibroblast-like synoviocytes
- hIAPP
human islet amyloid polypeptide
- HIV
human immunodeficiency virus
- IC50
half-maximal inhibitory concentration
- ICAM-1
intracellular adhesion molecule-1
- IR
insulin receptor
- IRS
insulin receptor substrate
- I/R
ischemia/reperfusion
- LFA-1
leukocyte function-associated antigen-1
- LPS
lipopolysaccharide
- mAb
monoclonal antibody
- MDA
malondialdehyde
- MDR
multi-drug resistant
- MRSA
methicillin-resistant S. aureus
- MIC
minimum inhibitory concentration
- MTX
methotrexate
- ND
neurodegenerative diseases
- Nva
norvaline
- OA
osteoarthritis
- OM
outer membrane
- PC
pheochromocytoma
- PDC
peptide-drug conjugate
- PEG
polyethylene glycol
- PET
positron emission tomography
- PMEN
polymyxin E nonapeptide
- PROTAC
proteolysis-targeting chimera
- PTP1B
protein tyrosine phosphatase 1B
- RA
rheumatoid arthritis
- ROS
reactive oxygen species
- SAR
structure-activity relationship
- SARS-CoV2
Severe Acutre Respiratory Syndrome Coronavirus 2
- SPP
similarity property principle
- TfR-P
transferrin receptor binding peptide
- Tle
tert-lecuine
- VISA
vancomycin-intermediate S. aureus
- VRE
vancomycin-resistant enterocci
- VRSA
vancomycin-resistant S. aureus
- KOR
κ-opioid receptor
- MOR
μ-opioid receptor
Biographies
Biographies
Trevor T. Dean is a Pharmaceutical Sciences graduate student in the Chemistry and Drug Discovery concentration at UIC. He completed his H.BSc in biochemistry at McGill University in 2019. He later completed his MSc at the University of Toronto in 2022 where he used molecular and structural biology techniques to study viral proteins. In the Moore lab, Trevor develops binding assays to study antimicrobial peptides such as apidaecin.
Juliet Jelu-Reyes is a Pharmaceutical Sciences graduate student in the Chemistry in Drug Discovery concentration at UIC. She received her B.S. in Chemistry from the University of Puerto Rico Rio Piedras in 2022. Her research focuses on the development of stapled peptides that modulate the estrogen receptor to address breast cancer.
A’Lester C. Allen is an IRACDA postdoc affiliated with the Moore lab at UIC. He graduated with a PhD in chemistry in 2022 from the University of California Santa Cruz, where he studied plasmonic nanoparticles to detect protein and lipid biomarkers using surface-enhanced Raman scattering. He holds an MS in materials engineering from San Jose State University, and a BS in Chemistry from Stanford University. He joined the Moore lab in 2022 and contributes to the apidaecin project.
Terry W. Moore is an Associate Professor of Pharmaceutical Sciences at the University of Illinois Chicago. He holds a PhD in organic chemistry from the University of Illinois at Urbana-Champaign, and a BA in biochemistry from Abilene Christian University. His research interests include small molecules and peptides active against a number of different molecular targets.
Footnotes
The authors declare no competing financial interest.
Contributor Information
Trevor T. Dean, Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois, 60612, United States
Juliet Jelu-Reyes, Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois, 60612, United States.
A’Lester C. Allen, Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois, 60612, United States
Terry W. Moore, Department of Pharmaceutical Sciences and University of Illinois Cancer Center, University of Illinois Chicago, Chicago, Illinois, 60612, United States
REFERENCES
- (1).Ngo HX; Garneau-Tsodikova S What Are the Drugs of the Future? Medchemcomm 2018, 9 (5), 757–758. 10.1039/c8md90019a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (2).Drucker DJ Advances in Oral Peptide Therapeutics. Nat Rev Drug Discov 2020, 19 (4), 277–289. 10.1038/s41573-019-0053-0. [DOI] [PubMed] [Google Scholar]
- (3).Santos GB; Ganesan A; Emery FS Oral Administration of Peptide-Based Drugs: Beyond Lipinski’s Rule. ChemMedChem 2016, 11 (20), 2245–2251. 10.1002/cmdc.201600288. [DOI] [PubMed] [Google Scholar]
- (4).Lau JL; Dunn MK Therapeutic Peptides: Historical Perspectives, Current Development Trends, and Future Directions. Bioorg Med Chem 2018, 26 (10), 2700–2707. 10.1016/j.bmc.2017.06.052. [DOI] [PubMed] [Google Scholar]
- (5).Diao L; Meibohm B Pharmacokinetics and Pharmacokinetic-Pharmacodynamic Correlations of Therapeutic Peptides. Clin Pharmacokinet 2013, 52 (10), 855–868. 10.1007/s40262-013-0079-0. [DOI] [PubMed] [Google Scholar]
- (6).Kumari S; Carmona AV; Tiwari AK; Trippier PC Amide Bond Bioisosteres: Strategies, Synthesis, and Successes. J Med Chem 2020, 63 (21), 12290–12358. 10.1021/acs.jmedchem.0c00530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (7).Skowron KJ; Speltz TE; Moore TW Recent Structural Advances in Constrained Helical Peptides. Med Res Rev 2019, 39 (2), 749–770. 10.1002/med.21540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (8).Khakshoor O; Demeler B; Nowick JS Macrocyclic Beta-Sheet Peptides That Mimic Protein Quaternary Structure through Intermolecular Beta-Sheet Interactions. J Am Chem Soc 2007, 129 (17), 5558–5569. 10.1021/ja068511u. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (9).Deming TJ Functional Modification of Thioether Groups in Peptides, Polypeptides, and Proteins. Bioconjug Chem 2017, 28 (3), 691–700. 10.1021/acs.bioconjchem.6b00696. [DOI] [PubMed] [Google Scholar]
- (10).Best MD Click Chemistry and Bioorthogonal Reactions: Unprecedented Selectivity in the Labeling of Biological Molecules. Biochemistry 2009, 48 (28), 6571–6584. 10.1021/bi9007726. [DOI] [PubMed] [Google Scholar]
- (11).Kim CH; Axup JY; Schultz PG Protein Conjugation with Genetically Encoded Unnatural Amino Acids. Curr Opin Chem Biol 2013, 17 (3), 412–419. 10.1016/j.cbpa.2013.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (12).Kurtzhals P; Østergaard S; Nishimura E; Kjeldsen T Derivatization with Fatty Acids in Peptide and Protein Drug Discovery. Nat Rev Drug Discov 2023, 22 (1), 59–80. 10.1038/s41573-022-00529-w. [DOI] [PubMed] [Google Scholar]
- (13).Mohan T; Kleinschek KS; Kargl R Polysaccharide Peptide Conjugates: Chemistry, Properties and Applications. Carbohydr Polym 2022, 280, 118875. 10.1016/j.carbpol.2021.118875. [DOI] [PubMed] [Google Scholar]
- (14).Hamley IW PEG-Peptide Conjugates. Biomacromolecules 2014, 15 (5), 1543–1559. 10.1021/bm500246w. [DOI] [PubMed] [Google Scholar]
- (15).Baumann A; Tuerck D; Prabhu S; Dickmann L; Sims J Pharmacokinetics, Metabolism and Distribution of PEGs and PEGylated Proteins: Quo Vadis? Drug Discov Today 2014, 19 (10), 1623–1631. 10.1016/j.drudis.2014.06.002. [DOI] [PubMed] [Google Scholar]
- (16).Otvos L; Ostorhazi E; Szabo D; Zumbrun SD; Miller LL; Halasohoris SA; Desai PD; Int Veldt SM; Kraus CN Synergy Between Proline-Rich Antimicrobial Peptides and Small Molecule Antibiotics Against Selected Gram-Negative Pathogens in Vitro and in Vivo. Front Chem 2018, 6, 309. 10.3389/fchem.2018.00309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (17).Hilchie AL; Sharon AJ; Haney EF; Hoskin DW; Bally MB; Franco OL; Corcoran JA; Hancock REW Mastoparan Is a Membranolytic Anti-Cancer Peptide That Works Synergistically with Gemcitabine in a Mouse Model of Mammary Carcinoma. Biochim Biophys Acta 2016, 1858 (12), 3195–3204. 10.1016/j.bbamem.2016.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (18).Chau CH; Steeg PS; Figg WD Antibody-Drug Conjugates for Cancer. Lancet 2019, 394 (10200), 793–804. 10.1016/S0140-6736(19)31774-X. [DOI] [PubMed] [Google Scholar]
- (19).Dumontet C; Reichert JM; Senter PD; Lambert JM; Beck A Antibody-Drug Conjugates Come of Age in Oncology. Nat Rev Drug Discov 2023, 22 (8), 641–661. 10.1038/s41573-023-00709-2. [DOI] [PubMed] [Google Scholar]
- (20).Gaston J; Maestrali N; Lalle G; Gagnaire M; Masiero A; Dumas B; Dabdoubi T; Radošević K; Berne P-F Intracellular Delivery of Therapeutic Antibodies into Specific Cells Using Antibody-Peptide Fusions. Sci Rep 2019, 9 (1), 18688. 10.1038/s41598-019-55091-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (21).Ritchie M; Tchistiakova L; Scott N Implications of Receptor-Mediated Endocytosis and Intracellular Trafficking Dynamics in the Development of Antibody Drug Conjugates. MAbs 2013, 5 (1), 13–21. 10.4161/mabs.22854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (22).Birrer MJ; Moore KN; Betella I; Bates RC Antibody-Drug Conjugate-Based Therapeutics: State of the Science. J Natl Cancer Inst 2019, 111 (6), 538–549. 10.1093/jnci/djz035. [DOI] [PubMed] [Google Scholar]
- (23).Brandsch M; Knütter I; Bosse-Doenecke E Pharmaceutical and Pharmacological Importance of Peptide Transporters. J Pharm Pharmacol 2008, 60 (5), 543–585. 10.1211/jpp.60.5.0002. [DOI] [PubMed] [Google Scholar]
- (24).Chari RVJ; Miller ML; Widdison WC Antibody-Drug Conjugates: An Emerging Concept in Cancer Therapy. Angew Chem Int Ed Engl 2014, 53 (15), 3796–3827. 10.1002/anie.201307628. [DOI] [PubMed] [Google Scholar]
- (25).Békés M; Langley DR; Crews CM PROTAC Targeted Protein Degraders: The Past Is Prologue. Nat Rev Drug Discov 2022, 21 (3), 181–200. 10.1038/s41573-021-00371-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (26).Sornay C; Vaur V; Wagner A; Chaubet G An Overview of Chemo- and Site-Selectivity Aspects in the Chemical Conjugation of Proteins. R Soc Open Sci 2022, 9 (1), 211563. 10.1098/rsos.211563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (27).Hamblett KJ; Senter PD; Chace DF; Sun MMC; Lenox J; Cerveny CG; Kissler KM; Bernhardt SX; Kopcha AK; Zabinski RF; Meyer DL; Francisco JA Effects of Drug Loading on the Antitumor Activity of a Monoclonal Antibody Drug Conjugate. Clin Cancer Res 2004, 10 (20), 7063–7070. 10.1158/1078-0432.CCR-04-0789. [DOI] [PubMed] [Google Scholar]
- (28).Matsuda Y; Mendelsohn BA Recent Advances in Drug-Antibody Ratio Determination of Antibody-Drug Conjugates. Chem Pharm Bull (Tokyo) 2021, 69 (10), 976–983. 10.1248/cpb.c21-00258. [DOI] [PubMed] [Google Scholar]
- (29).Adem YT; Schwarz KA; Duenas E; Patapoff TW; Galush WJ; Esue O Auristatin Antibody Drug Conjugate Physical Instability and the Role of Drug Payload. Bioconjug Chem 2014, 25 (4), 656–664. 10.1021/bc400439x. [DOI] [PubMed] [Google Scholar]
- (30).Ashman N; Bargh JD; Spring DR Non-Internalising Antibody-Drug Conjugates. Chem Soc Rev 2022, 51 (22), 9182–9202. 10.1039/d2cs00446a. [DOI] [PubMed] [Google Scholar]
- (31).Sheyi R; de la Torre BG; Albericio F Linkers: An Assurance for Controlled Delivery of Antibody-Drug Conjugate. Pharmaceutics 2022, 14 (2), 396. 10.3390/pharmaceutics14020396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (32).Yamada K; Ito Y Recent Chemical Approaches for Site-Specific Conjugation of Native Antibodies: Technologies toward Next-Generation Antibody-Drug Conjugates. Chembiochem 2019, 20 (21), 2729–2737. 10.1002/cbic.201900178. [DOI] [PubMed] [Google Scholar]
- (33).Guidotti G; Brambilla L; Rossi D Cell-Penetrating Peptides: From Basic Research to Clinics. Trends Pharmacol Sci 2017, 38 (4), 406–424. 10.1016/j.tips.2017.01.003. [DOI] [PubMed] [Google Scholar]
- (34).Cheng AV; Wuest WM Signed, Sealed, Delivered: Conjugate and Prodrug Strategies as Targeted Delivery Vectors for Antibiotics. ACS Infect Dis 2019, 5 (6), 816–828. 10.1021/acsinfecdis.9b00019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (35).Lu J; Jiang F; Lu A; Zhang G Linkers Having a Crucial Role in Antibody-Drug Conjugates. Int J Mol Sci 2016, 17 (4), 561. 10.3390/ijms17040561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (36).Bargh JD; Isidro-Llobet A; Parker JS; Spring DR Cleavable Linkers in Antibody-Drug Conjugates. Chem Soc Rev 2019, 48 (16), 4361–4374. 10.1039/c8cs00676h. [DOI] [PubMed] [Google Scholar]
- (37).Szijj PA; Kostadinova KA; Spears RJ; Chudasama V Tyrosine Bioconjugation - an Emergent Alternative. Org Biomol Chem 2020, 18 (44), 9018–9028. 10.1039/d0ob01912g. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (38).Chavda VP; Solanki HK; Davidson M; Apostolopoulos V; Bojarska J Peptide-Drug Conjugates: A New Hope for Cancer Management. Molecules 2022, 27 (21), 7232. 10.3390/molecules27217232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (39).Wang Y; Cheetham AG; Angacian G; Su H; Xie L; Cui H Peptide-Drug Conjugates as Effective Prodrug Strategies for Targeted Delivery. Adv Drug Deliv Rev 2017, 110–111, 112–126. 10.1016/j.addr.2016.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (40).Richman DD Antiviral Drug Resistance. Antiviral Res 2006, 71 (2–3), 117–121. 10.1016/j.antiviral.2006.03.004. [DOI] [PubMed] [Google Scholar]
- (41).Ng WM; Stelfox AJ; Bowden TA Unraveling Virus Relationships by Structure-Based Phylogenetic Classification. Virus Evol 2020, 6 (1), veaa003. 10.1093/ve/veaa003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (42).Dean TT; Serrão VHB; Lee JE Structure of the Core Postfusion Porcine Endogenous Retrovirus Fusion Protein. mBio 2022, 13 (1), e0292021. 10.1128/mbio.02920-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (43).De Clercq E Emerging Anti-HIV Drugs. Expert Opin Emerg Drugs 2005, 10 (2), 241–273. 10.1517/14728214.10.2.241. [DOI] [PubMed] [Google Scholar]
- (44).Ramdas P; Sahu AK; Mishra T; Bhardwaj V; Chande A From Entry to Egress: Strategic Exploitation of the Cellular Processes by HIV-1. Front Microbiol 2020, 11, 559792. 10.3389/fmicb.2020.559792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (45).Azimi FC; Dean TT; Minari K; Basso LGM; Vance TDR; Serrão VHB A Frame-by-Frame Glance at Membrane Fusion Mechanisms: From Viral Infections to Fertilization. Biomolecules 2023, 13 (7), 1130. 10.3390/biom13071130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (46).Liu S; Lu H; Niu J; Xu Y; Wu S; Jiang S Different from the HIV Fusion Inhibitor C34, the Anti-HIV Drug Fuzeon (T-20) Inhibits HIV-1 Entry by Targeting Multiple Sites in Gp41 and Gp120. J Biol Chem 2005, 280 (12), 11259–11273. 10.1074/jbc.M411141200. [DOI] [PubMed] [Google Scholar]
- (47).Dingens AS; Arenz D; Overbaugh J; Bloom JD Massively Parallel Profiling of HIV-1 Resistance to the Fusion Inhibitor Enfuvirtide. Viruses 2019, 11 (5), 439. 10.3390/v11050439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (48).Shi W; Cai L; Lu L; Wang C; Wang K; Xu L; Zhang S; Han H; Jiang X; Zheng B; Jiang S; Liu K Design of Highly Potent HIV Fusion Inhibitors Based on Artificial Peptide Sequences. Chem Commun (Camb) 2012, 48 (94), 11579–11581. 10.1039/c2cc35973a. [DOI] [PubMed] [Google Scholar]
- (49).Zhu X; Zhu Y; Ye S; Wang Q; Xu W; Su S; Sun Z; Yu F; Liu Q; Wang C; Zhang T; Zhang Z; Zhang X; Xu J; Du L; Liu K; Lu L; Zhang R; Jiang S Improved Pharmacological and Structural Properties of HIV Fusion Inhibitor AP3 over Enfuvirtide: Highlighting Advantages of Artificial Peptide Strategy. Sci Rep 2015, 5, 13028. 10.1038/srep13028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (50).Chong H; Yao X; Qiu Z; Sun J; Zhang M; Waltersperger S; Wang M; Liu S-L; Cui S; He Y Short-Peptide Fusion Inhibitors with High Potency against Wild-Type and Enfuvirtide-Resistant HIV-1. FASEB J 2013, 27 (3), 1203–1213. 10.1096/fj.12-222547. [DOI] [PubMed] [Google Scholar]
- (51).Frey G; Rits-Volloch S; Zhang X-Q; Schooley RT; Chen B; Harrison SC Small Molecules That Bind the Inner Core of Gp41 and Inhibit HIV Envelope-Mediated Fusion. Proc Natl Acad Sci U S A 2006, 103 (38), 13938–13943. 10.1073/pnas.0601036103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (52).Wang C; Shi W; Cai L; Lu L; Wang Q; Zhang T; Li J; Zhang Z; Wang K; Xu L; Jiang X; Jiang S; Liu K Design, Synthesis, and Biological Evaluation of Highly Potent Small Molecule-Peptide Conjugates as New HIV-1 Fusion Inhibitors. J Med Chem 2013, 56 (6), 2527–2539. 10.1021/jm3018964. [DOI] [PubMed] [Google Scholar]
- (53).Jiang S; Lu H; Liu S; Zhao Q; He Y; Debnath AK N-Substituted Pyrrole Derivatives as Novel Human Immunodeficiency Virus Type 1 Entry Inhibitors That Interfere with the Gp41 Six-Helix Bundle Formation and Block Virus Fusion. Antimicrob Agents Chemother 2004, 48 (11), 4349–4359. 10.1128/AAC.48.11.4349-4359.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (54).Wang C; Lu L; Na H; Li X; Wang Q; Jiang X; Xu X; Yu F; Zhang T; Li J; Zhang Z; Zheng B; Liang G; Cai L; Jiang S; Liu K Conjugation of a Nonspecific Antiviral Sapogenin with a Specific HIV Fusion Inhibitor: A Promising Strategy for Discovering New Antiviral Therapeutics. J Med Chem 2014, 57 (17), 7342–7354. 10.1021/jm500763m. [DOI] [PubMed] [Google Scholar]
- (55).Wang C; Wang X; Wang H; Pu J; Li Q; Li J; Liu Y; Lu L; Jiang SA “Two-Birds-One-Stone” Approach toward the Design of Bifunctional Human Immunodeficiency Virus Type 1 Entry Inhibitors Targeting the CCR5 Coreceptor and Gp41 N-Terminal Heptad Repeat Region. J Med Chem 2021, 64 (15), 11460–11471. 10.1021/acs.jmedchem.1c00781. [DOI] [PubMed] [Google Scholar]
- (56).Wang C; Wang H; Wang X; Sun L; Wang Q; Li Q; Liang R; Dou D; Yu F; Lu L; Jiang S Multitargeted Drug Design Strategy for Discovery of Short-Peptide-Based HIV-1 Entry Inhibitors with High Potency. Eur J Med Chem 2023, 252, 115294. 10.1016/j.ejmech.2023.115294. [DOI] [PubMed] [Google Scholar]
- (57).Imamura S; Ichikawa T; Nishikawa Y; Kanzaki N; Takashima K; Niwa S; Iizawa Y; Baba M; Sugihara Y Discovery of a Piperidine-4-Carboxamide CCR5 Antagonist (TAK-220) with Highly Potent Anti-HIV-1 Activity. J Med Chem 2006, 49 (9), 2784–2793. 10.1021/jm051034q. [DOI] [PubMed] [Google Scholar]
- (58).Dong M; Lu L; Li H; Wang X; Lu H; Jiang S; Dai Q Design, Synthesis, and Biological Activity of Novel 1,4-Disubstituted Piperidine/Piperazine Derivatives as CCR5 Antagonist-Based HIV-1 Entry Inhibitors. Bioorg Med Chem Lett 2012, 22 (9), 3284–3286. 10.1016/j.bmcl.2012.03.019. [DOI] [PubMed] [Google Scholar]
- (59).Rashad AA; Song L-R; Holmes AP; Acharya K; Zhang S; Wang Z-L; Gary E; Xie X; Pirrone V; Kutzler MA; Long Y-Q; Chaiken I Bifunctional Chimera That Coordinately Targets Human Immunodeficiency Virus 1 Envelope Gp120 and the Host-Cell CCR5 Coreceptor at the Virus-Cell Interface. J Med Chem 2018, 61 (11), 5020–5033. 10.1021/acs.jmedchem.8b00477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (60).Meng G; Pu J; Li Y; Han A; Tian Y; Xu W; Zhang T; Li X; Lu L; Wang C; Jiang S; Liu K Design and Biological Evaluation of M-Xylene Thioether-Stapled Short Helical Peptides Targeting the HIV-1 Gp41 Hexameric Coiled-Coil Fusion Complex. J Med Chem 2019, 62 (19), 8773–8783. 10.1021/acs.jmedchem.9b00882. [DOI] [PubMed] [Google Scholar]
- (61).Wang X; Xu W; Liu Z; Wu Y; Wang Q; Cao M; Ying T; He N; Lu L; Jiang S A Toxin-Conjugated Recombinant Protein Targeting Gp120 and Gp41 for Inactivating HIV-1 Virions and Killing Latency-Reversing Agent-Reactivated Latent Cells. mBio 2022, 13 (1), e0338421. 10.1128/mbio.03384-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (62).La Ruche G; Souarès Y; Armengaud A; Peloux-Petiot F; Delaunay P; Desprès P; Lenglet A; Jourdain F; Leparc-Goffart I; Charlet F; Ollier L; Mantey K; Mollet T; Fournier JP; Torrents R; Leitmeyer K; Hilairet P; Zeller H; Van Bortel W; Dejour-Salamanca D; Grandadam M; Gastellu-Etchegorry M First Two Autochthonous Dengue Virus Infections in Metropolitan France, September 2010. Euro Surveill 2010, 15 (39), 19676. [PubMed] [Google Scholar]
- (63).Perera R; Kuhn RJ Structural Proteomics of Dengue Virus. Curr Opin Microbiol 2008, 11 (4), 369–377. 10.1016/j.mib.2008.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (64).Nitsche C; Holloway S; Schirmeister T; Klein CD Biochemistry and Medicinal Chemistry of the Dengue Virus Protease. Chem Rev 2014, 114 (22), 11348–11381. 10.1021/cr500233q. [DOI] [PubMed] [Google Scholar]
- (65).Leung D; Schroder K; White H; Fang NX; Stoermer MJ; Abbenante G; Martin JL; Young PR; Fairlie DP Activity of Recombinant Dengue 2 Virus NS3 Protease in the Presence of a Truncated NS2B Co-Factor, Small Peptide Substrates, and Inhibitors. J Biol Chem 2001, 276 (49), 45762–45771. 10.1074/jbc.M107360200. [DOI] [PubMed] [Google Scholar]
- (66).Li J; Lim SP; Beer D; Patel V; Wen D; Tumanut C; Tully DC; Williams JA; Jiricek J; Priestle JP; Harris JL; Vasudevan SG Functional Profiling of Recombinant NS3 Proteases from All Four Serotypes of Dengue Virus Using Tetrapeptide and Octapeptide Substrate Libraries. J Biol Chem 2005, 280 (31), 28766–28774. 10.1074/jbc.M500588200. [DOI] [PubMed] [Google Scholar]
- (67).Yin Z; Patel SJ; Wang W-L; Wang G; Chan W-L; Rao KRR; Alam J; Jeyaraj DA; Ngew X; Patel V; Beer D; Lim SP; Vasudevan SG; Keller TH Peptide Inhibitors of Dengue Virus NS3 Protease. Part 1: Warhead. Bioorg Med Chem Lett 2006, 16 (1), 36–39. 10.1016/j.bmcl.2005.09.062. [DOI] [PubMed] [Google Scholar]
- (68).Nitsche C; Steuer C; Klein CD Arylcyanoacrylamides as Inhibitors of the Dengue and West Nile Virus Proteases. Bioorg Med Chem 2011, 19 (24), 7318–7337. 10.1016/j.bmc.2011.10.061. [DOI] [PubMed] [Google Scholar]
- (69).Mendgen T; Steuer C; Klein CD Privileged Scaffolds or Promiscuous Binders: A Comparative Study on Rhodanines and Related Heterocycles in Medicinal Chemistry. J Med Chem 2012, 55 (2), 743–753. 10.1021/jm201243p. [DOI] [PubMed] [Google Scholar]
- (70).Nitsche C; Behnam MAM; Steuer C; Klein CD Retro Peptide-Hybrids as Selective Inhibitors of the Dengue Virus NS2B-NS3 Protease. Antiviral Res 2012, 94 (1), 72–79. 10.1016/j.antiviral.2012.02.008. [DOI] [PubMed] [Google Scholar]
- (71).Nitsche C; Schreier VN; Behnam MAM; Kumar A; Bartenschlager R; Klein CD Thiazolidinone-Peptide Hybrids as Dengue Virus Protease Inhibitors with Antiviral Activity in Cell Culture. J Med Chem 2013, 56 (21), 8389–8403. 10.1021/jm400828u. [DOI] [PubMed] [Google Scholar]
- (72).Kaur Manjal S; Kaur R; Bhatia R; Kumar K; Singh V; Shankar R; Kaur R; Rawal RK Synthetic and Medicinal Perspective of Thiazolidinones: A Review. Bioorg Chem 2017, 75, 406–423. 10.1016/j.bioorg.2017.10.014. [DOI] [PubMed] [Google Scholar]
- (73).Behrouz S; Kühl N; Klein CD N-Sulfonyl Peptide-Hybrids as a New Class of Dengue Virus Protease Inhibitors. Eur J Med Chem 2023, 251, 115227. 10.1016/j.ejmech.2023.115227. [DOI] [PubMed] [Google Scholar]
- (74).Dražić T; Kopf S; Corridan J; Leuthold MM; Bertoša B; Klein CD Peptide-β-Lactam Inhibitors of Dengue and West Nile Virus NS2B-NS3 Protease Display Two Distinct Binding Modes. J Med Chem 2020, 63 (1), 140–156. 10.1021/acs.jmedchem.9b00759. [DOI] [PubMed] [Google Scholar]
- (75).Ghaffar KA; Hussein WM; Khalil ZG; Capon RJ; Skwarczynski M; Toth I Levofloxacin and Indolicidin for Combination Antimicrobial Therapy. Curr Drug Deliv 2015, 12 (1), 108–114. 10.2174/1567201811666140910094050. [DOI] [PubMed] [Google Scholar]
- (76).Chen H; Liu C; Chen D; Madrid K; Peng S; Dong X; Zhang M; Gu Y Bacteria-Targeting Conjugates Based on Antimicrobial Peptide for Bacteria Diagnosis and Therapy. Mol Pharm 2015, 12 (7), 2505–2516. 10.1021/acs.molpharmaceut.5b00053. [DOI] [PubMed] [Google Scholar]
- (77).Bera S; Zhanel GG; Schweizer F Synthesis and Antibacterial Activity of Amphiphilic Lysine-Ligated Neomycin B Conjugates. Carbohydr Res 2011, 346 (5), 560–568. 10.1016/j.carres.2011.01.015. [DOI] [PubMed] [Google Scholar]
- (78).Yadav S; Mahato M; Pathak R; Jha D; Kumar B; Deka SR; Gautam HK; Sharma AK Multifunctional Self-Assembled Cationic Peptide Nanostructures Efficiently Carry Plasmid DNA in Vitro and Exhibit Antimicrobial Activity with Minimal Toxicity. J Mater Chem B 2014, 2 (30), 4848–4861. 10.1039/c4tb00657g. [DOI] [PubMed] [Google Scholar]
- (79).Zhang F; Zhong C; Yao J; Zhang J; Zhang T; Li B; Gou S; Ni J Antimicrobial Peptides–Antibiotics Combination: An Effective Strategy Targeting Drug‐resistant Gram‐negative Bacteria. Peptide Science 2022, 114 (4), e24261. 10.1002/pep2.24261. [DOI] [Google Scholar]
- (80).Domalaon R; Zhanel GG; Schweizer F Short Antimicrobial Peptides and Peptide Scaffolds as Promising Antibacterial Agents. Curr Top Med Chem 2016, 16 (11), 1217–1230. 10.2174/1568026615666150915112459. [DOI] [PubMed] [Google Scholar]
- (81).Gao X; Ding J; Liao C; Xu J; Liu X; Lu W Defensins: The Natural Peptide Antibiotic. Adv Drug Deliv Rev 2021, 179, 114008. 10.1016/j.addr.2021.114008. [DOI] [PubMed] [Google Scholar]
- (82).David AA; Park SE; Parang K; Tiwari RK Antibiotics-Peptide Conjugates Against Multidrug-Resistant Bacterial Pathogens. Curr Top Med Chem 2018, 18 (22), 1926–1936. 10.2174/1568026619666181129141524. [DOI] [PubMed] [Google Scholar]
- (83).Zeiders SM; Chmielewski J Antibiotic-Cell-Penetrating Peptide Conjugates Targeting Challenging Drug-Resistant and Intracellular Pathogenic Bacteria. Chem Biol Drug Des 2021, 98 (5), 762–778. 10.1111/cbdd.13930. [DOI] [PubMed] [Google Scholar]
- (84).Levine DP Vancomycin: A History. Clin Infect Dis 2006, 42 Suppl 1, S5–12. 10.1086/491709. [DOI] [PubMed] [Google Scholar]
- (85).Blaskovich MAT; Hansford KA; Butler MS; Jia Z; Mark AE; Cooper MA Developments in Glycopeptide Antibiotics. ACS Infect Dis 2018, 4 (5), 715–735. 10.1021/acsinfecdis.7b00258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (86).Stogios PJ; Savchenko A Molecular Mechanisms of Vancomycin Resistance. Protein Sci 2020, 29 (3), 654–669. 10.1002/pro.3819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (87).Jelinkova P; Splichal Z; Jimenez AMJ; Haddad Y; Mazumdar A; Sur VP; Milosavljevic V; Kopel P; Buchtelova H; Guran R; Zitka O; Richtera L; Hegerova D; Heger Z; Moulick A; Adam V Novel Vancomycin-Peptide Conjugate as Potent Antibacterial Agent against Vancomycin-Resistant Staphylococcus Aureus. Infect Drug Resist 2018, 11, 1807–1817. 10.2147/IDR.S160975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (88).Jiang Y; Han M; Bo Y; Feng Y; Li W; Wu JR; Song Z; Zhao Z; Tan Z; Chen Y; Xue T; Fu Z; Kuo SH; Lau GW; Luijten E; Cheng J “Metaphilic” Cell-Penetrating Polypeptide-Vancomycin Conjugate Efficiently Eradicates Intracellular Bacteria via a Dual Mechanism. ACS Cent Sci 2020, 6 (12), 2267–2276. 10.1021/acscentsci.0c00893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (89).Blaskovich MAT; Hansford KA; Gong Y; Butler MS; Muldoon C; Huang JX; Ramu S; Silva AB; Cheng M; Kavanagh AM; Ziora Z; Premraj R; Lindahl F; Bradford TA; Lee JC; Karoli T; Pelingon R; Edwards DJ; Amado M; Elliott AG; Phetsang W; Daud NH; Deecke JE; Sidjabat HE; Ramaologa S; Zuegg J; Betley JR; Beevers APG; Smith RAG; Roberts JA; Paterson DL; Cooper MA Protein-Inspired Antibiotics Active against Vancomycin- and Daptomycin-Resistant Bacteria. Nat Commun 2018, 9 (1), 22. 10.1038/s41467-017-02123-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (90).Umstätter F; Domhan C; Hertlein T; Ohlsen K; Mühlberg E; Kleist C; Zimmermann S; Beijer B; Klika KD; Haberkorn U; Mier W; Uhl P Vancomycin Resistance Is Overcome by Conjugation of Polycationic Peptides. Angew Chem Int Ed Engl 2020, 59 (23), 8823–8827. 10.1002/anie.202002727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (91).Antonoplis A; Zang X; Huttner MA; Chong KKL; Lee YB; Co JY; Amieva MR; Kline KA; Wender PA; Cegelski L A Dual-Function Antibiotic-Transporter Conjugate Exhibits Superior Activity in Sterilizing MRSA Biofilms and Killing Persister Cells. J Am Chem Soc 2018, 140 (47), 16140–16151. 10.1021/jacs.8b08711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (92).Shi W; Chen F; Zou X; Jiao S; Wang S; Hu Y; Lan L; Tang F; Huang W Design, Synthesis, and Antibacterial Evaluation of Vancomycin-LPS Binding Peptide Conjugates. Bioorg Med Chem Lett 2021, 45, 128122. 10.1016/j.bmcl.2021.128122. [DOI] [PubMed] [Google Scholar]
- (93).van Groesen E; Slingerland CJ; Innocenti P; Mihajlovic M; Masereeuw R; Martin NI Vancomyxins: Vancomycin-Polymyxin Nonapeptide Conjugates That Retain Anti-Gram-Positive Activity with Enhanced Potency against Gram-Negative Strains. ACS Infect Dis 2021, 7 (9), 2746–2754. 10.1021/acsinfecdis.1c00318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (94).Agnati LF; Zoli M; Biagini G; Fuxe K Neuronal Plasticity and Ageing Processes in the Frame of the ‘Red Queen Theory.’ Acta Physiologica Scandinavica 1992, 145 (4), 301–309. 10.1111/j.1748-1716.1992.tb09370.x. [DOI] [PubMed] [Google Scholar]
- (95).Agnatic LF; Benfenati F; Solfrini V; Biagini G; Fuxe K; Guidolin D; Carani C; Zini I Brain Aging and Neuronal Plasticity. Ann NY Acad Sci 1992, 673 (1 Physiopatholo), 180–186. 10.1111/j.1749-6632.1992.tb27451.x. [DOI] [PubMed] [Google Scholar]
- (96).Gitler AD; Dhillon P; Shorter J Neurodegenerative Disease: Models, Mechanisms, and a New Hope. Disease Models & Mechanisms 2017, 10 (5), 499–502. 10.1242/dmm.030205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (97).Gammon K Neurodegenerative Disease: Brain Windfall. Nature 2014, 515 (7526), 299–300. 10.1038/nj7526-299a. [DOI] [PubMed] [Google Scholar]
- (98).Kumar A; Singh A; Ekavali. A Review on Alzheimer’s Disease Pathophysiology and Its Management: An Update. Pharmacological Reports 2015, 67 (2), 195–203. 10.1016/j.pharep.2014.09.004. [DOI] [PubMed] [Google Scholar]
- (99).Scheltens P; Blennow K; Breteler MMB; De Strooper B; Frisoni GB; Salloway S; Van Der Flier WM Alzheimer’s Disease. The Lancet 2016, 388 (10043), 505–517. 10.1016/S0140-6736(15)01124-1. [DOI] [PubMed] [Google Scholar]
- (100).Wang J; Gu BJ; Masters CL; Wang Y-J A Systemic View of Alzheimer Disease — Insights from Amyloid-β Metabolism beyond the Brain. Nat Rev Neurol 2017, 13 (10), 612–623. 10.1038/nrneurol.2017.111. [DOI] [PubMed] [Google Scholar]
- (101).Esch T; Stefano GB; Fricchione GL; Benson H The Role of Stress in Neurodegenerative Diseases and Mental Disorders. Neuro Endocrinol Lett 2002, 23 (3), 199–208. [PubMed] [Google Scholar]
- (102).Lamptey RNL; Chaulagain B; Trivedi R; Gothwal A; Layek B; Singh J A Review of the Common Neurodegenerative Disorders: Current Therapeutic Approaches and the Potential Role of Nanotherapeutics. IJMS 2022, 23 (3), 1851. 10.3390/ijms23031851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (103).Allan SM; Rothwell NJ Inflammation in Central Nervous System Injury. Phil. Trans. R. Soc. Lond. B 2003, 358 (1438), 1669–1677. 10.1098/rstb.2003.1358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (104).Brouwer-DudokdeWit AC; Savenije A; Zoeteweij MW; Maat-Kievit A; Tibben A A Hereditary Disorder In the Family and the Family Life Cycle: Huntington Disease as a Paradigm. Family Process 2002, 41 (4), 677–692. 10.1111/j.1545-5300.2002.00677.x. [DOI] [PubMed] [Google Scholar]
- (105).Thomsen T; Kewitz H Selective Inhibition of Human Acetylcholinesterase by Galanthamine in Vitro and in Vivo. Life Sciences 1990, 46 (21), 1553–1558. 10.1016/0024-3205(90)90429-U. [DOI] [PubMed] [Google Scholar]
- (106).Nakagawa R; Takashi O; Kobayashi H; Yamaoka T; Yajima T; Tanimura A; Kato T; Yoshizawa K Long-Term Effect of Galantamine on Cognitive Function in Patients with Alzheimer’s Disease versus a Simulated Disease Trajectory: An Observational Study in the Clinical Setting. NDT 2017, Volume 13, 1115–1124. 10.2147/NDT.S133145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (107).Baseman A; Hager K; Nye JS; Brashear R; Han J; Sano M; Davis B; Richards H Effects of Galantamine in a 2-Year, Randomized, Placebo-Controlled Study in Alzheimer's Disease. NDT 2014, 391. 10.2147/NDT.S57909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (108).Hardy J; Allsop D Amyloid Deposition as the Central Event in the Aetiology of Alzheimer’s Disease. Trends in Pharmacological Sciences 1991, 12, 383–388. 10.1016/0165-6147(91)90609-V. [DOI] [PubMed] [Google Scholar]
- (109).Georgieva M; Vezenkov L; Ivanov, Tch.; Ivanova, C. Design and Synthesis of New γ-Secretase Inhibitors. Comptes Rendus de l’Academie Bulgare des Sciences 2004, 57, 9. [Google Scholar]
- (110).Vezenkov L; Georgieva M; Danalev D; Ivanov T; Ivanova G Synthesis and Characterization of New Galanthamine Derivatives Comprising Peptide Moiety. PPL 2009, 16 (9), 1024–1028. 10.2174/092986609789055412. [DOI] [PubMed] [Google Scholar]
- (111).Vezenkov L; Sevalle J; Danalev D; Ivanov T; Bakalova A; Georgieva M; Checler F Galantamine-Based Hybrid Molecules with Acetylcholinesterase, Butyrylcholinesterase and γ-Secretase Inhibition Activities. CAR 2012, 9 (5), 600–605. 10.2174/156720512800618044. [DOI] [PubMed] [Google Scholar]
- (112).Reid GA; Chilukuri N; Darvesh S Butyrylcholinesterase and the Cholinergic System. Neuroscience 2013, 234, 53–68. 10.1016/j.neuroscience.2012.12.054. [DOI] [PubMed] [Google Scholar]
- (113).Kitazume S; Tachida Y; Oka R; Shirotani K; Saido TC; Hashimoto Y Alzheimer’s β-Secretase, β-Site Amyloid Precursor Protein-Cleaving Enzyme, Is Responsible for Cleavage Secretion of a Golgi-Resident Sialyltransferase. Proc. Natl. Acad. Sci. U.S.A 2001, 98 (24), 13554–13559. 10.1073/pnas.241509198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (114).Ivanov I; Vezenkov L; Danalev D New Peptide Analogues of β-Secretase Inhibitor OM 99–2 Modified in P3 Position. In Collection Symposium Series; Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic: Prague, 2009; pp 47–50. 10.1135/css200911047. [DOI] [Google Scholar]
- (115).Vezenkov LT; Tsekova DS; Kostadinova I; Mihaylova R; Vassilev NG; Danchev ND Synthesis of New Galanthamine-Peptide Derivatives Designed for Prevention and Treatment of Alzheimer’s Disease. CAR 2019, 16 (3), 183–192. 10.2174/1567205016666190228123923. [DOI] [PubMed] [Google Scholar]
- (116).Hankes LV; Coenen HH; Rota E; Langen KJ; Herzog H; Wutz W; Stoecklin G; Feinendegen LE Effect of Huntington’s and Alzheimer’s Diseases on the Transport of Nicotinic Acid or Nicotinamide Across the Human Blood-Brain Barrier. In Kynurenine and Serotonin Pathways; Schwarcz R, Young SN, Brown RR, Eds.; Advances in Experimental Medicine and Biology; Springer New York: Boston, MA, 1991; Vol. 294, pp 675–678. 10.1007/978-1-4684-5952-4_91. [DOI] [PubMed] [Google Scholar]
- (117).Penberthy WT Nicotinic Acid-Mediated Activation of Both Membrane and Nuclear Receptors towards Therapeutic Glucocorticoid Mimetics for Treating Multiple Sclerosis. PPAR Research 2009, 2009, 1–11. 10.1155/2009/853707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (118).Vezenkov LT; Danalev DL; Iwanov I; Lozanov V; Atanasov A; Todorova R; Vassilev N; Karadjova V Synthesis and Biological Study of New Galanthamine-Peptide Derivatives Designed for Prevention and Treatment of Alzheimer’s Disease. Amino Acids 2022, 54 (6), 897–910. 10.1007/s00726-022-03167-z. [DOI] [PubMed] [Google Scholar]
- (119).Jarrett JT; Berger EP; Lansbury PT The Carboxy Terminus of the .Beta. Amyloid Protein Is Critical for the Seeding of Amyloid Formation: Implications for the Pathogenesis of Alzheimer’s Disease. Biochemistry 1993, 32 (18), 4693–4697. 10.1021/bi00069a001. [DOI] [PubMed] [Google Scholar]
- (120).Jan A; Gokce O; Luthi-Carter R; Lashuel HA The Ratio of Monomeric to Aggregated Forms of Aβ40 and Aβ42 Is an Important Determinant of Amyloid-β Aggregation, Fibrillogenesis, and Toxicity. Journal of Biological Chemistry 2008, 283 (42), 28176–28189. 10.1074/jbc.M803159200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (121).Olofsson A; Lindhagen-Persson M; Sauer-Eriksson AE; Öhman A Amide Solvent Protection Analysis Demonstrates That Amyloid-β(1–40) and Amyloid-β(1–42) Form Different Fibrillar Structures under Identical Conditions. Biochemical Journal 2007, 404 (1), 63–70. 10.1042/BJ20061561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (122).Olofsson A; Sauer-Eriksson AE; Öhman A The Solvent Protection of Alzheimer Amyloid-β-(1–42) Fibrils as Determined by Solution NMR Spectroscopy. Journal of Biological Chemistry 2006, 281 (1), 477–483. 10.1074/jbc.M508962200. [DOI] [PubMed] [Google Scholar]
- (123).Varadarajan S; Yatin S; Aksenova M; Butterfield DA Review: Alzheimer’s Amyloid β-Peptide-Associated Free Radical Oxidative Stress and Neurotoxicity. Journal of Structural Biology 2000, 130 (2–3), 184–208. 10.1006/jsbi.2000.4274. [DOI] [PubMed] [Google Scholar]
- (124).Smith DG; Cappai R; Barnham KJ The Redox Chemistry of the Alzheimer’s Disease Amyloid β Peptide. Biochimica et Biophysica Acta (BBA) - Biomembranes 2007, 1768 (8), 1976–1990. 10.1016/j.bbamem.2007.02.002. [DOI] [PubMed] [Google Scholar]
- (125).Feng Y; Wang X Antioxidant Therapies for Alzheimer’s Disease. Oxidative Medicine and Cellular Longevity 2012, 2012, 1–17. 10.1155/2012/472932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (126).Arai T; Ohno A; Kazunori M; Kakizawa T; Kuwata H; Ozawa T; Shibanuma M; Hara S; Ishida S; Kurihara M; Miyata N; Nakagawa H; Fukuhara K Design, Synthesis, and Evaluation of Trolox-Conjugated Amyloid-β C-Terminal Peptides for Therapeutic Intervention in an in Vitro Model of Alzheimer’s Disease. Bioorganic & Medicinal Chemistry 2016, 24 (18), 4138–4143. 10.1016/j.bmc.2016.06.057. [DOI] [PubMed] [Google Scholar]
- (127).Linning P; Haussmann U; Beyer I; Weidlich S; Schieb H; Wiltfang J; Klafki H-W; Knölker H-J Optimisation of BACE1 Inhibition of Tripartite Structures by Modification of Membrane Anchors, Spacers and Pharmacophores - Development of Potential Agents for the Treatment of Alzheimer’s Disease. Org Biomol Chem 2012, 10 (41), 8216–8235. 10.1039/c2ob26103k. [DOI] [PubMed] [Google Scholar]
- (128).Schieb H; Weidlich S; Schlechtingen G; Linning P; Jennings G; Gruner M; Wiltfang J; Klafki H-W; Knölker H-J Structural Design, Solid-Phase Synthesis and Activity of Membrane-Anchored β-Secretase Inhibitors on Aβ Generation from Wild-Type and Swedish-Mutant APP. Chem. Eur. J 2010, 16 (48), 14412–14423. 10.1002/chem.201002878. [DOI] [PubMed] [Google Scholar]
- (129).Heo J-M; Park J; Kim J Retro Diels–Alder-Triggered Self-Assembly of a Polymerizable Macrocyclic Diacetylene. Org. Biomol. Chem 2023, 21 (31), 6302–6306. 10.1039/D3OB00953J. [DOI] [PubMed] [Google Scholar]
- (130).Pardridge WM Re-Engineering Biopharmaceuticals for Delivery to Brain with Molecular Trojan Horses. Bioconjugate Chem. 2008, 19 (7), 1327–1338. 10.1021/bc800148t. [DOI] [PubMed] [Google Scholar]
- (131).Wängler C; Chowdhury S; Höfner G; Djurova P; Purisima EO; Bartenstein P; Wängler B; Fricker G; Wanner KT; Schirrmacher R Shuttle–Cargo Fusion Molecules of Transport Peptides and the hD 2/3 Receptor Antagonist Fallypride: A Feasible Approach To Preserve Ligand–Receptor Binding? J. Med. Chem. 2014, 57 (10), 4368–4381. 10.1021/jm5004123. [DOI] [PubMed] [Google Scholar]
- (132).Ó Murchú SC; Slyne AD Opioids, Plasticity and Phrenic Motor Performance: Investigating the off-Target Effects of Acute Morphine Administration. J Physiol 2021, 599 (19), 4411–4412. 10.1113/JP282068. [DOI] [PubMed] [Google Scholar]
- (133).Darcq E; Kieffer BL Opioid Receptors: Drivers to Addiction? Nat Rev Neurosci 2018, 19 (8), 499–514. 10.1038/s41583-018-0028-x. [DOI] [PubMed] [Google Scholar]
- (134).Contet C; Kieffer BL; Befort K Mu Opioid Receptor: A Gateway to Drug Addiction. Curr Opin Neurobiol 2004, 14 (3), 370–378. 10.1016/j.conb.2004.05.005. [DOI] [PubMed] [Google Scholar]
- (135).Santino F; Gentilucci L Design of κ-Opioid Receptor Agonists for the Development of Potential Treatments of Pain with Reduced Side Effects. Molecules 2023, 28 (1), 346. 10.3390/molecules28010346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (136).Muratspahić E; Deibler K; Han J; Tomašević N; Jadhav KB; Olivé-Marti A-L; Hochrainer N; Hellinger R; Koehbach J; Fay JF; Rahman MH; Hegazy L; Craven TW; Varga BR; Bhardwaj G; Appourchaux K; Majumdar S; Muttenthaler M; Hosseinzadeh P; Craik DJ; Spetea M; Che T; Baker D; Gruber CW Design and Structural Validation of Peptide-Drug Conjugate Ligands of the Kappa-Opioid Receptor. Nat Commun 2023, 14 (1), 8064. 10.1038/s41467-023-43718-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (137).Che T; Majumdar S; Zaidi SA; Ondachi P; McCorvy JD; Wang S; Mosier PD; Uprety R; Vardy E; Krumm BE; Han GW; Lee M-Y; Pardon E; Steyaert J; Huang X-P; Strachan RT; Tribo AR; Pasternak GW; Carroll FI; Stevens RC; Cherezov V; Katritch V; Wacker D; Roth BL Structure of the Nanobody-Stabilized Active State of the Kappa Opioid Receptor. Cell 2018, 172 (1–2), 55–67.e15. 10.1016/j.cell.2017.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (138).Cichewicz RH; Kouzi SA Chemistry, Biological Activity, and Chemotherapeutic Potential of Betulinic Acid for the Prevention and Treatment of Cancer and HIV Infection. Med. Res. Rev 2004, 24 (1), 90–114. 10.1002/med.10053. [DOI] [PubMed] [Google Scholar]
- (139).Tolstikova TG; Sorokina IV; Tolstikov GA; Tolstikov AG; Flekhter OB Biological Activity and Pharmacological Prospects of Lupane Terpenoids: II. Semisynthetic Lupane Derivatives. Russ J Bioorg Chem 2006, 32 (3), 261–276. 10.1134/S1068162006030083. [DOI] [PubMed] [Google Scholar]
- (140).Ghaffari Moghaddam M; Bin H. Ahmad F; Samzadeh-Kermani A Biological Activity of Betulinic Acid: A Review. PP 2012, 03 (02), 119–123. 10.4236/pp.2012.32018. [DOI] [Google Scholar]
- (141).Drag-Zalesinska M; Kulbacka J; Saczko J; Wysocka T; Zabel M; Surowiak P; Drag M Esters of Betulin and Betulinic Acid with Amino Acids Have Improved Water Solubility and Are Selectively Cytotoxic toward Cancer Cells. Bioorganic & Medicinal Chemistry Letters 2009, 19 (16), 4814–4817. 10.1016/j.bmcl.2009.06.046. [DOI] [PubMed] [Google Scholar]
- (142).Govdi AI; Sokolova NV; Sorokina IV; Baev DS; Tolstikova TG; Mamatyuk VI; Fadeev DS; Vasilevsky SF; Nenajdenko VG Synthesis of New Betulinic Acid–Peptide Conjugates and in Vivo and in Silico Studies of the Influence of Peptide Moieties on the Triterpenoid Core Activity. Med. Chem. Commun 2015, 6 (1), 230–238. 10.1039/C4MD00236A. [DOI] [Google Scholar]
- (143).Dinkova-Kostova AT; Liby KT; Stephenson KK; Holtzclaw WD; Gao X; Suh N; Williams C; Risingsong R; Honda T; Gribble GW; Sporn MB; Talalay P Extremely Potent Triterpenoid Inducers of the Phase 2 Response: Correlations of Protection against Oxidant and Inflammatory Stress. Proc. Natl. Acad. Sci. U.S.A 2005, 102 (12), 4584–4589. 10.1073/pnas.0500815102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (144).Liu H; Dinkova-Kostova AT; Talalay P Coordinate Regulation of Enzyme Markers for Inflammation and for Protection against Oxidants and Electrophiles. Proc. Natl. Acad. Sci. U.S.A 2008, 105 (41), 15926–15931. 10.1073/pnas.0808346105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (145).Vasilevsky SF; Govdi AI; Sorokina IV; Tolstikova TG; Baev DS; Tolstikov GA; Mamatuyk VI; Alabugin IV Rapid Access to New Bioconjugates of Betulonic Acid via Click Chemistry. Bioorganic & Medicinal Chemistry Letters 2011, 21 (1), 62–65. 10.1016/j.bmcl.2010.11.072. [DOI] [PubMed] [Google Scholar]
- (146).Govdi AI; Vasilevsky SF; Sokolova NV; Sorokina IV; Tolstikova TG; Nenajdenko VG Betulonic Acid–Peptide Conjugates: Synthesis and Evaluation of Anti-Inflammatory Activity. Mendeleev Communications 2013, 23 (5), 260–261. 10.1016/j.mencom.2013.09.007. [DOI] [Google Scholar]
- (147).Cross M; Smith E; Hoy D; Carmona L; Wolfe F; Vos T; Williams B; Gabriel S; Lassere M; Johns N; Buchbinder R; Woolf A; March L The Global Burden of Rheumatoid Arthritis: Estimates from the Global Burden of Disease 2010 Study. Ann Rheum Dis 2014, 73 (7), 1316–1322. 10.1136/annrheumdis-2013-204627. [DOI] [PubMed] [Google Scholar]
- (148).Homma A; Sato H; Okamachi A; Emura T; Ishizawa T; Kato T; Matsuura T; Sato S; Tamura T; Higuchi Y; Watanabe T; Kitamura H; Asanuma K; Yamazaki T; Ikemi M; Kitagawa H; Morikawa T; Ikeya H; Maeda K; Takahashi K; Nohmi K; Izutani N; Kanda M; Suzuki R Novel Hyaluronic Acid–Methotrexate Conjugates for Osteoarthritis Treatment. Bioorganic & Medicinal Chemistry 2009, 17 (13), 4647–4656. 10.1016/j.bmc.2009.04.063. [DOI] [PubMed] [Google Scholar]
- (143).Hamstra DA; Pagé M; Maybaum J; Rehemtulla A Expression of Endogenously Activated Secreted or Cell Surface Carboxypeptidase A Sensitizes Tumor Cells to Methotrexate-α-Peptide Prodrugs1. Cancer Research 2000, 60 (3), 657–665. [PubMed] [Google Scholar]
- (149).Smal MA; Dong Z; Cheung HTA; Asano Y; Escoffier L; Costello M; Tattersall MHN Activation and Cytotoxicity of 2-α-Aminoacyl Prodrugs of Methotrexate. Biochemical Pharmacology 1995, 49 (4), 567–574. 10.1016/0006-2952(94)00456-V. [DOI] [PubMed] [Google Scholar]
- (150).Homma A; Sato H; Tamura T; Okamachi A; Emura T; Ishizawa T; Kato T; Matsuura T; Sato S; Higuchi Y; Watanabe T; Kitamura H; Asanuma K; Yamazaki T; Ikemi M; Kitagawa H; Morikawa T; Ikeya H; Maeda K; Takahashi K; Nohmi K; Izutani N; Kanda M; Suzuki R Synthesis and Optimization of Hyaluronic Acid–Methotrexate Conjugates to Maximize Benefit in the Treatment of Osteoarthritis. Bioorganic & Medicinal Chemistry 2010, 18 (3), 1062–1075. 10.1016/j.bmc.2009.12.053. [DOI] [PubMed] [Google Scholar]
- (151).Majumdar S; Anderson ME; Xu CR; Yakovleva TV; Gu LC; Malefyt TR; Siahaan TJ Methotrexate (MTX)–cIBR Conjugate for Targeting MTX to Leukocytes: Conjugate Stability and In Vivo Efficacy in Suppressing Rheumatoid Arthritis. Journal of Pharmaceutical Sciences 2012, 101 (9), 3275–3291. 10.1002/jps.23164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (152).Anderson ME; Siahaan TJ Targeting ICAM-1/LFA-1 Interaction for Controlling Autoimmune Diseases: Designing Peptide and Small Molecule Inhibitors. Peptides 2003, 24 (3), 487–501. 10.1016/S0196-9781(03)00083-4. [DOI] [PubMed] [Google Scholar]
- (153).Hersmann GHW; Kriegsmann J; Simon J; Hüttich C; Bräuer R Expression of Cell Adhesion Molecules and Cytokines in Murine Antigen-Induced Arthritis. Cell Adhesion and Communication 1998, 6 (1), 69–82. 10.3109/15419069809069761. [DOI] [PubMed] [Google Scholar]
- (154).Bi W; Bi Y; Xue P; Zhang Y; Gao X; Wang Z; Li M; Baudy-Floc’h M; Ngerebara N; Michael Gibson K; Bi L A New Class of β-Carboline Alkaloid-Peptide Conjugates with Therapeutic Efficacy in Acute Limb Ischemia/Reperfusion Injury. European Journal of Medicinal Chemistry 2011, 46 (5), 1453–1462. 10.1016/j.ejmech.2011.01.021. [DOI] [PubMed] [Google Scholar]
- (155).Bi W; Bi Y; Xue P; Zhang Y; Gao X; Wang Z; Li M; Baudy-Floc’h M; Ngerebara N; Li X; Gibson KM; Bi L Novel β-Carboline-Tripeptide Conjugates Attenuate Mesenteric Ischemia/Reperfusion Injury in the Rat. European Journal of Medicinal Chemistry 2011, 46 (6), 2441–2452. 10.1016/j.ejmech.2011.03.029. [DOI] [PubMed] [Google Scholar]
- (156).Sun M-S; Jin H; Sun X; Huang S; Zhang F-L; Guo Z-N; Yang Y Free Radical Damage in Ischemia-Reperfusion Injury: An Obstacle in Acute Ischemic Stroke after Revascularization Therapy. Oxidative Medicine and Cellular Longevity 2018, 2018, 1–17. 10.1155/2018/3804979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (157).Wu L; Xiong X; Wu X; Ye Y; Jian Z; Zhi Z; Gu L Targeting Oxidative Stress and Inflammation to Prevent Ischemia-Reperfusion Injury. Front. Mol. Neurosci 2020, 13, 28. 10.3389/fnmol.2020.00028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (158).Susanna Y.H T; I-Tong M; Dickens BF Antioxidative Properties of Harmane and β-Carboline Alkaloids. Biochemical Pharmacology 1991, 42 (3), 459–464. 10.1016/0006-2952(91)90305-O. [DOI] [PubMed] [Google Scholar]
- (159).Moura DJ; Richter MF; Boeira JM; Pegas Henriques JA; Saffi J Antioxidant Properties of -Carboline Alkaloids Are Related to Their Antimutagenic and Antigenotoxic Activities. Mutagenesis 2007, 22 (4), 293–302. 10.1093/mutage/gem016. [DOI] [PubMed] [Google Scholar]
- (160).Herraiz T; Galisteo J Tetrahydro-β-Carboline Alkaloids Occur in Fruits and Fruit Juices. Activity as Antioxidants and Radical Scavengers. J. Agric. Food Chem 2003, 51 (24), 7156–7161. 10.1021/jf030324h. [DOI] [PubMed] [Google Scholar]
- (161).Herraiz T; Galisteo J Tetrahydro-β-Carboline Alkaloids That Occur in Foods and Biological Systems Act as Radical Scavengers and Antioxidants in the ABTS Assay. Free Radical Research 2002, 36 (8), 923–928. 10.1080/1071576021000005762. [DOI] [PubMed] [Google Scholar]
- (162).Bi W; Bi Y; Xue P; Zhang Y; Gao X; Wang Z; Li M; Baudy-Floc’h M; Ngerebara N; Michael Gibson K; Bi L A New Class of β-Carboline Alkaloid-Peptide Conjugates with Therapeutic Efficacy in Acute Limb Ischemia/Reperfusion Injury. European Journal of Medicinal Chemistry 2011, 46 (5), 1453–1462. 10.1016/j.ejmech.2011.01.021. [DOI] [PubMed] [Google Scholar]
