Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect
ACS Publications. Most Trusted. Most Cited. Most Read
Structure–Activity Relationship of Antibody–Oligonucleotide Conjugates: Evaluating Bioconjugation Strategies for Antibody–siRNA Conjugates for Drug Development
My Activity
CONTENT TYPES

Figure 1Loading Img
  • Open Access
Article

Structure–Activity Relationship of Antibody–Oligonucleotide Conjugates: Evaluating Bioconjugation Strategies for Antibody–siRNA Conjugates for Drug Development
Click to copy article linkArticle link copied!

  • Michael Cochran
    Michael Cochran
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
  • Danny Arias
    Danny Arias
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    More by Danny Arias
  • Rob Burke
    Rob Burke
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    More by Rob Burke
  • David Chu
    David Chu
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    More by David Chu
  • Gulin Erdogan
    Gulin Erdogan
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
  • Michael Hood
    Michael Hood
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    More by Michael Hood
  • Philip Kovach
    Philip Kovach
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
  • Hae Won Kwon
    Hae Won Kwon
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    More by Hae Won Kwon
  • Yanling Chen
    Yanling Chen
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    More by Yanling Chen
  • Michael Moon
    Michael Moon
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    More by Michael Moon
  • Christopher D. Miller
    Christopher D. Miller
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
  • Hanhua Huang
    Hanhua Huang
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    More by Hanhua Huang
  • Arthur Levin
    Arthur Levin
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    More by Arthur Levin
  • Venkata Ramana Doppalapudi*
    Venkata Ramana Doppalapudi
    Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    *Email: [email protected]
Open PDFSupporting Information (1)

Journal of Medicinal Chemistry

Cite this: J. Med. Chem. 2024, 67, 17, 14852–14867
Click to copy citationCitation copied!
https://doi.org/10.1021/acs.jmedchem.4c00802
Published August 28, 2024

Copyright © 2024 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY-NC-ND 4.0 .

Abstract

Click to copy section linkSection link copied!

Antibody–oligonucleotide conjugates are a promising class of therapeutics for extrahepatic delivery of small interfering ribonucleic acids (siRNAs). These conjugates can be optimized for improved delivery and mRNA knockdown (KD) through understanding of structure–activity relationships. In this study, we systematically examined factors including antibody isotype, siRNA chemistry, linkers, conjugation chemistry, PEGylation, and drug-to-antibody ratios (DARs) for their impact on bioconjugation, pharmacokinetics (PK), siRNA delivery, and bioactivity. Conjugation site (cysteine, lysine, and Asn297 glycan) and DAR proved critical for optimal conjugate PK and siRNA delivery. SiRNA chemistry including 2′ sugar modifications and positioning of phosphorothioates were found to be critical for delivery and duration of action. By utilizing cleavable and noncleavable linkers, we demonstrated the impact of linkers on PK and mRNA KD. To achieve optimal properties of antibody–siRNA conjugates, a careful selection of siRNA chemistry, DAR, conjugation sites, linkers, and antibody isotype is necessary.

This publication is licensed under

CC-BY-NC-ND 4.0 .
  • cc licence
  • by licence
  • nc licence
  • nd licence
Copyright © 2024 The Authors. Published by American Chemical Society

Introduction

Click to copy section linkSection link copied!

Antibodies and oligonucleotides, two powerful classes of molecules, have diverse applications in biology and medicine. As highly specific proteins, antibodies recognize and bind to a wide range of targets. Their proven effectiveness as receptor antagonists and as delivery vehicles for small molecules makes them potent therapeutics. (1−4)
Oligonucleotides exert their therapeutic effect through multiple mechanisms, including inducing the degradation of mRNA or promoting splice modulation. (5−9) Over the last 25 years, more than 16 oligonucleotide therapeutics have been approved for multiple therapeutic indications, including homozygous familial hypercholesterolemia, spinal muscular atrophy, Duchenne muscular dystrophy, hereditary transthyretin-mediated amyloidosis, familial chylomicronemia syndrome, acute hepatic porphyria, and primary hyperoxaluria. (10)
Small interfering ribonucleic acids (siRNAs) are 20–25 nucleotide-long, double-stranded RNA molecules containing a complementary sense and antisense strand. When inside the cell, these duplexes load into the Ago2 protein. Following this, the sense strand of the duplex siRNA is cleaved, and its fragments ejected, leaving behind a functional RNA-induced silencing complex (RISC) that comprises the antisense strand and Ago2. The RISC then uses the antisense strand to recognize and bind to complementary mRNA sequences within the cell. Once fully hybridized with the mRNA, Ago2 cleaves the mRNA between target bases 10 and 11, resulting in mRNA degradation and reduced gene expression. siRNAs are proven therapeutic agents, especially in diseases caused by the overexpression of certain genes or the presence of harmful genetic mutations. By selectively targeting specific disease-related RNA, siRNA-based therapies hold promise for treating a wide range of conditions, including certain types of cancer, viral infections, and genetic disorders. (5−9)
siRNA therapeutics face three main challenges: selectivity, stability, and delivery. Selectivity is primarily tackled through bioinformatics or chemical modifications, which can help predict the efficacy of candidate oligonucleotides and ensure they are designed to minimize off-target effects. (11) Stability is addressed by chemical modifications, such as phosphorothioate linkages or 2′-O-methyl modifications, which protect the oligonucleotides from degradation by nucleases in the bloodstream and other tissues. Delivery is an issue because different tissue types can present unique barriers, requiring specific targeted delivery strategies dependent on the target tissue. siRNA delivery to the liver is achieved with lipid nanoparticle formulations and conjugation of ligands such as N-acetylgalactosamine (GalNAc), which has a high affinity for asialoglycoprotein receptors (ASGPRs) that are predominantly expressed on hepatic parenchymal cells. Also, intrathecal delivery of siRNA modified with C16, or 2′-O-hexadecyl has produced potent and durable knockdown (KD) in the central nervous system (CNS). (12) However, there remains a significant unmet need for technologies that can effectively deliver oligonucleotides systemically to other tissues. (13) Owing to their inherent specificity for cell surface receptors and their demonstrated capability to transport small molecules to a variety of tissues, antibodies have emerged as promising vehicles for oligonucleotide delivery. Antibody–oligonucleotide conjugates (AOCs) present a promising solution for overcoming the challenges associated with oligonucleotide delivery.
Previous research by different groups has explored the use of antibody-mediated targeted delivery of oligonucleotides, yielding varying degrees of success. Seminal work by Cuellar et al. (2015) showed that siRNA oligonucleotides could be conjugated to thiol-containing monoclonal antibodies (mAbs), resulting in active, stable conjugates in vitro. (14) They showed that the mAb-siRNA conjugates retained a similar binding affinity and gene-silencing capacity to that of their constituent mAb or siRNA. However, when tested in mouse tumor xenograft studies, their activity was modest. Sela et al. (2023) recently reported on a new method for delivering antisense-oligonucleotides (ASOs) to the CNS for treating neurological disorders. (15) They conjugated the ASO to an antibody that could cross the blood–brain barrier. The study found promising in vitro activity and in vivo pharmacokinetics (PK) behavior, suggesting potential for future ASO-based CNS drugs. Of particular interest is the targeted delivery of various oligonucleotides to muscle for the treatment of muscle disorders via a transferrin receptor 1 (TfR1)-mediated approach. (16) Recent publications, such as the work by Malecova et al. (2023), have demonstrated effective delivery of diverse oligonucleotides to muscle tissue. (17)
While other studies have demonstrated delivery of oligonucleotides conjugated to mAbs and antigen-binding fragments, there is currently a lack of information on comprehensive SARs of antibody–siRNA conjugates. (16,18−20) This publication will provide a comprehensive SAR of antibody–siRNA conjugates, including optimal linker selection, points of conjugation on the antibody and siRNA, bioconjugation strategies to link an antibody and siRNA oligonucleotide, and the impact of siRNA oligonucleotide-to-antibody ratios on activity. The AOC approach offers a flexible strategy to increase the potential delivery of oligonucleotides to target tissues.

AOC Naming Convention

Click to copy section linkSection link copied!

In the rapidly evolving field of AOCs, the need for a clear, standardized naming convention is paramount because a multitude of variables exist, such as the type of antibody, its target, the linker used, and the siRNA conjugated. Our naming convention, detailed below, offers a systematic approach using specific symbols and abbreviations to denote each component of an AOC.
Take the following conjugates as examples: αhEGFR-Cys-MCC-siKRAS, αmTfR1-Cys-MCC-siMstn, αmTfR1-Cys-PDP-siMstn (for disulfide linkers). Here, “α” means “anti” to indicate that the antibody binds to EGFR (epidermal growth factor receptor) or TfR1. “h” or “m” denote whether the antibody is human (h) or mouse (m) specific. “Cys” or “Lys” represent the amino acids cysteine (Cys) or lysine (Lys) that the linker is attached to on the antibody. “MCC” or “PDP” are the linkers, 4-(N-maleimidomethyl)cyclohexane-1-carboxyamide and 3-(2-Pyridyldithio)propionamide. The “siKRAS” or “siMstn” are the siRNAs, targeting KRAS mRNA and Mstn mRNA, respectively, that are conjugated to the antibody. For example, “αhEGFR-Cys-MCC-siKRAS” would be an anti-EGFR antibody that is specific to humans, with an MCC linker attached to a cysteine residue, and conjugated to an siRNA targeting KRAS.
In our naming convention for linkers, we use the linker name, rather than the reagent used in the reaction. This is because during the reaction, a part of the reagent is removed. For instance, instead of using “SMCC” (the reagent) as the name of the linker, we simply use “MCC”. This approach ensures that the names we use are representative of the actual structure of the AOCs. However, in the Experimental Section, we use the full reagent name when discussing the linker reaction with the siRNA; for example, SMCC (reagent) versus MCC (linker).

Results and Discussion

Click to copy section linkSection link copied!

We first describe a general overview of the AOC synthesis used to generate antibody–siRNA conjugates. AOCs can be generated using any antibody isotype and siRNA with a conjugation handle, using cysteine, lysine or Asn297 conjugation methods and their corresponding linkers. The conditions and yields may vary, based on the constructs being used, but the general scheme remains the same.
We then detail the results of bioconjugation reaction and purification steps that reduce the risk of product aggregation and yield a highly pure AOC product. We then describe our investigations into antibody–siRNA conjugate SARs. We evaluated siRNA chemical modifications, siRNA conjugation locations on the mAb, cleavable versus noncleavable linkers, linker location on the siRNA, the addition of polyethylene glycol (PEG), and antibody isotypes.
Table S1 describes the antibody target, linker, siRNA sequence and chemical modification pattern for each antibody–siRNA conjugate. Furthermore, Table S2 provides a comprehensive list of the siRNA conjugation components, detailing the siRNA conjugation handles, linker reagent structures, linker-siRNA structures, antibody reactive groups, and final antibody–siRNA conjugate structures.

AOC Synthesis

General Synthetic Scheme for Antibody–siRNA Conjugate Generation

Scheme 1 outlines the general synthetic scheme we use to generate antibody–siRNA conjugates. For cysteine-conjugated AOCs, we first couple a linker-maleimide to the amino group at the 5′ end of the sense strand of a duplex siRNA using N-hydroxysuccinimide (NHS) chemistry. (21,22) This generates an intermediate siRNA-linker-maleimide (Scheme 1a).

Scheme 1

Scheme 1. (a) Synthetic Scheme for Conjugating an siRNA with a Linker; (b) Antibody–siRNA Conjugate Synthesis via Interchain Disulfide (Cysteine) Conjugation; (c) Antibody–siRNA Conjugate Synthesis via Asn297 Conjugation; (d) Antibody–siRNA Conjugate Synthesis via Lysine Conjugation
We then partially reduce the interchain disulfide of the antibody with tris(2-carboxyethyl)phosphine (TCEP) (Scheme 1b). This means the disulfide bonds between the cysteine residues in the antibody are broken, exposing the cysteine residue thiol groups, which can then be used for conjugation with the oligonucleotide. The antibody is then conjugated to the siRNA-linker-maleimide. The resulting intermediate conjugate is processed through anion exchange chromatography, dehydroascorbic acid (DHAA) oxidation, N-ethylmaleimide (NEM) capping, and buffer exchange. This results in an antibody–siRNA conjugate with a drug-to-antibody ratio of 1 (DAR1). Scheme 1b depicts random cysteine conjugation. In this paper, we will also discuss site-specific cysteine conjugation.
In addition to the cysteine conjugation shown in Scheme 1b, AOCs can be synthesized via Asn297 (Scheme 1c) and lysine (Scheme 1d) conjugations. (23−25) For synthesis via Asn297, we modify the N-linked glycans at Asn297 enzymatically to azide-activate the antibody on each heavy chain. A bifunctional dibenzocyclooctyne (DBCO)-PEG-trans-cyclooctene (TCO) linker is added to the azide conjugation handle on the antibody to generate an antibody with a TCO conjugation handle at the Asn297 location. In a separate reaction, methyltetrazine-siRNA is generated by conjugating methyltetrazine NHS ester to the siRNA amine conjugation handle. A TCO-methyltetrazine click chemistry is performed between the functionalized antibody and siRNA to generate an AOC with a DAR of 1. For synthesis via lysine, the antibody is activated using the noncleavable linker succinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC); the NHS ester of SMCC reacts with the amino groups of the lysine residues attaching reactive maleimide groups to the lysine side chain. The maleimide-activated antibody is then reacted with the thiol-containing siRNA and the resulting intermediate conjugate is then processed through anion exchange chromatography and buffer exchange, resulting in a DAR1 antibody–siRNA conjugate.
Figure 1 shows a typical structure of an siRNA molecule conjugated with a 4-(N-maleimidomethyl)cyclohexane-1-carboxyamide (MCC) linker through a C6 amine linker. This paper describes AOCs generated with siRNAs conjugated at the 5′ and 3′ termini of the sense strand. There are potential advantages of each of these strategies, including using the steric bulk of the conjugation to protect against 3′ nucleases as well as preventing unwanted loading of the sense strand into RISC by conjugating to the 5′ end. (26)

Figure 1

Figure 1. Typical structure of an siRNA molecule. MCC linker: a heterobifunctional cross-linker that contains two reactive groups: an NHS ester and a maleimide. The NHS ester reacts with primary amines, while the maleimide reacts with sulfhydryl groups. This allows the MCC to form stable bonds with both amine- and sulfhydryl-containing molecules. C6 amino: a linker between the MCC and the siRNA. It provides a sufficient distance between the siRNA and the MCC to minimize any potential steric hindrance or interference with the siRNA’s function. Sense strand 5′ end: the 5′ end of the sense strand of the siRNA. The 5′ end is where the MCC is attached via the C6 amine linker. Sense strand 3′ end: the 3′ end of the sense strand of the siRNA. Antisense strand: designed to be complementary to the target mRNA sequence. It guides the RISC to the target mRNA. Phosphodiester and phosphorothioate bonds: the bonds that connect the nucleotides in the siRNA. Phosphodiester bonds are the standard bonds in RNA, while phosphorothioate bonds are modified versions that contain a sulfur atom, which can provide increased stability to the siRNA.

Throughout this paper, AOC dose levels are based on the siRNA component of the AOC. Calculations to convert siRNA to mAb or AOC concentrations are provided in the Experimental Section. A 1 mg/kg siRNA dose is equivalent to a 10.6 mg/kg mAb dose, and an 11.6 mg/kg DAR1 AOC dose. While the molecular weight of the siRNA and AOC may change with sequence, modifications, and linkers, the impact of these changes is less than 5% of the total MW and does not significantly impact these ratios.

Optimization of Disulfide Bond Reduction and Capping in Cys Conjugation Method

The cysteine DAR 1 AOCs generated by partially reducing the interchain disulfide bonds of an antibody have 1–7 unreacted thiols depending upon the number of reduced disulfides in an antibody molecule. These free cysteines could lead to aggregates, disulfide scrambling, and impact the conjugate stability due to a fewer number of disulfide bonds holding the conjugate together. To preemptively solve these potential challenges, we used oxidation with DHAA to reform the reduced excess disulfide bonds, then a capping of the remaining one cysteine of the isolated DAR1 AOC using NEM alkylation to form a thioether.
We evaluated the number of free cysteines on a DAR1 conjugate (Figure 2). After TCEP reduction, siRNA conjugation, and purification using strong anion exchange chromatography (SAX), DAR1 αhEGFR-siRNA AOCs (αhEGFR-Cys-MCC-siAR) without DHAA oxidation or NEM capping had more than one free cysteine available, indicating that some DAR1 conjugate molecules contain at least one other reduced disulfide bond. This isolated DAR1 AOC was split into three treatment groups: no treatment (DAR1), five equivalents of NEM to cap unreacted cysteine (DAR1 + NEM), and 10 equiv of oxidizing DHAA (DAR1 + DHAA). NEM capping blocked nearly all free cysteines (0.13 free cysteines per antibody) of DAR1 AOC, while DHAA oxidation left one free cysteine available for potential conjugation (0.89 free cysteines per antibody). AOCs with a DAR of 1 were at least 95% pure by strong anion exchange chromatography and size exclusion chromatography (SEC). The results suggest that after oxidation with DHAA, the αhEGFR hIgG1 DAR1 AOC used in this experiment will have approximately one free cysteine available to conjugate other functional groups (such as peptides or fluorescent dyes) or that free cysteine must be capped by alkylation to prevent AOC dimer formation in storage conditions (data not shown). These results are specific to the antibody used in this experiment. It is expected that each antibody will have its own degree of reduction after TCEP treatment. However, the presence of free cysteines and the requirement for alkylation to reduce dimer formation have been observed for DAR1 AOCs made with two other hIgG1 antibodies (data not shown).

Figure 2

Figure 2. Evaluation of free cysteines available for conjugation on an αhEGFR-Cys-MCC-siAR DAR1 AOC following treatments with TCEP, NEM, and DHAA (n = 3).

siRNA Chemical Modifications

The susceptibility of the siRNA internucleotide phosphodiester bond to degradation by endonucleases or exonucleases upon systemic administration hinders the accumulation of intact therapeutic siRNA in target tissues. It is important for siRNAs to be stable in circulation as well as in tissues to show not only initial target mRNA KD, but also long duration of action after deposition in the target tissues. Chemically modifying siRNAs, such as replacement of 2′–OH with a 2′-deoxy-2′-fluoro (2′-F), 2′-O-methyl (2′-OMe), or 2′-O-methoxyethyl (2′-MOE) group, as well as substitutions of certain internucleotide phosphates with phosphorothioates (P═S) is a widely used strategy to improve siRNA stability in circulation as well as intracellular environments. (27,28) One commonly used modification pattern includes two P═S at each terminus of the antisense strand, two P═S at the 5′ end of the sense strand, and either one or no P═S on the 3′ end of the sense strand, which is frequently used for GalNAc conjugates with a 3′-pyrrolidine linker. (10,29) We tested the same pattern with a C6 amine linker as an AOC and found it to be either less active or to have low durability with AOCs in vivo, as shown in Figure 3. We presume this is due to nuclease-mediated cleavage of siRNA from the unprotected end of the sense strand in mice.

Figure 3

Figure 3. Impact of siRNA chemical modifications on siRNA stability and activity in targeting Mstn through evaluation of tissue siRNA concentration (TC) and percent mRNA remaining relative to phosphate buffered saline (PBS) in gastrocnemius muscle of mice treated with αmTfR1-Cys-MCC-siMstn AOCs. aAll sense strands were duplexed with the antisense strand: vpUusUfsAoUoUoAfUoUoUoGoUoUoCoUfUoUfGoCoCosUosUo. Abbreviations: X refers to bases evaluated (AUGC): Xo = 2’O-methyl, Xf = 2’Fluoro, Xb = LNA, Xu = UNA-2’MOE, s = phosphorothioate, vp = vinylphosphonate.

To improve the stability and activity of siRNAs on AOCs in muscle tissue, we explored combinations of sugar modifications including locked nucleic acid (LNA) and unlocked nucleic acid (UNA), which are known to improve siRNA stability in biological matrices, and phosphorothioate patterns. We used AOCs consisting of an antimouse TfR1 mAb (αmTfR1) conjugated to siRNAs targeting the myostatin (Mstn) gene with the MCC linker (αmTfR1-Cys-MCC-siMstn). The modification pattern of each sense strand is shown in Figure 3. Each sense strand was duplexed with the same antisense strand (vpUusUfsAoUoUoAfUoUoUoGoUoUoCoUfUoUfGoCoCosUosUo) containing an unlocked 2′-O-methoxymethyl uridine base with a vinylphosphonate at the 5′ end (Scheme S1). A 5′-phosphate is required for loading into the mid-domain of the Argonaut2 protein; however, natural phosphorylation of the siRNA by Clp1 kinase can be impacted by chemical modifications to the siRNA. 5′-vinylphosphonates, metabolically stable 5′-phosphate mimics, which have been shown to improve tissue accumulation and efficacy of conjugated siRNAs in vivo, (30,31) were included in this experiment to reduce the potential impact of new modifications on activity. The unlocked sugar with 2′-O-methoxyethyl is believed to provide flexibility to load into the mid-domain of Argonaut2 along with steric bulk to protect against nucleases.
We administered 1 mg/kg (siRNA) doses to mice at Day 0, then sacrificed four animals from each treatment at each time point: 24 h, and 2, 4, 6, and 8 weeks. We measured Mstn mRNA expression and siRNA concentrations in the gastrocnemius muscle at each time point. Among the sugar modifications, UNA was found to be more stable than 2′-O-methyl, while LNA was more stable than UNA. This trend is true for siRNAs with no P═S on the termini of the sense strand (groups 1, 2, 4, and 6), as well as siRNAs that have one P═S at each end of the sense strand (groups 3, 5, and 7). The 5′ end of the sense strand may be the most vulnerable to degradation in the absence of P═S groups, which can be seen by the reduced KD detected at Weeks 2 and 4. In the absence of P═S, the addition of 2 UNA or LNA (groups 4 and 6) at each termini of the sense strand improved KD at Weeks 2 and 4 compared with the 2′-O-methyl (group 2); however, by Weeks 6 and 8 it is clear that UNA and LNA in the absence of P═S are not sufficient due to the loss of KD. With all three sugar modifications (2′-O-methyl, UNA, and LNA) adding a single P═S to each side of the sense strand improves activity from Weeks 4 through 8, indicating that these more stable modifications can lead to longer durations of silencing. The two most active and stable modification patterns were found to be two P═S at each terminus of the sense and antisense strands as well as the pattern with two LNAs and one P═S at each terminus of the sense and two P═S at each terminus of the antisense strand; both maintain better than 80% KD over 8 weeks while all other patterns begin to show reduced KD by Week 8 (Figure 3). As demonstrated by Brown et al. (2020), stable siRNAs can remain in acidic compartments of the cell and act as a depot that results in continuous RISC loading beyond 2 weeks and sustained KD, despite decreasing siRNA concentrations in the tissue. (32) Although the data are shown only for one antibody and siRNA conjugate, several other AOCs with different siRNA sequences and antibodies showed similar stability patterns in vivo. These findings highlight the importance of carefully designing and selecting chemical modification patterns to optimize siRNA stability and activity for therapeutic applications.

siRNA Conjugation to mAb Via Cysteine, Lysine, and Sugar Locations

Once we identified the optimal siRNA chemical modification pattern for in vivo delivery and long duration of action, we next evaluated the differences in various conjugation positions on the antibody, including cysteine, lysine, and sugar locations. We assessed differences in plasma PK in mice for antibody–siRNA conjugates comprised of an αhEGFR mAb, which does not cross-react with mouse EGFR, conjugated to an siRNA targeting KRAS (siKRAS) at different sites on the antibody. The conjugation methods included random cysteine (TCEP reduction of disulfide followed by conjugation with siRNA-maleimide), random lysine (by nonselective addition of a TCO linker to lysine side chains), and glycan conjugation at Asn297 (sugar functionalization with TCO linker) (Figure 4a). AOCs were evaluated at a dose level of 0.5 mg/kg (siRNA). The full siRNA sequences for test articles evaluated in this experiment and throughout the manuscript are listed in Table S1.

Figure 4

Figure 4. (a) Plasma PK analysis of αhEGFR-siKRAS AOCs conjugated to the mAb cysteine, lysine, and Asn297 as measured by percent of injected dose (% ID) versus time (h). (b) Mouse plasma PK of DAR1 αhTfR1-siMstn AOCs generated via random cysteine versus cysteine engineered at position 188 of the light chain.

Lysine and glycan conjugation of siRNA exhibited increased plasma clearance [46 and 11% lower area under the curve (AUC)], respectively, compared to cysteine conjugation (Figure 4a; SEM n = 4). The reduced plasma exposure of the lysine-conjugated siRNA is likely attributed to the greater heterogeneity and lower in vivo stability of lysine conjugates. While glycan conjugation is well tolerated for many antibody–drug conjugates (ADCs), it is not clear if the small reduction in plasma exposure of the glycan-conjugated AOC is the result of linker stability or the location of the siRNA on the antibody.
We also investigated the impact of random cysteine conjugation versus site-specific cysteine engineering on plasma clearance, since some locations of cysteine on the antibody show improved plasma PK of ADCs. (33) For random cysteine conjugation, we used αhTfR1-Cys-MCC-siMstn, while for site-specific cysteine engineering we used αhTfR1-K188C-MCC-siMstn, where the cysteine was engineered at position lysine 188 of the antibody light chain. No significant differences in plasma clearance were observed when conjugating siRNA to a random cysteine (Figure 4b).
While these data indicate cysteine conjugates may have advantages over glycan and lysine conjugates, additional studies are required to elucidate differences between PK, tissue delivery, and efficacy. Based on our data, additional results described herein will focus on random cysteine conjugation. Random cysteine conjugation was selected based on the relative ease of conjugate synthesis from most antibodies without the necessity of molecular engineering, less heterogeneity, clinical evaluation as shown by numerous ADCs, and manufacturability.

Drug to Antibody Ratio

We investigated the impact of DAR on AOC plasma clearance by assessing AOCs comprised of αhEGFR mAb conjugated to one, two, or three siRNAs targeting DMPK (αhEGFR-Cys-MCC-siDMPK). An antibody that does not bind to a target in mouse was selected to show the impact of DAR on plasma clearance in the absence of target-mediated drug deposition. We observed clear trends of faster plasma clearance as a function of increasing DAR (Figure 5a). Figure 5b shows the clearance rate of DAR2 is nearly five times greater than the clearance rate of DAR1 (0.101 vs 0.019 mL/h) calculated from 0 to 96 h. To determine if increasing DAR impacts delivery to tissue or activity in muscle, αmTfR1-Cys-MCC-siMstn conjugates with a DAR of 1 and 2 were tested in mice. The rapid clearance from plasma observed with DAR2 is likely caused by greater nonproductive uptake by the liver. This rapid uptake by the liver leads to lower delivery and activity in the muscle (Figure S1). AOCs with a higher DAR may have altered physicochemical properties, such as increased size and significantly greater negative charge density, which can affect their interactions with plasma proteins and clearance mechanisms. Examples of the increased size and charge density of DAR2 conjugates include a left shift on SEC (Figure S2) and a right shift on strong anion exchange (Figure S9a). Protein interaction data are not available for these conjugates but binding of oligonucleotides has been observed by others. (34,35)

Figure 5

Figure 5. (a) Mouse plasma PK of AOCs (αhEGFR-Cys-MCC-siDMPK) comprised of αhEGFR mAb conjugated to one, two, or three siRNAs targeting DMPK mRNA. (b) Noncompartmental analysis of plasma PK data (0–96 h).

Oligonucleotides themselves have been observed to interact strongly with plasma and cell surface proteins and it is possible that increasing the DAR may increase the avidity of these interactions. (36−38) A similar phenomenon of rapid DAR2 clearance with ASO-antibody conjugates (ASOs are single strands and typically fully phosphorothioated oligonucleotides) has been observed by us (unpublished) and others. (15) However, phosphorodiamidate morpholino oligonucleotide (PMO)-antibody conjugates (charge-neutral oligonucleotides) with up to DAR9.7 have no impact on plasma PK, (39) suggesting the charge and/or phosphorothioate content in siRNA and ASO DAR2 AOCs is driving clearance. Based on these data, we selected DAR1 for further evaluation throughout this paper.

Cysteine Conjugates with Cleavable Versus Non-cleavable Linkers

After selecting random cysteine conjugation as the conjugation strategy for antibody–siRNA conjugates, we focused on evaluating the impact of linker on plasma clearance. We evaluated AOCs using various cleavable and noncleavable linkers to assess their impact on in vivo stability as measured by plasma PK. To attain ideal pharmacokinetic profiles for active siRNA and desired effectiveness, it is necessary to regulate both the stability of antibody-conjugates and the release of siRNA. Favorable systemic stability is sought to safeguard linker-siRNA from hydrolysis while circulating. Simultaneously, efficient cleavage of linkers and release of siRNAs are crucial for accumulating a satisfactory quantity of active siRNA and meeting efficacy end points. Understanding these nuances is crucial for optimizing the design and functionality of antibody–siRNA conjugates. Table S2 summarizes the structures of the linkers. We assessed cleavable linkers including reducible disulfide (SS) linkers SS(methyl) and SS(gem-dimethyl) which exhibit increasing steric hindrance around the disulfide bond. Additionally, we evaluated a cathepsin-cleavable valine-citrulline (VC) linker, 3-(2-pyridyldithio)propionate (PDP) and methyl-(2-pyridyldithio)toluene (MPT). Noncleavable linkers included MCC, m-maleimidobenzoyl (MB), 4-((4-(cyanoethynyl)benzoyl)oxy) (CB), and bismaleimide (BisMal).
The impact of these linkers on AOC plasma PK was tested on conjugates with a 5000 Da MW PEG (PEG5k) attached to the sense strand at the end opposite of antibody conjugation. The benefits of PEGylation for AOC stability are included in a subsequent section. We conducted mouse PK studies with AOCs comprised of an αhEGFR antibody conjugated to siEGFR-PEG5k (PEG5k conjugated to the 3′ end of siEGFR by a stable thioether). We included a stable MCC linker between the mAb and siRNA (αhEGFR-Cys-MCC-siEGFR-PEG5k) and compared it with an enzyme-cleavable VC linker (αhEGFR-Cys-VC-siEGFR-PEG5k). Further iterations of these AOCs were synthesized to evaluate the impact of a cleavable SS linker between the siRNA and PEG5k (αhEGFR-Cys-siEGFR-SS-PEG5k and αhEGFR-VC-siEGFR-SS-PEG5k). We measured the plasma PK of these AOCs over time (Figure 6a,b) and observed no significant differences in clearance of the siRNA. However, our stem-loop assay for the detection of the antisense strand is insensitive to the presence of a PEG group attached to the sense strand, meaning the stability of the PEG5k group attached via a disulfide bond remains unclear. Furthermore, it is possible that a large PEG at the end of the siRNA could influence linker stability, for instance by limiting enzyme accessibility to the VC linker. However, considering the radius of gyration for PEG5k (∼3 nm) and the length of a 21-nucleotide siRNA duplex (∼7.5 nm), it is unlikely that PEG attached to the 3′ end of the sense strand would have a significant impact on the stability of the linker at the 5′ end. Further studies are needed to fully elucidate the impact of these modifications on the PK of AOCs.

Figure 6

Figure 6. (a,b) Mouse PK of AOCs containing cleavable linkers.

Further mouse PK studies were conducted with AOCs comprised of αhEGFR mAb conjugated to siKRAS via different cleavable disulfide linkers (PDP, MPT, SS[methyl], and SS[gem-dimethyl]) between the mAb and siRNA. (40) These AOCs each contained a PEG5k conjugated directly to the siKRAS. We dosed the AOCs at 0.5 mg/kg (siRNA) (Figure 6b). These results demonstrate that the linker stability matters for AOCs, and that increasing steric bulk around the disulfide (SS) linker is required to maintain plasma stability of these cleavable linkers. Incorporating the gem-dimethyl group into the disulfide linker reduced the clearance by more than 40 times (0.951 to 0.02 mL/h for PDP vs SS[gem-dimethyl] as shown in Table S3), suggesting that increased bulk slows down disulfide cleavage in circulation. Similar results have been observed with ADCs demonstrating the impact of disulfide linker length and steric hindrance on PK and efficacy. (41,42) The rest of this manuscript will focus on noncleavable (stable) linkers other than the data shown in Figure 9.

Further Exploration of Non-cleavable Linkers (MCC, CB, MB, and BisMal)

αhEGFR-siKRAS AOC In Vitro Stability

To compare the in vitro stability of different noncleavable linkers, we conjugated a human immunoglobulin (Ig) G1 antibody targeting EGFR (αhEGFR) to KRAS siRNA using four different noncleavable linkers (MCC, CB, MB, and BisMal). We tested the linker stability by observing siRNA release from the antibody after incubating it at 37 °C in PBS with 0.5 mM glutathione (Figure S3). Free antibody was measured by using strong-anion exchange high-performance liquid chromatography to indicate the loss of KRAS siRNA. Our results showed that αhEGFR-siKRAS AOCs containing CB, MB, and BisMal linkers were more stable than the MCC AOC in vitro, matching expectations from the literature. (43)

Linker Impact on mTfR1 Antibody-Based siRNA Delivery to Muscles and Other Tissues

We synthesized and evaluated αmTfR1 antibody–siHprt conjugates via four different linkers (MCC, CB, MB, and BisMal) to understand the impact of in vitro linker stability differences on the AOC plasma PK, siRNA tissue delivery and target mRNA KD efficacy in a mouse PK and Hprt mRNA KD study. Despite differences in in vitro stability, we observed nearly identical plasma clearance profiles, as well as siRNA concentration and mRNA reduction in skeletal muscle, for all four conjugates (Figure 7). While others have shown that cytotoxic ADCs targeting tumors have improved activity when using stabilized linkers, (44) our data show that for αmTfR1 conjugates, there is no improvement in plasma exposure, tissue delivery, or KD when using linkers more stable than MCC. This result is best explained by Figure 7a showing greater than 80% of the dosed material is no longer in plasma after 4 h. This rapid target-mediated drug disposition is expected for a highly expressed receptor like TfR1 and results in the majority of the AOC being taken into the tissue before any differences in linker stability could have an impact. It would be intriguing to explore whether these linker patterns persist with AOCs featuring extended plasma residence.

Figure 7

Figure 7. (a–c) PK/PD studies with αmTfR1–siHprt AOCs containing four different noncleavable linkers.

Linker Location on the siRNA

The spatial arrangement of the linker on the siRNA molecule could play a pivotal role in determining the stability of the siRNA itself. This influence is likely attributed to heightened steric hindrance, where the size and the proximity of the antibody creates obstacles to enzymes that can affect the stability of the siRNA as part of the conjugate as well as after release inside the cell. Understanding the impact of linker placement on siRNA stability is crucial for optimizing the design and performance of antibody–siRNA conjugates, especially in the context of therapeutic applications where stability is a critical factor for efficacy and duration of action. AOCs were synthesized to evaluate the effect of linker locations at the 3′ or 5′ end of the siRNA sense strand on the target mRNA KD in different tissue types. The AOCs we evaluated were comprised of an αmTfR1 antibody conjugated via an MCC linker to an siHprt at either the 3′ or 5′ end: αmTfR1-Cys-MCC-N-5′-siHprt and αmTfR1-Cys-MCC-N-3′-siHprt. We measured the KD of these AOCs in four tissues: muscle, heart, liver, and lung (Table S1 and Figure S4). These results demonstrate that the siRNA could be linked at either the 3′ or 5′ end of the sense strand without an impact on KD in the four tissues assessed. This also indicates that any remnant of the linker or the antibody at the 3′ or 5′ end of the siRNA sense strand does not impact RISC loading of duplex siRNA. These data show that the concentration of siRNA in tissue (Figure S4e) does not necessarily correlate with activity when comparing across tissues. While 20 nM concentrations in muscle generate approximately 70% KD, greater than 200 nM concentrations can be measured in the liver without achieving the same amount of activity. These data suggest that AOC activity can be impacted by siRNA delivery as well as cell type and, potentially, mechanism of siRNA internalization.
We synthesized additional AOCs to further evaluate the impact of linker positions in the middle of the siRNA sense strand. αhEGFR antibody was lysine conjugated to siKRAS with bicyclo[6.1.0]nonyne (BCN) linkers at siRNA positions 7, 13, or 17 on the sense strand off a 2′ amine (Table S1 and Figure S5). Although we hypothesized that conjugating to the backbone of the siRNA would enhance protection against potential nucleases by antibody steric interference, we observed no impact on plasma PK based on the siRNA backbone linker location.
The impact of siRNA conjugation position on in vivo potency was tested in both liver and muscle with two different targeting antibodies. To assess the impact of siRNA conjugation positions on KD in the liver, we synthesized AOCs containing antimouse ASGPR mAb (αmASGPR) conjugated to siHprt via a BisMal linker at three different positions: 5′ end versus positions 8 or 14 of the sense strand via 2′ primary amine (Figure 8). We measured percent Hprt mRNA expression in the liver. The αmASGPR-Cys-BisMal-siHprt (5′) demonstrated higher KD versus backbone positions 8 or 14 on the sense strand at lower doses, indicating potential interference in siRNA RISC loading with a linker attached at 2′ locations. It is important to note that while conjugates at 2′ positions 8 and 14 were less active at lower doses, they were able to achieve the same maximum KD as the 5′ conjugate when the dose was increased to 2 mg/kg.

Figure 8

Figure 8. (a,b) Liver KD analysis of αmASGPR–siHprt AOCs containing BisMal linkers to siRNA at the 5′ end and positions (pos) 8 or 14 of the sense strand at 96 h postdose.

Additional AOCs were synthesized to evaluate the impact of siRNA conjugation sites on KD in the gastrocnemius. These AOCs were comprised of an αmTfR1 conjugated to siHprt with either a stable MCC or cleavable VC linker (Figure 9a). The MCC linker was evaluated at the 5′ end as well as positions 8 and 14 on the sense strand via a 2′ primary amine. Better gastrocnemius KD was observed for the MCC linker at the 5′ end versus the backbone positions tested for the αmTfR1–siHprt AOCs at 0.2 mg/kg (p < 0.05), once again indicating the potential impact on the RISC loading of the released siRNA with linkers at positions 8 and 14 on the sense strand. An enzyme-cleavable VC linker was then evaluated at position 8 of the sense strand via a 2′ primary amine. The αmTfR1-Cys-VC-siHprt (2′pos8 sense strand) AOC was synthesized to test the hypothesis that the self-immolating enzyme-cleavable linker would have the greatest impact at the center of the sense strand because of the potential for the noncleavable linker to have more steric bulk and prevent RISC loading. We based this hypothesis on in vitro research with similar modifications. (12) However, the AOC with the cleavable VC linker at position 8 of the sense strand showed lower activity compared with a noncleavable MCC linker 5′ end conjugation at lower doses, indicating that propyl amine at 2′ of position 8 of the siRNA sense strand is unlikely to be an optimal 2′ substitution for RISC loading and target gene KD in muscle or liver. The VC linker cleavage kinetics for antibody–siRNA conjugates are not known. It is conceivable that the VC linker at position 8 could be more hindered than at the 5′ end resulting in slower release of siRNA. The Hprt mRNA KD reaches saturation beyond a dose of 1 mg/kg, yet the concentration of siRNA in tissues continues to rise (Figure 9b), indicating that receptor capacity is not the limiting factor. Our hypothesis to explain the absence of a dose response beyond 1 mg/kg, despite higher siRNA concentrations in tissues, is that Hprt is expressed in both myotubes and other cell types (such as fibroblasts and endothelial cells), whereas siRNA delivery primarily targets muscle cells.

Figure 9

Figure 9. (a) Expression of Hprt in muscle and (b) siRNA tissue concentration after treatment with αmTfR1–siHprt AOCs with MCC and VC linkers at various positions on the siHprt at 96 h postdose.

We recognize the extensive work done around internucleotide triester, 2′-linker, and nucleobase conjugation approaches that identified SAR trends for GalNAc and other ligand conjugates. (45) Our study did not delve into these aspects; therefore, we caution against overgeneralization of our findings and recommend further studies to comprehensively understand the impact of siRNA conjugation sites on KD in different tissues and under varying conditions.

Impact of PEG on AOC Activity

PEG increases solubility, reduces nonspecific protein binding, provides protection from enzymes, and reduces clearance of various molecules such as proteins. (46) To assess the impact of PEG on the plasma PK of an AOC, we evaluated a set of AOCs with PEG on the inside versus the outside of siRNA (i.e., mAb-PEG5k-siRNA vs mAb-siRNA-PEG5k). The delivery and KD of PEG AOCs in mice with lymph node carcinoma of the prostate (LNCaP) xenograft were also assessed. These mice were treated with AOCs comprised of antihuman prostate-specific membrane antigen antibody (αhPSMA) conjugated to EGFR siRNA (siEGFR). The PEG was placed either between the mAb and siRNA or on the outside of the siRNA: αhPSMA-Cys-MCC-siEGFR, αhPSMA-Cys-MCC-siEGFR-PEG5k, and αhPSMA-Cys-PEG5k-siEGFR (Table S1 and Figure S6). These AOCs were designed to test the hypothesis that placing PEG on the outside of the siRNA would protect it from nucleases and increase its stability. Observations showed no significant change in plasma PK based on the size or placement of the PEG. However, placing PEG on the outside led to more KD and higher tissue concentration in the tumor, indicating a potential increase in the intracellular stability of siRNA. Furthermore, the placement of PEG5k externally is likely to have an impact on the hydrophilicity of the conjugate. This could influence the tumor-to-liver ratio and subsequent mRNA reduction. In addition, PEG might shield some of the siRNA charge, which could further affect its interaction with cellular components and its overall effectiveness. These concepts are well described in ADCs. (46)
To further evaluate the potential impact of PEGylation on siRNA in the muscle, AOCs were synthesized using an αmTfR1 antibody conjugated at the cysteine via an MCC linker to an siCtnnb1 sense strand with and without PEG5k on the outside (Figure S7). Tissue PK data of these αmTfR1–siCtnnb1 AOCs suggest that PEG5k attached to the 3′ end of siCtnnb1 sense strand (while conjugating the 5′ end to the antibody) increases the amount of siRNA detected in gastrocnemius as well as Ctnnb1 mRNA KD. Specifically, a 1 mg/kg dose with PEG achieved the same tissue concentration as the 3 mg/kg dose without PEG. Moreover, the KD at 1 mg/kg with PEG was comparable to the KD at 3 mg/kg without PEG.
Our data indicate that including PEG in the AOC architecture could help with siRNA tissue PK, likely resulting in increased gene KD efficacy. Further research is needed to understand the optimal size of the PEG needed in these AOCs.

Applicability of siRNA Bioconjugation across Multiple Antibody Isotypes

We can apply siRNA bioconjugation to a variety of antibody isotypes, including human (h) IgG1, hIgG2, hIgG4, rat IgG2a, and rabbit IgG. The structure of the antibody and conditions used for partial reduction determine the conjugation efficiency and DAR distribution of random cysteine conjugation of siRNA. Each antibody isotype requires different ratios of reagents to maximize DAR1, and IgG4 predominantly gives DAR2 (Table 1). We evaluated antibodies for conjugation, including antimouse TfR1, anti-PSMA, anti-EGFR, and antimouse ASGPR. Interestingly, for hIgG4, the use of a monofunctional versus bifunctional linker (MCC, BisMal) influenced the ratio of DAR1 and DAR ≥ 2. This linker-dependent change in DAR distribution may be due to the Fab arm exchange observed in IgG4 antibodies and the ability of the BisMal linker to stabilize the DAR1 structure. While DAR1 yields may vary greatly across antibody isotypes, we can obtain sufficient yields of pure DAR1 for preclinical research. The observed variation in DAR distribution between antibody isotypes for AOCs is consistent with the differences observed with ADCs generated with different isotypes. (47) This correlation is anticipated, given the similar conditions used for partial reduction of the antibodies. However, AOCs present a unique challenge as they require the isolation of only DAR1. This is further complicated by the high MW and negative charge of siRNAs which creates steric and charge repulsion that can slow reactions for less exposed positions on the antibody.
Table 1. Summary of Reaction Conditions and DAR Distributions for Various mAb Isotypes (%)
mAb isotypeTCEP eq(linker) siRNA eqDAR0 (%)DAR1 (%)DAR ≥ 2 (%)isolated DAR 1 yield (%)
hIgG12(MCC) 125571846
hIgG26(MCC) 127403334
hIgG41.75(MCC) 1.2547223116
hIgG41.75(BisMal) 1.2546312325
Rat IgG2a4(MCC) 1.1538412136
Rabbit IgG4(MCC) 1.126413335

Conclusions

Click to copy section linkSection link copied!

AOCs are complex bioconjugates and the data herein illustrate a comprehensive SAR evaluation of siRNA AOCs. This evaluation has allowed us to optimize siRNA chemistries, select linkers, identify points of conjugation on the antibody and siRNA, develop strategies to conjugate an antibody to an siRNA oligonucleotide, and understand the impact of oligonucleotide-to-antibody ratios on AOC activity. Our findings show the importance of selecting appropriate siRNA chemical modifications to improve tissue delivery in vivo and achieve higher target gene KD in tissues of interest. Our platform can accommodate various points of conjugation on the antibody, offering flexibility to choose between cysteine, site-specific cysteine, and sugar conjugations. Initial studies indicate that lysine conjugates are not optimal for AOCs, but additional studies are needed to understand the differences. The siRNA-to-antibody ratio is limited to 1, likely due to high charge of each siRNA and the potential phosphorothioate-mediated nonspecific binding of DAR ≥ 2 AOCs. The AOC platform can accommodate most noncleavable linker chemistries when conjugated on both ends of the sense strand, indicating that remnants of linker attached to the released siRNA sense strand, after AOC digestion in lysosomes, may not interfere with Ago2 protein binding and RISC loading. We found no significant differences in plasma PK, siRNA tissue delivery, and target gene KD between noncleavable linkers like MCC, BisMal, CB, and MB with rapidly internalized αmTfR1 AOCs. Additional evaluation with AOCs that show longer plasma PK are needed to tease out any differences between these noncleavable linkers. Results from our evaluation of cleavable linkers demonstrate that AOCs with less sterically hindered disulfide linkers like PDP, MPT and SS(methyl) are less stable than more sterically hindered disulfide SS(gem-dimethyl). The reduced plasma clearance of AOCs correlated with the increasing steric hindrance around the disulfide bond. The VC linker offered no significant improvement over stable MCC or BisMal linkers, indicating the tolerance for remnant linker attached to siRNA for Ago2 binding and RISC loading. The exploration of conjugation positions, other than 5′ or 3′ end positions on the sense strand, on both strands of siRNA also revealed that the internal conjugation locations show lower activity than the 5′ end conjugation. This is likely due to interference of siRNA duplex interaction with either Ago2 protein or RISC loading. Addition of 5000 molecular weight PEG on the other side of siRNA in an AOC, by sandwiching siRNA between antibody and PEG, showed approximately three-fold improved tissue delivery to the targeted tumor cells with αhPSMA AOC and a five-fold increase in muscle with αmTfR1 AOC. The increased delivery of siRNA resulted in higher target gene KD. PEG is likely providing steric shielding to nucleases and may reduce nonspecific protein binding of AOC.
Overall, our SAR study provides important insights into the design and optimization of siRNA AOCs, identifying key factors that can influence PK, tissue delivery, and efficacy of these complex bioconjugates. These findings are consistent across multiple antibody targets and siRNAs, suggesting these design principles can be applied to the entire AOC class. By understanding the SAR of AOCs, researchers can continue to develop new and improved therapeutics for a range of diseases and conditions.

Experimental Section

Click to copy section linkSection link copied!

siRNA Synthesis

The oligonucleotide siRNA sequences were assembled on solid phase using well-described and standard phosphoramidite methodology. Single strands were synthesized on a MerMade 12 synthesizer (BioAutomation) with phosphoramidite monomers (2′OMe, 2′F, LNAs, inverted abasic, C6NH2, and C6SSC6dT) and universal controlled pore glass solid support purchased from commercial sources. The vinylphosphonate UNA-MOE-U phosphoramidite monomer was synthesized in-house as per the synthesis shown in Scheme S1. (48) All phosphoramidites were used at a concentration of 100 mM in 100% anhydrous acetonitrile utilizing a 2.5 molar excess of ETT activator in acetonitrile with 2 × 6 min coupling time. Oxidation of phosphite linkages was achieved using 0.02 M I2 in 9:1 acetonitrile/H2O for 60 s. Sulfurization was achieved with 0.05 M DDTT (3-dimethylaminomethylene amino-3H-1,2,4-dithiazole-5-thione) in 60/40 pyridine/acetonitrile for 6 min per cycle. Capping was performed with tetrahydrofuran (THF)/pyridine/acetic anhydride for 60 s and 16% methyl imidazole in THF for 60 s per cycle. Deblocking was performed with 3% trichloroacetic acid in acetonitrile for 2 × 40 s per cycle. Cleavage of siRNA from solid support and deprotection of all protecting groups was accomplished using a 1:1 ratio of 28% aqueous ammonia and 40% methylamine in methanol for 2 h at room temperature. Crude oligonucleotides were purified using strong anion exchange with phosphate buffers (pH 7.2) containing sodium bromide; the desired fractions were chosen based on LCMS analysis, then pooled and desalted. The identification and purity of the single strands were determined by LCMS and RP-HPLC followed by lyophilization. The single strands were then dissolved in H2O and equal molar amounts of antisense and sense strand were annealed by heating to 65 °C for 5 min and cooling to room temperature to form the final duplex, which was analyzed by anion exchange chromatography.

Characterization of Oligonucleotides

Single-strand purities were measured by ion-pair RP-HPLC using an XBridge C18 OST column (Waters, Milford MA; 2.1 × 50 mm, 2.5 μm) with a gradient from 6 to 35% mobile phase B over 15 min at a flow rate of 0.25 mL/min and column temperature of 65 °C. Mobile phase A consisted of 100 mM 1,1,1,3,3,3-hexafluoroisopropanol (HFIP), 16.3 mM triethylamine (TEA), and 1% methanol. Mobile phase B consisted of 100 mM HFIP, 16.3 mM TEA, and 95% methanol. All single strands were at least 85% pure. Single-strand identities were confirmed by mass spectrometry (MS), on LCQ Deca XP + Ion Trap MS (Thermo Finnigan, San Jose, CA), used in line with ion-pair reversed-phase HPLC. The MS was run in negative ion mode with a capillary temperature of 315 °C and voltage of −100 V. Data analysis was performed with ProMass deconvolution software (Novatia, Newtown, PA). Duplex purity was measured by anion exchange chromatography using a DNAPac-200 column (Thermo Fisher, Waltham, MA; 4 × 250 mm) with a gradient of 15–58% mobile phase B over 9 min with a flow rate of 1 mL/min. Mobile phase A contained 20 mM sodium phosphate buffer and 20% ethanol; mobile phase B consisted of mobile phase A plus 800 mM NaClO4. All duplexes were at least 90% pure.

siRNA-PEG Synthesis

The siRNA sense strands used for conjugation were synthesized with a C6 amino or C6 sulfur linker at the 5′- or 3′-end, or 2′-internal modifications (C3 amino). siRNAs with 3′ sense strand C6SSC6dT modifications were dissolved in 100 mM phosphate buffer pH 7.4 then reduced with 20 eq TCEP at room temperature. Excess TCEP was removed using ultrafiltration into 20 mM phosphate buffer with 150 mM sodium chloride then added to 3 equiv of maleimide-PEG5k or 5 equiv of orthopyridyl disulfide PEG5k (Laysan Bio, Arab AL) for a reducible linkage. Resulting siRNA-PEG was purified using the strong anion exchange chromatography method described for AOCs below. The desired fractions were then isolated using ultrafiltration with 3 kDa molecular weight cutoff (MWCO). siRNA-PEG purity was measured by anion exchange chromatography using an Agilent 1200 with a Thermo Fisher ProPac SAX-10 BioLC column (250 × 4 mm) at ambient temperature. siRNA-PEG was separated from unconjugated siRNA and excess PEG with a gradient of 10–60% mobile phase B over 8 min and flow rate at 0.75 mL/min, monitoring at 260 nm. Mobile phase A was 10 mM Tris Base pH 7.2 with 20% ethanol, mobile phase B contained A plus 1.5 M sodium chloride. All siRNA-PEG purities were greater than 95%.

Linker-siRNA Generation

SiRNA duplex in 50 mM phosphate buffer at a concentration of 100 mg/mL was pH adjusted with sodium hydroxide to pH 7.4. The siRNA was then diluted with dimethyl sulfoxide (DMSO) to achieve a final DMSO concentration of 50% once the linker was added. Linker (10 equiv to the siRNA) dissolved in DMSO was added to the siRNA and mixed at room temperature for 30 min. The progress of the reaction was monitored by MS, and the absence of a sense strand indicated the completion of the conjugation. Excess linker was removed using ultrafiltration with a 3 kDa MWCO. Linker-siRNA purity was not determined and was used in situ. Disulfide linkers used were succinimidyl 3-(2-pyridyldithio)propionate (SPDP), 4-succinimidyloxycarbonyl-alpha-methyl-α(2-pyridyldithio)toluene (SMPT), SS[methyl] (2,5-dioxopyrrolidin-1-yl 4-(pyridine-2-yldisulfanyl)pentanoate), and SS[gem-dimethyl] 2,5-dioxopyrrolidin-1-yl 4-methyl-4-((5-nitropyridin-2-yl)disulfanyl)pentanoate (Annova Chem, San Diego, CA). Maleimide linkers used were NHS-PEG5k-maleimide (Laysan Bio, Arab, AL), maleimidocaproyl-l-valine-l-citrulline-p-aminobenzyl alcohol p-nitrophenyl carbonate (MC-Val-Cit–PAB-PNP) (Broadpharm, San Diego, CA), succinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC) (Thermo Fisher, Waltham, MA), bis-maleimide-dPEG4-TFP ester (BisMal) (Quanta Biodesign, Plain City, OH), sodium 4-((4-(cyanoethynyl)benzoyl)oxy)-2,3,5,6-tetrafluorobenzenesulfonate (CBTF), and MB-N-hydroxysuccinimide ester (Sigma, St. Louis, MO). Click linkers used were TCO-PEG4-NHS, NHS-PEG1-Azide (Broadpharm, San Diego, CA), and methyltetrazine-PEG4-NHS (Sigma, St. Louis, MO).

Cysteine, Lysine, and Glycan Conjugations

Cysteine-conjugated AOCs were generated using a standard random cysteine conjugation method. The interchain disulfide bonds of the antibody were partially reduced with TCEP (Thermo Fisher, Waltham, MA) at 37 °C for 2–4 h prior to conjugation with a maleimide linker-oligonucleotide (molar equivalents of TCEP and siRNA-linker required for conjugation to each antibody isotype are listed in Table 1). Lysine-conjugated AOCs were generated using a standard random lysine conjugation method (with the degree of conjugation relative to the antibody). The antibody at 10 mg/mL in PBS (pH 7.4) was incubated with 2 equiv relative to antibody of NHS-linker (TCO, maleimide, azide) at room temperature for 90 min. Excess linker was removed using 50 kDa MWCO ultrafiltration prior to conjugation with 1.75 equiv of a functionalized (tetrazine, sulfhydryl, BCN)-oligonucleotide. Glycan-conjugated AOCs were generated using Asn297 TCO modified antibody from Fisher Scientific, using SiteClick and conjugated with a methyl tetrazine-oligonucleotide. All reaction mixtures after the conjugation were purified on fast protein liquid chromatography using a preparative anion exchange purification method to isolate the DAR1 antibody–siRNA conjugate.

Preparative Anion Exchange Purification

SAX purification was performed on an ÄKTA Pure system with a Sartobind Q (Sartorius, Göttingen, Germany; Nano 3 mL with 8 mm bed height) at ambient temperature with a gradient of 20–40% mobile phase B over 20 column volumes and a flow rate of 3 mL/min, monitoring at ultraviolet (UV) 220, 260, and 280 nm. Mobile phase A was 20 mM sodium phosphate buffer pH 7.2; mobile phase B contained A plus 1.5 M sodium chloride (Figure S8).

Overview of Cysteine Conjugation Using NEM Capping with or without DHAA Oxidation

AOCs (αhEGFR-siKRAS) were generated via cysteine conjugation. Cysteine conjugates were made by partially reducing antibody interchain disulfide bonds with TCEP to react with maleimide on the siRNA. On human IgG1 antibodies, the primary location of siRNA conjugation is on the cysteine of the heavy chain previously forming the heavy chain–light chain disulfide. (47) During this process, some cysteine remained unreacted.

Overview of Free Cysteine Determination

The average number of free cysteines on DAR1 AOCs was estimated by quantifying the amount of fluorescein-5-maleimide (AnaSpec, Fremont, CA) able to react with the antibody relative to a standard curve measured by SEC-HPLC. For each replicate, 0.25 mg of mAb or AOC in PBS at 5 mg/mL was incubated with 5 mol equiv of fluorescein-5-maleimide (5 μL at 0.715 mg/mL in DMSO) at room temperature for 1 h protected from light (5 equiv was selected based on optimization experiments to determine the amount of fluorescein-5-maleimide needed to saturate the free cysteine reaction on mAb reduced with 1.5 EQ of TCEP). After a 3× dilution, 30 μL of each sample was tested by SEC-HPLC (Yarra SEC-3000 column, 1 mL/min flow rate, mobile phase: phosphate buffer +300 mM NaCl) the absorbance at 495 nm was measured and the AUCs for peaks corresponding to the antibody or DAR1 AOC were calculated and compared with a standard curve of fluorescein-5-maleimide to quantify the number of cysteine available for reaction.

Purity Statement

AOCs with a DAR of 1 are at least 95% pure by strong anion exchange chromatography and SEC. The single-strand oligonucleotides used to make the duplex siRNAs for conjugation were at least 85% pure by the appropriate HPLC methods and identified by MS.

Animal Studies

All animal procedures were approved by the Institutional Animal Care and Use Committee of Explora BioLabs in accordance with guidelines of the Association for Assessment and Accreditation of Laboratory Animal Care. Mice were housed in ventilated cages under a 12 h light–dark cycle and fed ad libitum with standard rodent chow. All in vivo experiments were conducted using four mice per group, each weighing ∼24 g.
In vivo studies in wild-type mice were performed in female Hsd/ICR (Envigo) mice. All the dosing was based on the siRNA weight, not the total conjugate (1 mg/kg = 1 mg siRNA/kg). Mice were dosed intravenously at 5 mL/kg. At end of study, mice were euthanized and tissues removed for analysis. For the assessment of delivery to tumors, male Crl:SHO-PrkdcscidHrhr (Charles River) mice were inoculated in the right flank subcutaneously with 7 million LNCaP cells in a mixture of 0.2 mL total of RPMI 1640 with matrigel (1:1). Mice were randomly grouped when tumors reached 200 mm3 and given an intravenous dose of the test article at 5 mL/kg. At end of study, or when tumors reached 2000 mm3, mice were euthanized and tissues removed for analysis.

Pharmacodynamic Evaluation

Tissue was processed for RNA extraction using the Zymo Direct-zol-96 RNA purification kit (Zymo Research Corporation, Irvine, CA). Once RNA quality was confirmed, cDNA was generated from purified RNA and used in quantitative reverse transcription polymerase chain reaction analysis for gene expression for each of the targets of interests. (49) The reference mRNA transcripts for Ppib were used in ΔΔCt calculations for tissue samples. (50)
To evaluate siRNA concentrations in plasma or tissues, custom stem-loop-RT-qPCR assays were developed as described previously. (51) For each siRNA, a specific stem-loop-RT-qPCR assay was designed to quantify the antisense strand, using custom DNA forward, reverse, and reverse transcription primers (Integrated DNA Technologies, Coralville, IA) and a custom Taqman probe (Thermo Fisher, Waltham, MA). Noncompartmental analysis was performed on individual mouse plasma total siRNA concentration versus time data to estimate PK parameters for AUC and clearance using Phoenix WinNonlin v8.3 (Certara, Princeton, NJ). An intravenous bolus administration model (Plasma 200–202) was used and AUC was calculated using the “linear-up log-down” calculation method.

AOC Characterization

The isolated DAR1 antibody–siRNA conjugate was then characterized by SAX, SEC, reduced capillary gel electrophoresis (RCGE), UV, and MS.

Analytical Anion Exchange Chromatography

After bioconjugation, the DAR distribution of the AOC reaction mixture was analyzed by SAX. SAX analysis was performed on an Agilent 1200 with Thermo Fisher ProPac SAX-10 BioLC column (250 × 4 mm) at ambient temperature with a gradient of 10–60% mobile phase B over 8 min and flow rate at 0.75 mL/min, monitoring at UV 220, 260, and 280 nm. Mobile phase A was 10 mM Tris Base pH 7.2 with 20% ethanol; mobile phase B contained A plus 1.5 M sodium chloride. DAR1 was then isolated by column chromatography (Figure S9a). The DAR1 final product was then characterized by SAX [same method as described above; UV (Figure S9b)], SEC (Figure S9c), RCGE (Figure S9d), and MS (Figure S9e).

UV Spectroscopy

AOC concentrations were determined using the absorbance at 280 nm and an extinction coefficient of 2.28 L·g–1·cm–1 for DAR1 and 3.43 L·g–1·cm–1 for DAR2. Absorbance ratios at 260 nm/280 nm were also measured to confirm DAR. The extinction coefficient used for DAR1 and DAR2 AOCs was calculated using the UV absorbance at 280 nM and the BCA measurement of antibody concentration (compared to a standard curve with the same unconjugated antibody) of more than 20 different purified conjugates. The values were within 13 and 9% (DAR1 and DAR2) of the values calculated using the mAb and siRNA extinction coefficients and the formula below. The concentration of the mAb portion of an AOC was used to calculate siRNA concentration and total AOC concentration using the siRNA MW of 14,100 Da and antibody MW of 150,000 Da. A 1 mg/kg siRNA dose corresponds to 10.6 mg/kg mAb dose and 11.6 mg/kg AOC dose.
[mAb]=A280smplε280,mAb+DARε280,siRNA
SiRNA single strand concentrations were determined by using the absorbance of each strand at 260 nM and an extinction coefficient estimated using the nearest neighbor method. Duplex siRNA concentrations were calculated based on the known amount of each single strand used and the appropriate dilutions. When needed, duplex extinction coefficients were estimated using the formula below including a correction factor of 0.8 to account for the hypochromic effect of duplexed nucleic acids.
ε260,siRNA=(ε260,antisensestrand+ε260,sensestrand)×0.8

Size Exclusion

SEC analysis was performed on an Agilent 1200 with a Yarra 3 μm SEC-300 column (300 × 7.8 mm) at ambient temperature with an isocratic flow of 55 mM potassium phosphate monobasic 62 mM sodium phosphate dibasic 100 mM sodium sulfate with 0.05% sodium azide pH 6.7 buffer with a flow rate of 1 mL/min for 15 min, monitoring at UV 260 and 280 nm.

Reduced Capillary Electrophoresis

Reduced capillary electrophoresis-sodium dodecyl sulfate was performed on ProteinSimple Maurice Plus instrument using Maurice CE-SDS PLUS cartridges according to manual instructions. Samples were prepared to 1 mg/mL by protein concentration. Internal standard and 2-mercaptoethanol were added, then heated to 70 °C for 10 min. Reduced samples were run with a sample load of 20 s at 4600 V, and separation for 25 min at 5750 V.

MS

Intact MS was performed by Thermo Fisher with a Q Exactive BioPharma. 5 μg of sample was loaded onto an Acquity UPLC protein BEH SEC column (4.5 × 100 mm, 1.7 μm) and eluted with 50 mM ammonium acetate at a flow rate of 0.3 mL/min and a column temperature of 55 °C. MS was run with a spray voltage of 4.2 kV and an ion transfer tube temperature of 320 °C.

Supporting Information

Click to copy section linkSection link copied!

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.4c00802.

  • Please see siRNA AOC Supplement. siRNA sequence and chemical modifications for evaluated test articles; siRNA conjugation comprehensive guide; noncompartmental analysis of plasma PK data in Figure 5b; plasma PK, tissue concentration, and mRNA expression of αmTfR1-Cys-MCC-siMstn conjugates; DAR1 versus DAR2 SEC comparison; in vitro stability of αhEGFR-siKRAS AOCs; KD and tissue concentration of αmTfR1-siHprt AOCs; plasma PK of αhEGFR-siKRAS AOCs; tumor delivery and KD of αhPSMA-siEGFR AOCs; PK of αmTfR1-siCtnnb1 AOCs; purification of antibody–siRNA conjugation reaction mixture by SAX; purification and characterization of DAR1 AOC; UNA vinylphosphonate (vpUq) amidite synthesis (PDF)

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

Click to copy section linkSection link copied!

  • Corresponding Author
    • Venkata Ramana Doppalapudi - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States Email: [email protected]
  • Authors
    • Michael Cochran - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    • Danny Arias - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    • Rob Burke - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United StatesPresent Address: Seawolf Therapeutics, One Sansome Street Suite 3630, San Francisco, CA 94104, USA
    • David Chu - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United StatesPresent Address: Capstan Therapeutics, 9880 Campus Point Drive, San Diego, CA 92121
    • Gulin Erdogan - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    • Michael Hood - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    • Philip Kovach - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    • Hae Won Kwon - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    • Yanling Chen - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    • Michael Moon - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United StatesPresent Address: Amgen Inc., 750 Gateway Blvd., Suite 100, South San Francisco, California 94080
    • Christopher D. Miller - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United StatesPresent Address: California North state University College of Medicine, 9700 W Taron Dr, Elk Grove, CA 95757
    • Hanhua Huang - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
    • Arthur Levin - Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125. San Diego, California 92121, United States
  • Author Contributions

    The manuscript was written through contributions of all authors. All authors have given approval to the final version of the manuscript.

  • Notes
    The authors declare the following competing financial interest(s): At the time this research was conducted, all authors were employees of Avidity Biosciences and may have equity in the company.

Acknowledgments

Click to copy section linkSection link copied!

Authors acknowledge John Manhard for generating antibodies with site-specific cysteine conjugation handles. Thanks to Baorui Lin and Zhanlei Wei of WuXi TIDES for their work on the synthesis and scale-up of vp-UNA-3′-phosphoramidite. Special thanks to Son Lam and Han Cho for technical review of the manuscript. The authors would like to thank Gemma Hall, DPhil, and Megan Hotard Jarrell, MS, from Lighthouse Medical Communications, New York, NY, USA, for providing medical writing support. Medical writing support was funded by Avidity Biosciences, San Diego, CA, USA, in accordance with Good Publication Practice (GPP3) guidelines.

Abbreviations

Click to copy section linkSection link copied!

% ID

percent of injected dose

ADC

antibody–drug conjugate

Ago2

Argonaute2

AOC

antibody–oligonucleotide conjugate

ASGPR

asialoglycoprotein receptor

ASO

antisense oligonucleotide

BCN

bicyclo[6.1.0]nonyne

BisMal

bismaleimide

CBTF

sodium 4-((4-(cyanoethynyl)benzoyl)oxy)-2,3,5,6-tetrafluorobenzenesulfonate

DAR

drug-to-antibody ratio

DHAA

dehydroascorbic acid

EGFR

epidermal growth factor receptor

Eq

equivalent

GalNAc

N-acetylgalactosamine

HFIP

1,1,1,3,3,3-hexafluoroisopropanol

HPLC

high-performance liquid chromatography

KD

knockdown

LNA

locked nucleic acid

LNCaP

lymph node carcinoma of the prostate

mAb

monoclonal antibody

MBS

m-maleimidobenzoyl-N-hydroxysuccinimide ester

MS

mass spectrometry

NEM

N-ethylmaleimide

NHS

N-hydroxysuccinimide

P═S

phosphorothioates

PBS

phosphate buffered saline

PEG

polyethylene glycol

PSMA

prostate-specific membrane antigen

RCGE

reduced capillary gel electrophoresis

RISC

RNA-induced silencing complex

RT-qPCR

quantitative reverse transcription polymerase chain reaction

SAR

structure–activity relationship

SAX

strong anion exchange chromatography

SEC

size exclusion chromatography

siRNA

small interfering RNA

SMCC

succinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate

SMPT

4-succinimidyloxycarbonyl-alpha-methyl-α(2-pyridyldithio)toluene

SPDP

succinimidyl 3-(2-pyridyldithio)propionate

SS

disulfide

TCEP

tris (2-carboxyethyl) phosphine

TEA

triethylamine

TfR1

transferrin receptor 1

UNA

unlocked nucleic acid

VC

valine-citrulline

References

Click to copy section linkSection link copied!

This article references 51 other publications.

  1. 1
    Arslan, F. B.; Ozturk, K.; Calis, S. Antibody-Mediated Drug Delivery. Int. J. Pharm. 2021, 596, 120268,  DOI: 10.1016/j.ijpharm.2021.120268
  2. 2
    Awwad, S.; Angkawinitwong, U. Overview of Antibody Drug Delivery. Pharmaceutics 2018, 10 (3), 83,  DOI: 10.3390/pharmaceutics10030083
  3. 3
    Dovgan, I.; Koniev, O.; Kolodych, S.; Wagner, A. Antibody-Oligonucleotide Conjugates as Therapeutic, Imaging, and Detection Agents. Bioconjugate Chem. 2019, 30 (10), 24832501,  DOI: 10.1021/acs.bioconjchem.9b00306
  4. 4
    Ovacik, M.; Lin, K. Tutorial on Monoclonal Antibody Pharmacokinetics and Its Considerations in Early Development. Clin. Transl. Sci. 2018, 11 (6), 540552,  DOI: 10.1111/cts.12567
  5. 5
    Chery, J. RNA Therapeutics: RNAi and Antisense Mechanisms and Clinical Applications. Postdoc J. 2016, 4 (7), 3550,  DOI: 10.14304/SURYA.JPR.V4N7.5
  6. 6
    Levin, A. A. Treating Disease at the RNA Level with Oligonucleotides. N. Engl. J. Med. 2019, 380 (1), 5770,  DOI: 10.1056/NEJMra1705346
  7. 7
    Burnett, J. C.; Rossi, J. J. RNA-Based Therapeutics: Current Progress and Future Prospects. Chem. Biol. 2012, 19 (1), 6071,  DOI: 10.1016/j.chembiol.2011.12.008
  8. 8
    Crooke, S. T.; Witztum, J. L.; Bennett, C. F.; Baker, B. F. RNA-Targeted Therapeutics. Cell Metab. 2018, 27 (4), 714739,  DOI: 10.1016/j.cmet.2018.03.004
  9. 9
    Hu, B.; Zhong, L.; Weng, Y.; Peng, L.; Huang, Y.; Zhao, Y.; Liang, X.-J. Therapeutic siRNA: State of the Art. Signal Transduct. Target. Ther. 2020, 5 (1), 101,  DOI: 10.1038/s41392-020-0207-x
  10. 10
    Egli, M.; Manoharan, M. Chemistry, Structure and Function of Approved Oligonucleotide Therapeutics. Nucleic Acids Res. 2023, 51 (6), 25292573,  DOI: 10.1093/nar/gkad067
  11. 11
    Janas, M. M.; Schlegel, M. K.; Harbison, C. E.; Yilmaz, V. O.; Jiang, Y.; Parmar, R.; Zlatev, I.; Castoreno, A.; Xu, H.; Shulga-Morskaya, S.; Rajeev, K. G.; Manoharan, M.; Keirstead, N. D.; Maier, M. A.; Jadhav, V. Selection of GalNAc-Conjugated siRNAs with Limited off-Target-Driven Rat Hepatotoxicity. Nat. Commun. 2018, 9 (1), 723,  DOI: 10.1038/s41467-018-02989-4
  12. 12
    Brown, K. M.; Nair, J. K.; Janas, M. M.; Anglero-Rodriguez, Y. I.; Dang, L. T. H.; Peng, H.; Theile, C. S.; Castellanos-Rizaldos, E.; Brown, C.; Foster, D.; Kurz, J.; Allen, J.; Maganti, R.; Li, J.; Matsuda, S.; Stricos, M.; Chickering, T.; Jung, M.; Wassarman, K.; Rollins, J.; Woods, L.; Kelin, A.; Guenther, D. C.; Mobley, M. W.; Petrulis, J.; McDougall, R.; Racie, T.; Bombardier, J.; Cha, D.; Agarwal, S.; Johnson, L.; Jiang, Y.; Lentini, S.; Gilbert, J.; Nguyen, T.; Chigas, S.; LeBlanc, S.; Poreci, U.; Kasper, A.; Rogers, A. B.; Chong, S.; Davis, W.; Sutherland, J. E.; Castoreno, A.; Milstein, S.; Schlegel, M. K.; Zlatev, I.; Charisse, K.; Keating, M.; Manoharan, M.; Fitzgerald, K.; Wu, J.-T.; Maier, M. A.; Jadhav, V. Expanding RNAi Therapeutics to Extrahepatic Tissues with Lipophilic Conjugates. Nat. Biotechnol. 2022, 40 (10), 15001508,  DOI: 10.1038/s41587-022-01334-x
  13. 13
    Roberts, T. C.; Langer, R.; Wood, M. J. A. Advances in Oligonucleotide Drug Delivery. Nat. Rev. Drug Discovery 2020, 19 (10), 673694,  DOI: 10.1038/s41573-020-0075-7
  14. 14
    Cuellar, T. L.; Barnes, D.; Nelson, C.; Tanguay, J.; Yu, S.-F.; Wen, X.; Scales, S. J.; Gesch, J.; Davis, D.; van Brabant Smith, A.; Leake, D.; Vandlen, R.; Siebel, C. W. Systematic Evaluation of Antibody-Mediated siRNA Delivery Using an Industrial Platform of THIOMAB-siRNA Conjugates. Nucleic Acids Res. 2015, 43 (2), 11891203,  DOI: 10.1093/nar/gku1362
  15. 15
    Sela, T.; Mansø, M.; Siegel, M.; Marban-Doran, C.; Ducret, A.; Niewöhner, J.; Ravn, J.; Martin, R. E.; Sommer, A.; Lohmann, S.; Krippendorff, B.-F.; Ladefoged, M.; Indlekofer, A.; Quaiser, T.; Bueddefeld, F.; Koller, E.; Mohamed, M. Y.; Oelschlaegel, T.; Gothelf, K. V.; Hofer, K.; Schumacher, F. F. Diligent Design Enables Antibody-ASO Conjugates with Optimal Pharmacokinetic Properties. Bioconjugate Chem. 2023, 34 (11), 20962111,  DOI: 10.1021/acs.bioconjchem.3c00393
  16. 16
    Sugo, T.; Terada, M.; Oikawa, T.; Miyata, K.; Nishimura, S.; Kenjo, E.; Ogasawara-Shimizu, M.; Makita, Y.; Imaichi, S.; Murata, S.; Otake, K.; Kikuchi, K.; Teratani, M.; Masuda, Y.; Kamei, T.; Takagahara, S.; Ikeda, S.; Ohtaki, T.; Matsumoto, H. Development of Antibody-siRNA Conjugate Targeted to Cardiac and Skeletal Muscles. J. Controlled Release 2016, 237, 113,  DOI: 10.1016/j.jconrel.2016.06.036
  17. 17
    Malecova, B.; Burke, R. S.; Cochran, M.; Hood, M. D.; Johns, R.; Kovach, P. R.; Doppalapudi, V. R.; Erdogan, G.; Arias, J. D.; Darimont, B.; Miller, C. D.; Huang, H.; Geall, A.; Younis, H. S.; Levin, A. A. Targeted Tissue Delivery of RNA Therapeutics Using Antibody-Oligonucleotide Conjugates (AOCs). Nucleic Acids Res. 2023, 51 (12), 59015910,  DOI: 10.1093/nar/gkad415
  18. 18
    Cao, W.; Li, R.; Pei, X.; Chai, M.; Sun, L.; Huang, Y.; Wang, J.; Barth, S.; Yu, F.; He, H. Antibody-siRNA Conjugates (ARC): Emerging siRNA Drug Formulation. Med. Drug Discovery 2022, 15, 100128,  DOI: 10.1016/j.medidd.2022.100128
  19. 19
    Chernikov, I. V.; Vlassov, V. V.; Chernolovskaya, E. L. Current Development of siRNA Bioconjugates: From Research to the Clinic. Front. Pharmacol 2019, 10, 444,  DOI: 10.3389/fphar.2019.00444
  20. 20
    Tai, W. Current Aspects of siRNA Bioconjugate for In Vitro and In Vivo Delivery. Molecules 2019, 24 (12), 2211,  DOI: 10.3390/molecules24122211
  21. 21
    Sun, M. M. C.; Beam, K. S.; Cerveny, C. G.; Hamblett, K. J.; Blackmore, R. S.; Torgov, M. Y.; Handley, F. G. M.; Ihle, N. C.; Senter, P. D.; Alley, S. C. Reduction-Alkylation Strategies for the Modification of Specific Monoclonal Antibody Disulfides. Bioconjugate Chem. 2005, 16 (5), 12821290,  DOI: 10.1021/bc050201y
  22. 22
    Shen, B.-Q.; Xu, K.; Liu, L.; Raab, H.; Bhakta, S.; Kenrick, M.; Parsons-Reponte, K. L.; Tien, J.; Yu, S.-F.; Mai, E.; Li, D.; Tibbitts, J.; Baudys, J.; Saad, O. M.; Scales, S. J.; McDonald, P. J.; Hass, P. E.; Eigenbrot, C.; Nguyen, T.; Solis, W. A.; Fuji, R. N.; Flagella, K. M.; Patel, D.; Spencer, S. D.; Khawli, L. A.; Ebens, A.; Wong, W. L.; Vandlen, R.; Kaur, S.; Sliwkowski, M. X.; Scheller, R. H.; Polakis, P.; Junutula, J. R. Conjugation Site Modulates the in Vivo Stability and Therapeutic Activity of Antibody-Drug Conjugates. Nat. Biotechnol. 2012, 30 (2), 184189,  DOI: 10.1038/nbt.2108
  23. 23
    Zhou, Q.; Stefano, J. E.; Manning, C.; Kyazike, J.; Chen, B.; Gianolio, D. A.; Park, A.; Busch, M.; Bird, J.; Zheng, X.; Simonds-Mannes, H.; Kim, J.; Gregory, R. C.; Miller, R. J.; Brondyk, W. H.; Dhal, P. K.; Pan, C. Q. Site-Specific Antibody-Drug Conjugation through Glycoengineering. Bioconjugate Chem. 2014, 25 (3), 510520,  DOI: 10.1021/bc400505q
  24. 24
    Li, X.; Fang, T.; Boons, G. Preparation of Well-Defined Antibody-Drug Conjugates through Glycan Remodeling and Strain-Promoted Azide-Alkyne Cycloadditions. Angew. Chem., Int. Ed. 2014, 53 (28), 71797182,  DOI: 10.1002/anie.201402606
  25. 25
    Brun, M.-P.; Gauzy-Lazo, L. Protocols for Lysine Conjugation. In Antibody-Drug Conjugates; Ducry, L., Ed.; Methods in Molecular Biology; Humana Press: Totowa, NJ, 2013; Vol. 1045, pp 173187. DOI: 10.1007/978-1-62703-541-5_10 .
  26. 26
    Kumar, P.; Parmar, R. G.; Brown, C. R.; Willoughby, J. L. S.; Foster, D. J.; Babu, I. R.; Schofield, S.; Jadhav, V.; Charisse, K.; Nair, J. K.; Rajeev, K. G.; Maier, M. A.; Egli, M.; Manoharan, M. 5′-Morpholino Modification of the Sense Strand of an siRNA Makes It a More Effective Passenger. Chem. Commun. 2019, 55 (35), 51395142,  DOI: 10.1039/C9CC00977A
  27. 27
    Allerson, C. R.; Sioufi, N.; Jarres, R.; Prakash, T. P.; Naik, N.; Berdeja, A.; Wanders, L.; Griffey, R. H.; Swayze, E. E.; Bhat, B. Fully 2‘-Modified Oligonucleotide Duplexes with Improved in Vitro Potency and Stability Compared to Unmodified Small Interfering RNA. J. Med. Chem. 2005, 48 (4), 901904,  DOI: 10.1021/jm049167j
  28. 28
    Foster, D. J.; Brown, C. R.; Shaikh, S.; Trapp, C.; Schlegel, M. K.; Qian, K.; Sehgal, A.; Rajeev, K. G.; Jadhav, V.; Manoharan, M.; Kuchimanchi, S.; Maier, M. A.; Milstein, S. Advanced siRNA Designs Further Improve In Vivo Performance of GalNAc-siRNA Conjugates. Mol. Ther. 2018, 26 (3), 708717,  DOI: 10.1016/j.ymthe.2017.12.021
  29. 29
    Nair, J. K.; Willoughby, J. L. S.; Chan, A.; Charisse, K.; Alam, M. R.; Wang, Q.; Hoekstra, M.; Kandasamy, P.; Kel’in, A. V.; Milstein, S.; Taneja, N.; O’Shea, J.; Shaikh, S.; Zhang, L.; Van Der Sluis, R. J.; Jung, M. E.; Akinc, A.; Hutabarat, R.; Kuchimanchi, S.; Fitzgerald, K.; Zimmermann, T.; Van Berkel, T. J. C.; Maier, M. A.; Rajeev, K. G.; Manoharan, M. Multivalent N -Acetylgalactosamine-Conjugated siRNA Localizes in Hepatocytes and Elicits Robust RNAi-Mediated Gene Silencing. J. Am. Chem. Soc. 2014, 136 (49), 1695816961,  DOI: 10.1021/ja505986a
  30. 30
    Haraszti, R. A.; Roux, L.; Coles, A. H.; Turanov, A. A.; Alterman, J. F.; Echeverria, D.; Godinho, B. M. D. C.; Aronin, N.; Khvorova, A. 5′-Vinylphosphonate Improves Tissue Accumulation and Efficacy of Conjugated siRNAs in Vivo. Nucleic Acids Res. 2017, 45 (13), 75817592,  DOI: 10.1093/nar/gkx507
  31. 31
    Parmar, R.; Willoughby, J. L. S.; Liu, J.; Foster, D. J.; Brigham, B.; Theile, C. S.; Charisse, K.; Akinc, A.; Guidry, E.; Pei, Y.; Strapps, W.; Cancilla, M.; Stanton, M. G.; Rajeev, K. G.; Sepp-Lorenzino, L.; Manoharan, M.; Meyers, R.; Maier, M. A.; Jadhav, V. 5′-(E)-Vinylphosphonate: A Stable Phosphate Mimic Can Improve the RNAi Activity of siRNA-GalNAc Conjugates. ChemBioChem 2016, 17 (11), 985989,  DOI: 10.1002/cbic.201600130
  32. 32
    Brown, C. R.; Gupta, S.; Qin, J.; Racie, T.; He, G.; Lentini, S.; Malone, R.; Yu, M.; Matsuda, S.; Shulga-Morskaya, S.; Nair, A. V.; Theile, C. S.; Schmidt, K.; Shahraz, A.; Goel, V.; Parmar, R. G.; Zlatev, I.; Schlegel, M. K.; Nair, J. K.; Jayaraman, M.; Manoharan, M.; Brown, D.; Maier, M. A.; Jadhav, V. Investigating the Pharmacodynamic Durability of GalNAc-siRNA Conjugates. Nucleic Acids Res. 2020, 48 (21), 1182711844,  DOI: 10.1093/nar/gkaa670
  33. 33
    Strop, P.; Liu, S.-H.; Dorywalska, M.; Delaria, K.; Dushin, R. G.; Tran, T.-T.; Ho, W.-H.; Farias, S.; Casas, M. G.; Abdiche, Y.; Zhou, D.; Chandrasekaran, R.; Samain, C.; Loo, C.; Rossi, A.; Rickert, M.; Krimm, S.; Wong, T.; Chin, S. M.; Yu, J.; Dilley, J.; Chaparro-Riggers, J.; Filzen, G. F.; O’Donnell, C. J.; Wang, F.; Myers, J. S.; Pons, J.; Shelton, D. L.; Rajpal, A. Location Matters: Site of Conjugation Modulates Stability and Pharmacokinetics of Antibody Drug Conjugates. Chem. Biol. 2013, 20 (2), 161167,  DOI: 10.1016/j.chembiol.2013.01.010
  34. 34
    Miller, C. M.; Donner, A. J.; Blank, E. E.; Egger, A. W.; Kellar, B. M.; Østergaard, M. E.; Seth, P. P.; Harris, E. N. Stabilin-1 and Stabilin-2 Are Specific Receptors for the Cellular Internalization of Phosphorothioate-Modified Antisense Oligonucleotides (ASOs) in the Liver. Nucleic Acids Res. 2016, 44 (6), 27822794,  DOI: 10.1093/nar/gkw112
  35. 35
    Lehot, V.; Kuhn, I.; Nothisen, M.; Erb, S.; Kolodych, S.; Cianférani, S.; Chaubet, G.; Wagner, A. Non-Specific Interactions of Antibody-Oligonucleotide Conjugates with Living Cells. Sci. Rep. 2021, 11 (1), 5881,  DOI: 10.1038/s41598-021-85352-w
  36. 36
    Rocca, C.; Dennin, S.; Gu, Y.; Kim, J.; Chigas, S.; Najarian, D.; Chong, S.; Gutierrez, S.; Butler, J.; Charisse, K.; Robbie, G.; Xu, Y.; Brown, K. Evaluation of Electrophoretic Mobility Shift Assay as a Method to Determine Plasma Protein Binding of siRNA. Bioanalysis 2019, 11 (21), 19271939,  DOI: 10.4155/bio-2019-0151
  37. 37
    Liang, X.; Sun, H.; Shen, W.; Crooke, S. T. Identification and Characterization of Intracellular Proteins That Bind Oligonucleotides with Phosphorothioate Linkages. Nucleic Acids Res. 2015, 43 (5), 29272945,  DOI: 10.1093/nar/gkv143
  38. 38
    Gaus, H.; Miller, C. M.; Seth, P. P.; Harris, E. N. Structural Determinants for the Interactions of Chemically Modified Nucleic Acids with the Stabilin-2 Clearance Receptor. Biochemistry 2018, 57 (14), 20612064,  DOI: 10.1021/acs.biochem.8b00126
  39. 39
    Structure−Activity Relationship of Antibody−Oligonucleotide Conjugates: Evaluating Bioconjugation Strategies for Antibody−Phosphorodiamidate Morpholino Oligomer Conjugates for Drug Development, Manuscript in Preparation.
  40. 40
    Dosio, F.; Brusa, P.; Cattel, L. Immunotoxins and Anticancer Drug Conjugate Assemblies: The Role of the Linkage between Components. Toxins 2011, 3 (7), 848883,  DOI: 10.3390/toxins3070848
  41. 41
    Su, D.; Kozak, K. R.; Sadowsky, J.; Yu, S.-F.; Fourie-O’Donohue, A.; Nelson, C.; Vandlen, R.; Ohri, R.; Liu, L.; Ng, C.; He, J.; Davis, H.; Lau, J.; Del Rosario, G.; Cosino, E.; Cruz-Chuh, J. D.; Ma, Y.; Zhang, D.; Darwish, M.; Cai, W.; Chen, C.; Zhou, H.; Lu, J.; Liu, Y.; Kaur, S.; Xu, K.; Pillow, T. H. Modulating Antibody-Drug Conjugate Payload Metabolism by Conjugation Site and Linker Modification. Bioconjugate Chem. 2018, 29 (4), 11551167,  DOI: 10.1021/acs.bioconjchem.7b00785
  42. 42
    Su, D.; Zhang, D. Linker Design Impacts Antibody-Drug Conjugate Pharmacokinetics and Efficacy via Modulating the Stability and Payload Release Efficiency. Front. Pharmacol 2021, 12, 687926,  DOI: 10.3389/fphar.2021.687926
  43. 43
    Kolodych, S.; Koniev, O.; Baatarkhuu, Z.; Bonnefoy, J.-Y.; Debaene, F.; Cianférani, S.; Van Dorsselaer, A.; Wagner, A. CBTF: New Amine-to-Thiol Coupling Reagent for Preparation of Antibody Conjugates with Increased Plasma Stability. Bioconjugate Chem. 2015, 26 (2), 197200,  DOI: 10.1021/bc500610g
  44. 44
    Lyon, R. P.; Setter, J. R.; Bovee, T. D.; Doronina, S. O.; Hunter, J. H.; Anderson, M. E.; Balasubramanian, C. L.; Duniho, S. M.; Leiske, C. I.; Li, F.; Senter, P. D. Self-Hydrolyzing Maleimides Improve the Stability and Pharmacological Properties of Antibody-Drug Conjugates. Nat. Biotechnol. 2014, 32 (10), 10591062,  DOI: 10.1038/nbt.2968
  45. 45
    Meade, B. R.; Gogoi, K.; Hamil, A. S.; Palm-Apergi, C.; Berg, A. V. D.; Hagopian, J. C.; Springer, A. D.; Eguchi, A.; Kacsinta, A. D.; Dowdy, C. F.; Presente, A.; Lönn, P.; Kaulich, M.; Yoshioka, N.; Gros, E.; Cui, X.-S.; Dowdy, S. F. Efficient Delivery of RNAi Prodrugs Containing Reversible Charge-Neutralizing Phosphotriester Backbone Modifications. Nat. Biotechnol. 2014, 32 (12), 12561261,  DOI: 10.1038/nbt.3078
  46. 46
    Burke, P. J.; Hamilton, J. Z.; Jeffrey, S. C.; Hunter, J. H.; Doronina, S. O.; Okeley, N. M.; Miyamoto, J. B.; Anderson, M. E.; Stone, I. J.; Ulrich, M. L.; Simmons, J. K.; McKinney, E. E.; Senter, P. D.; Lyon, R. P. Optimization of a PEGylated Glucuronide-Monomethylauristatin E Linker for Antibody-Drug Conjugates. Mol. Cancer Ther. 2017, 16 (1), 116123,  DOI: 10.1158/1535-7163.MCT-16-0343
  47. 47
    Wiggins, B.; Liu-Shin, L.; Yamaguchi, H.; Ratnaswamy, G. Characterization of Cysteine-Linked Conjugation Profiles of Immunoglobulin G1 and Immunoglobulin G2 Antibody-Drug Conjugates. J. Pharm. Sci. 2015, 104 (4), 13621372,  DOI: 10.1002/jps.24338
  48. 48
    Prakash, T. P.; Lima, W. F.; Murray, H. M.; Li, W.; Kinberger, G. A.; Chappell, A. E.; Gaus, H.; Seth, P. P.; Bhat, B.; Crooke, S. T.; Swayze, E. E. Identification of Metabolically Stable 5′-Phosphate Analogs That Support Single-Stranded siRNA Activity. Nucleic Acids Res. 2015, 43 (6), 29933011,  DOI: 10.1093/nar/gkv162
  49. 49
    Green, M. R.; Sambrook, J. Quantification of RNA by Real-Time Reverse Transcription-Polymerase Chain Reaction (RT-PCR). Cold Spring Harb. Protoc. 2018, 2018 (10), pdb.prot095042,  DOI: 10.1101/pdb.prot095042
  50. 50
    Livak, K. J.; Schmittgen, T. D. Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2-ΔΔCT Method. Methods 2001, 25 (4), 402408,  DOI: 10.1006/meth.2001.1262
  51. 51
    Chen, C. Real-Time Quantification of microRNAs by Stem-Loop RT-PCR. Nucleic Acids Res. 2005, 33 (20), e179  DOI: 10.1093/nar/gni178

Cited By

Click to copy section linkSection link copied!

This article has not yet been cited by other publications.

Open PDF

Journal of Medicinal Chemistry

Cite this: J. Med. Chem. 2024, 67, 17, 14852–14867
Click to copy citationCitation copied!
https://doi.org/10.1021/acs.jmedchem.4c00802
Published August 28, 2024

Copyright © 2024 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY-NC-ND 4.0 .

Article Views

5433

Altmetric

-

Citations

-
Learn about these metrics

Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.

Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.

The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.

  • Abstract

    Scheme 1

    Scheme 1. (a) Synthetic Scheme for Conjugating an siRNA with a Linker; (b) Antibody–siRNA Conjugate Synthesis via Interchain Disulfide (Cysteine) Conjugation; (c) Antibody–siRNA Conjugate Synthesis via Asn297 Conjugation; (d) Antibody–siRNA Conjugate Synthesis via Lysine Conjugation

    Figure 1

    Figure 1. Typical structure of an siRNA molecule. MCC linker: a heterobifunctional cross-linker that contains two reactive groups: an NHS ester and a maleimide. The NHS ester reacts with primary amines, while the maleimide reacts with sulfhydryl groups. This allows the MCC to form stable bonds with both amine- and sulfhydryl-containing molecules. C6 amino: a linker between the MCC and the siRNA. It provides a sufficient distance between the siRNA and the MCC to minimize any potential steric hindrance or interference with the siRNA’s function. Sense strand 5′ end: the 5′ end of the sense strand of the siRNA. The 5′ end is where the MCC is attached via the C6 amine linker. Sense strand 3′ end: the 3′ end of the sense strand of the siRNA. Antisense strand: designed to be complementary to the target mRNA sequence. It guides the RISC to the target mRNA. Phosphodiester and phosphorothioate bonds: the bonds that connect the nucleotides in the siRNA. Phosphodiester bonds are the standard bonds in RNA, while phosphorothioate bonds are modified versions that contain a sulfur atom, which can provide increased stability to the siRNA.

    Figure 2

    Figure 2. Evaluation of free cysteines available for conjugation on an αhEGFR-Cys-MCC-siAR DAR1 AOC following treatments with TCEP, NEM, and DHAA (n = 3).

    Figure 3

    Figure 3. Impact of siRNA chemical modifications on siRNA stability and activity in targeting Mstn through evaluation of tissue siRNA concentration (TC) and percent mRNA remaining relative to phosphate buffered saline (PBS) in gastrocnemius muscle of mice treated with αmTfR1-Cys-MCC-siMstn AOCs. aAll sense strands were duplexed with the antisense strand: vpUusUfsAoUoUoAfUoUoUoGoUoUoCoUfUoUfGoCoCosUosUo. Abbreviations: X refers to bases evaluated (AUGC): Xo = 2’O-methyl, Xf = 2’Fluoro, Xb = LNA, Xu = UNA-2’MOE, s = phosphorothioate, vp = vinylphosphonate.

    Figure 4

    Figure 4. (a) Plasma PK analysis of αhEGFR-siKRAS AOCs conjugated to the mAb cysteine, lysine, and Asn297 as measured by percent of injected dose (% ID) versus time (h). (b) Mouse plasma PK of DAR1 αhTfR1-siMstn AOCs generated via random cysteine versus cysteine engineered at position 188 of the light chain.

    Figure 5

    Figure 5. (a) Mouse plasma PK of AOCs (αhEGFR-Cys-MCC-siDMPK) comprised of αhEGFR mAb conjugated to one, two, or three siRNAs targeting DMPK mRNA. (b) Noncompartmental analysis of plasma PK data (0–96 h).

    Figure 6

    Figure 6. (a,b) Mouse PK of AOCs containing cleavable linkers.

    Figure 7

    Figure 7. (a–c) PK/PD studies with αmTfR1–siHprt AOCs containing four different noncleavable linkers.

    Figure 8

    Figure 8. (a,b) Liver KD analysis of αmASGPR–siHprt AOCs containing BisMal linkers to siRNA at the 5′ end and positions (pos) 8 or 14 of the sense strand at 96 h postdose.

    Figure 9

    Figure 9. (a) Expression of Hprt in muscle and (b) siRNA tissue concentration after treatment with αmTfR1–siHprt AOCs with MCC and VC linkers at various positions on the siHprt at 96 h postdose.

  • References


    This article references 51 other publications.

    1. 1
      Arslan, F. B.; Ozturk, K.; Calis, S. Antibody-Mediated Drug Delivery. Int. J. Pharm. 2021, 596, 120268,  DOI: 10.1016/j.ijpharm.2021.120268
    2. 2
      Awwad, S.; Angkawinitwong, U. Overview of Antibody Drug Delivery. Pharmaceutics 2018, 10 (3), 83,  DOI: 10.3390/pharmaceutics10030083
    3. 3
      Dovgan, I.; Koniev, O.; Kolodych, S.; Wagner, A. Antibody-Oligonucleotide Conjugates as Therapeutic, Imaging, and Detection Agents. Bioconjugate Chem. 2019, 30 (10), 24832501,  DOI: 10.1021/acs.bioconjchem.9b00306
    4. 4
      Ovacik, M.; Lin, K. Tutorial on Monoclonal Antibody Pharmacokinetics and Its Considerations in Early Development. Clin. Transl. Sci. 2018, 11 (6), 540552,  DOI: 10.1111/cts.12567
    5. 5
      Chery, J. RNA Therapeutics: RNAi and Antisense Mechanisms and Clinical Applications. Postdoc J. 2016, 4 (7), 3550,  DOI: 10.14304/SURYA.JPR.V4N7.5
    6. 6
      Levin, A. A. Treating Disease at the RNA Level with Oligonucleotides. N. Engl. J. Med. 2019, 380 (1), 5770,  DOI: 10.1056/NEJMra1705346
    7. 7
      Burnett, J. C.; Rossi, J. J. RNA-Based Therapeutics: Current Progress and Future Prospects. Chem. Biol. 2012, 19 (1), 6071,  DOI: 10.1016/j.chembiol.2011.12.008
    8. 8
      Crooke, S. T.; Witztum, J. L.; Bennett, C. F.; Baker, B. F. RNA-Targeted Therapeutics. Cell Metab. 2018, 27 (4), 714739,  DOI: 10.1016/j.cmet.2018.03.004
    9. 9
      Hu, B.; Zhong, L.; Weng, Y.; Peng, L.; Huang, Y.; Zhao, Y.; Liang, X.-J. Therapeutic siRNA: State of the Art. Signal Transduct. Target. Ther. 2020, 5 (1), 101,  DOI: 10.1038/s41392-020-0207-x
    10. 10
      Egli, M.; Manoharan, M. Chemistry, Structure and Function of Approved Oligonucleotide Therapeutics. Nucleic Acids Res. 2023, 51 (6), 25292573,  DOI: 10.1093/nar/gkad067
    11. 11
      Janas, M. M.; Schlegel, M. K.; Harbison, C. E.; Yilmaz, V. O.; Jiang, Y.; Parmar, R.; Zlatev, I.; Castoreno, A.; Xu, H.; Shulga-Morskaya, S.; Rajeev, K. G.; Manoharan, M.; Keirstead, N. D.; Maier, M. A.; Jadhav, V. Selection of GalNAc-Conjugated siRNAs with Limited off-Target-Driven Rat Hepatotoxicity. Nat. Commun. 2018, 9 (1), 723,  DOI: 10.1038/s41467-018-02989-4
    12. 12
      Brown, K. M.; Nair, J. K.; Janas, M. M.; Anglero-Rodriguez, Y. I.; Dang, L. T. H.; Peng, H.; Theile, C. S.; Castellanos-Rizaldos, E.; Brown, C.; Foster, D.; Kurz, J.; Allen, J.; Maganti, R.; Li, J.; Matsuda, S.; Stricos, M.; Chickering, T.; Jung, M.; Wassarman, K.; Rollins, J.; Woods, L.; Kelin, A.; Guenther, D. C.; Mobley, M. W.; Petrulis, J.; McDougall, R.; Racie, T.; Bombardier, J.; Cha, D.; Agarwal, S.; Johnson, L.; Jiang, Y.; Lentini, S.; Gilbert, J.; Nguyen, T.; Chigas, S.; LeBlanc, S.; Poreci, U.; Kasper, A.; Rogers, A. B.; Chong, S.; Davis, W.; Sutherland, J. E.; Castoreno, A.; Milstein, S.; Schlegel, M. K.; Zlatev, I.; Charisse, K.; Keating, M.; Manoharan, M.; Fitzgerald, K.; Wu, J.-T.; Maier, M. A.; Jadhav, V. Expanding RNAi Therapeutics to Extrahepatic Tissues with Lipophilic Conjugates. Nat. Biotechnol. 2022, 40 (10), 15001508,  DOI: 10.1038/s41587-022-01334-x
    13. 13
      Roberts, T. C.; Langer, R.; Wood, M. J. A. Advances in Oligonucleotide Drug Delivery. Nat. Rev. Drug Discovery 2020, 19 (10), 673694,  DOI: 10.1038/s41573-020-0075-7
    14. 14
      Cuellar, T. L.; Barnes, D.; Nelson, C.; Tanguay, J.; Yu, S.-F.; Wen, X.; Scales, S. J.; Gesch, J.; Davis, D.; van Brabant Smith, A.; Leake, D.; Vandlen, R.; Siebel, C. W. Systematic Evaluation of Antibody-Mediated siRNA Delivery Using an Industrial Platform of THIOMAB-siRNA Conjugates. Nucleic Acids Res. 2015, 43 (2), 11891203,  DOI: 10.1093/nar/gku1362
    15. 15
      Sela, T.; Mansø, M.; Siegel, M.; Marban-Doran, C.; Ducret, A.; Niewöhner, J.; Ravn, J.; Martin, R. E.; Sommer, A.; Lohmann, S.; Krippendorff, B.-F.; Ladefoged, M.; Indlekofer, A.; Quaiser, T.; Bueddefeld, F.; Koller, E.; Mohamed, M. Y.; Oelschlaegel, T.; Gothelf, K. V.; Hofer, K.; Schumacher, F. F. Diligent Design Enables Antibody-ASO Conjugates with Optimal Pharmacokinetic Properties. Bioconjugate Chem. 2023, 34 (11), 20962111,  DOI: 10.1021/acs.bioconjchem.3c00393
    16. 16
      Sugo, T.; Terada, M.; Oikawa, T.; Miyata, K.; Nishimura, S.; Kenjo, E.; Ogasawara-Shimizu, M.; Makita, Y.; Imaichi, S.; Murata, S.; Otake, K.; Kikuchi, K.; Teratani, M.; Masuda, Y.; Kamei, T.; Takagahara, S.; Ikeda, S.; Ohtaki, T.; Matsumoto, H. Development of Antibody-siRNA Conjugate Targeted to Cardiac and Skeletal Muscles. J. Controlled Release 2016, 237, 113,  DOI: 10.1016/j.jconrel.2016.06.036
    17. 17
      Malecova, B.; Burke, R. S.; Cochran, M.; Hood, M. D.; Johns, R.; Kovach, P. R.; Doppalapudi, V. R.; Erdogan, G.; Arias, J. D.; Darimont, B.; Miller, C. D.; Huang, H.; Geall, A.; Younis, H. S.; Levin, A. A. Targeted Tissue Delivery of RNA Therapeutics Using Antibody-Oligonucleotide Conjugates (AOCs). Nucleic Acids Res. 2023, 51 (12), 59015910,  DOI: 10.1093/nar/gkad415
    18. 18
      Cao, W.; Li, R.; Pei, X.; Chai, M.; Sun, L.; Huang, Y.; Wang, J.; Barth, S.; Yu, F.; He, H. Antibody-siRNA Conjugates (ARC): Emerging siRNA Drug Formulation. Med. Drug Discovery 2022, 15, 100128,  DOI: 10.1016/j.medidd.2022.100128
    19. 19
      Chernikov, I. V.; Vlassov, V. V.; Chernolovskaya, E. L. Current Development of siRNA Bioconjugates: From Research to the Clinic. Front. Pharmacol 2019, 10, 444,  DOI: 10.3389/fphar.2019.00444
    20. 20
      Tai, W. Current Aspects of siRNA Bioconjugate for In Vitro and In Vivo Delivery. Molecules 2019, 24 (12), 2211,  DOI: 10.3390/molecules24122211
    21. 21
      Sun, M. M. C.; Beam, K. S.; Cerveny, C. G.; Hamblett, K. J.; Blackmore, R. S.; Torgov, M. Y.; Handley, F. G. M.; Ihle, N. C.; Senter, P. D.; Alley, S. C. Reduction-Alkylation Strategies for the Modification of Specific Monoclonal Antibody Disulfides. Bioconjugate Chem. 2005, 16 (5), 12821290,  DOI: 10.1021/bc050201y
    22. 22
      Shen, B.-Q.; Xu, K.; Liu, L.; Raab, H.; Bhakta, S.; Kenrick, M.; Parsons-Reponte, K. L.; Tien, J.; Yu, S.-F.; Mai, E.; Li, D.; Tibbitts, J.; Baudys, J.; Saad, O. M.; Scales, S. J.; McDonald, P. J.; Hass, P. E.; Eigenbrot, C.; Nguyen, T.; Solis, W. A.; Fuji, R. N.; Flagella, K. M.; Patel, D.; Spencer, S. D.; Khawli, L. A.; Ebens, A.; Wong, W. L.; Vandlen, R.; Kaur, S.; Sliwkowski, M. X.; Scheller, R. H.; Polakis, P.; Junutula, J. R. Conjugation Site Modulates the in Vivo Stability and Therapeutic Activity of Antibody-Drug Conjugates. Nat. Biotechnol. 2012, 30 (2), 184189,  DOI: 10.1038/nbt.2108
    23. 23
      Zhou, Q.; Stefano, J. E.; Manning, C.; Kyazike, J.; Chen, B.; Gianolio, D. A.; Park, A.; Busch, M.; Bird, J.; Zheng, X.; Simonds-Mannes, H.; Kim, J.; Gregory, R. C.; Miller, R. J.; Brondyk, W. H.; Dhal, P. K.; Pan, C. Q. Site-Specific Antibody-Drug Conjugation through Glycoengineering. Bioconjugate Chem. 2014, 25 (3), 510520,  DOI: 10.1021/bc400505q
    24. 24
      Li, X.; Fang, T.; Boons, G. Preparation of Well-Defined Antibody-Drug Conjugates through Glycan Remodeling and Strain-Promoted Azide-Alkyne Cycloadditions. Angew. Chem., Int. Ed. 2014, 53 (28), 71797182,  DOI: 10.1002/anie.201402606
    25. 25
      Brun, M.-P.; Gauzy-Lazo, L. Protocols for Lysine Conjugation. In Antibody-Drug Conjugates; Ducry, L., Ed.; Methods in Molecular Biology; Humana Press: Totowa, NJ, 2013; Vol. 1045, pp 173187. DOI: 10.1007/978-1-62703-541-5_10 .
    26. 26
      Kumar, P.; Parmar, R. G.; Brown, C. R.; Willoughby, J. L. S.; Foster, D. J.; Babu, I. R.; Schofield, S.; Jadhav, V.; Charisse, K.; Nair, J. K.; Rajeev, K. G.; Maier, M. A.; Egli, M.; Manoharan, M. 5′-Morpholino Modification of the Sense Strand of an siRNA Makes It a More Effective Passenger. Chem. Commun. 2019, 55 (35), 51395142,  DOI: 10.1039/C9CC00977A
    27. 27
      Allerson, C. R.; Sioufi, N.; Jarres, R.; Prakash, T. P.; Naik, N.; Berdeja, A.; Wanders, L.; Griffey, R. H.; Swayze, E. E.; Bhat, B. Fully 2‘-Modified Oligonucleotide Duplexes with Improved in Vitro Potency and Stability Compared to Unmodified Small Interfering RNA. J. Med. Chem. 2005, 48 (4), 901904,  DOI: 10.1021/jm049167j
    28. 28
      Foster, D. J.; Brown, C. R.; Shaikh, S.; Trapp, C.; Schlegel, M. K.; Qian, K.; Sehgal, A.; Rajeev, K. G.; Jadhav, V.; Manoharan, M.; Kuchimanchi, S.; Maier, M. A.; Milstein, S. Advanced siRNA Designs Further Improve In Vivo Performance of GalNAc-siRNA Conjugates. Mol. Ther. 2018, 26 (3), 708717,  DOI: 10.1016/j.ymthe.2017.12.021
    29. 29
      Nair, J. K.; Willoughby, J. L. S.; Chan, A.; Charisse, K.; Alam, M. R.; Wang, Q.; Hoekstra, M.; Kandasamy, P.; Kel’in, A. V.; Milstein, S.; Taneja, N.; O’Shea, J.; Shaikh, S.; Zhang, L.; Van Der Sluis, R. J.; Jung, M. E.; Akinc, A.; Hutabarat, R.; Kuchimanchi, S.; Fitzgerald, K.; Zimmermann, T.; Van Berkel, T. J. C.; Maier, M. A.; Rajeev, K. G.; Manoharan, M. Multivalent N -Acetylgalactosamine-Conjugated siRNA Localizes in Hepatocytes and Elicits Robust RNAi-Mediated Gene Silencing. J. Am. Chem. Soc. 2014, 136 (49), 1695816961,  DOI: 10.1021/ja505986a
    30. 30
      Haraszti, R. A.; Roux, L.; Coles, A. H.; Turanov, A. A.; Alterman, J. F.; Echeverria, D.; Godinho, B. M. D. C.; Aronin, N.; Khvorova, A. 5′-Vinylphosphonate Improves Tissue Accumulation and Efficacy of Conjugated siRNAs in Vivo. Nucleic Acids Res. 2017, 45 (13), 75817592,  DOI: 10.1093/nar/gkx507
    31. 31
      Parmar, R.; Willoughby, J. L. S.; Liu, J.; Foster, D. J.; Brigham, B.; Theile, C. S.; Charisse, K.; Akinc, A.; Guidry, E.; Pei, Y.; Strapps, W.; Cancilla, M.; Stanton, M. G.; Rajeev, K. G.; Sepp-Lorenzino, L.; Manoharan, M.; Meyers, R.; Maier, M. A.; Jadhav, V. 5′-(E)-Vinylphosphonate: A Stable Phosphate Mimic Can Improve the RNAi Activity of siRNA-GalNAc Conjugates. ChemBioChem 2016, 17 (11), 985989,  DOI: 10.1002/cbic.201600130
    32. 32
      Brown, C. R.; Gupta, S.; Qin, J.; Racie, T.; He, G.; Lentini, S.; Malone, R.; Yu, M.; Matsuda, S.; Shulga-Morskaya, S.; Nair, A. V.; Theile, C. S.; Schmidt, K.; Shahraz, A.; Goel, V.; Parmar, R. G.; Zlatev, I.; Schlegel, M. K.; Nair, J. K.; Jayaraman, M.; Manoharan, M.; Brown, D.; Maier, M. A.; Jadhav, V. Investigating the Pharmacodynamic Durability of GalNAc-siRNA Conjugates. Nucleic Acids Res. 2020, 48 (21), 1182711844,  DOI: 10.1093/nar/gkaa670
    33. 33
      Strop, P.; Liu, S.-H.; Dorywalska, M.; Delaria, K.; Dushin, R. G.; Tran, T.-T.; Ho, W.-H.; Farias, S.; Casas, M. G.; Abdiche, Y.; Zhou, D.; Chandrasekaran, R.; Samain, C.; Loo, C.; Rossi, A.; Rickert, M.; Krimm, S.; Wong, T.; Chin, S. M.; Yu, J.; Dilley, J.; Chaparro-Riggers, J.; Filzen, G. F.; O’Donnell, C. J.; Wang, F.; Myers, J. S.; Pons, J.; Shelton, D. L.; Rajpal, A. Location Matters: Site of Conjugation Modulates Stability and Pharmacokinetics of Antibody Drug Conjugates. Chem. Biol. 2013, 20 (2), 161167,  DOI: 10.1016/j.chembiol.2013.01.010
    34. 34
      Miller, C. M.; Donner, A. J.; Blank, E. E.; Egger, A. W.; Kellar, B. M.; Østergaard, M. E.; Seth, P. P.; Harris, E. N. Stabilin-1 and Stabilin-2 Are Specific Receptors for the Cellular Internalization of Phosphorothioate-Modified Antisense Oligonucleotides (ASOs) in the Liver. Nucleic Acids Res. 2016, 44 (6), 27822794,  DOI: 10.1093/nar/gkw112
    35. 35
      Lehot, V.; Kuhn, I.; Nothisen, M.; Erb, S.; Kolodych, S.; Cianférani, S.; Chaubet, G.; Wagner, A. Non-Specific Interactions of Antibody-Oligonucleotide Conjugates with Living Cells. Sci. Rep. 2021, 11 (1), 5881,  DOI: 10.1038/s41598-021-85352-w
    36. 36
      Rocca, C.; Dennin, S.; Gu, Y.; Kim, J.; Chigas, S.; Najarian, D.; Chong, S.; Gutierrez, S.; Butler, J.; Charisse, K.; Robbie, G.; Xu, Y.; Brown, K. Evaluation of Electrophoretic Mobility Shift Assay as a Method to Determine Plasma Protein Binding of siRNA. Bioanalysis 2019, 11 (21), 19271939,  DOI: 10.4155/bio-2019-0151
    37. 37
      Liang, X.; Sun, H.; Shen, W.; Crooke, S. T. Identification and Characterization of Intracellular Proteins That Bind Oligonucleotides with Phosphorothioate Linkages. Nucleic Acids Res. 2015, 43 (5), 29272945,  DOI: 10.1093/nar/gkv143
    38. 38
      Gaus, H.; Miller, C. M.; Seth, P. P.; Harris, E. N. Structural Determinants for the Interactions of Chemically Modified Nucleic Acids with the Stabilin-2 Clearance Receptor. Biochemistry 2018, 57 (14), 20612064,  DOI: 10.1021/acs.biochem.8b00126
    39. 39
      Structure−Activity Relationship of Antibody−Oligonucleotide Conjugates: Evaluating Bioconjugation Strategies for Antibody−Phosphorodiamidate Morpholino Oligomer Conjugates for Drug Development, Manuscript in Preparation.
    40. 40
      Dosio, F.; Brusa, P.; Cattel, L. Immunotoxins and Anticancer Drug Conjugate Assemblies: The Role of the Linkage between Components. Toxins 2011, 3 (7), 848883,  DOI: 10.3390/toxins3070848
    41. 41
      Su, D.; Kozak, K. R.; Sadowsky, J.; Yu, S.-F.; Fourie-O’Donohue, A.; Nelson, C.; Vandlen, R.; Ohri, R.; Liu, L.; Ng, C.; He, J.; Davis, H.; Lau, J.; Del Rosario, G.; Cosino, E.; Cruz-Chuh, J. D.; Ma, Y.; Zhang, D.; Darwish, M.; Cai, W.; Chen, C.; Zhou, H.; Lu, J.; Liu, Y.; Kaur, S.; Xu, K.; Pillow, T. H. Modulating Antibody-Drug Conjugate Payload Metabolism by Conjugation Site and Linker Modification. Bioconjugate Chem. 2018, 29 (4), 11551167,  DOI: 10.1021/acs.bioconjchem.7b00785
    42. 42
      Su, D.; Zhang, D. Linker Design Impacts Antibody-Drug Conjugate Pharmacokinetics and Efficacy via Modulating the Stability and Payload Release Efficiency. Front. Pharmacol 2021, 12, 687926,  DOI: 10.3389/fphar.2021.687926
    43. 43
      Kolodych, S.; Koniev, O.; Baatarkhuu, Z.; Bonnefoy, J.-Y.; Debaene, F.; Cianférani, S.; Van Dorsselaer, A.; Wagner, A. CBTF: New Amine-to-Thiol Coupling Reagent for Preparation of Antibody Conjugates with Increased Plasma Stability. Bioconjugate Chem. 2015, 26 (2), 197200,  DOI: 10.1021/bc500610g
    44. 44
      Lyon, R. P.; Setter, J. R.; Bovee, T. D.; Doronina, S. O.; Hunter, J. H.; Anderson, M. E.; Balasubramanian, C. L.; Duniho, S. M.; Leiske, C. I.; Li, F.; Senter, P. D. Self-Hydrolyzing Maleimides Improve the Stability and Pharmacological Properties of Antibody-Drug Conjugates. Nat. Biotechnol. 2014, 32 (10), 10591062,  DOI: 10.1038/nbt.2968
    45. 45
      Meade, B. R.; Gogoi, K.; Hamil, A. S.; Palm-Apergi, C.; Berg, A. V. D.; Hagopian, J. C.; Springer, A. D.; Eguchi, A.; Kacsinta, A. D.; Dowdy, C. F.; Presente, A.; Lönn, P.; Kaulich, M.; Yoshioka, N.; Gros, E.; Cui, X.-S.; Dowdy, S. F. Efficient Delivery of RNAi Prodrugs Containing Reversible Charge-Neutralizing Phosphotriester Backbone Modifications. Nat. Biotechnol. 2014, 32 (12), 12561261,  DOI: 10.1038/nbt.3078
    46. 46
      Burke, P. J.; Hamilton, J. Z.; Jeffrey, S. C.; Hunter, J. H.; Doronina, S. O.; Okeley, N. M.; Miyamoto, J. B.; Anderson, M. E.; Stone, I. J.; Ulrich, M. L.; Simmons, J. K.; McKinney, E. E.; Senter, P. D.; Lyon, R. P. Optimization of a PEGylated Glucuronide-Monomethylauristatin E Linker for Antibody-Drug Conjugates. Mol. Cancer Ther. 2017, 16 (1), 116123,  DOI: 10.1158/1535-7163.MCT-16-0343
    47. 47
      Wiggins, B.; Liu-Shin, L.; Yamaguchi, H.; Ratnaswamy, G. Characterization of Cysteine-Linked Conjugation Profiles of Immunoglobulin G1 and Immunoglobulin G2 Antibody-Drug Conjugates. J. Pharm. Sci. 2015, 104 (4), 13621372,  DOI: 10.1002/jps.24338
    48. 48
      Prakash, T. P.; Lima, W. F.; Murray, H. M.; Li, W.; Kinberger, G. A.; Chappell, A. E.; Gaus, H.; Seth, P. P.; Bhat, B.; Crooke, S. T.; Swayze, E. E. Identification of Metabolically Stable 5′-Phosphate Analogs That Support Single-Stranded siRNA Activity. Nucleic Acids Res. 2015, 43 (6), 29933011,  DOI: 10.1093/nar/gkv162
    49. 49
      Green, M. R.; Sambrook, J. Quantification of RNA by Real-Time Reverse Transcription-Polymerase Chain Reaction (RT-PCR). Cold Spring Harb. Protoc. 2018, 2018 (10), pdb.prot095042,  DOI: 10.1101/pdb.prot095042
    50. 50
      Livak, K. J.; Schmittgen, T. D. Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2-ΔΔCT Method. Methods 2001, 25 (4), 402408,  DOI: 10.1006/meth.2001.1262
    51. 51
      Chen, C. Real-Time Quantification of microRNAs by Stem-Loop RT-PCR. Nucleic Acids Res. 2005, 33 (20), e179  DOI: 10.1093/nar/gni178
  • Supporting Information

    Supporting Information


    The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.4c00802.

    • Please see siRNA AOC Supplement. siRNA sequence and chemical modifications for evaluated test articles; siRNA conjugation comprehensive guide; noncompartmental analysis of plasma PK data in Figure 5b; plasma PK, tissue concentration, and mRNA expression of αmTfR1-Cys-MCC-siMstn conjugates; DAR1 versus DAR2 SEC comparison; in vitro stability of αhEGFR-siKRAS AOCs; KD and tissue concentration of αmTfR1-siHprt AOCs; plasma PK of αhEGFR-siKRAS AOCs; tumor delivery and KD of αhPSMA-siEGFR AOCs; PK of αmTfR1-siCtnnb1 AOCs; purification of antibody–siRNA conjugation reaction mixture by SAX; purification and characterization of DAR1 AOC; UNA vinylphosphonate (vpUq) amidite synthesis (PDF)


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.