Triantennary N-acetylgalactosamine (GalNAc) conjugation to small interfering RNA (siRNA) and antisense oligonucleotides (ASO) is today's leading strategy for efficient, selective hepatic delivery of oligonucleotide drugs. By exploiting the highly abundant asialoglycoprotein receptor (ASGPR) on hepatocytes, GalNAc conjugates transform systemic dosing into targeted hepatocyte uptake, enabling large increases in on-target potency, prolonged duration of action, and substantially lower clinical doses compared with non-targeted oligonucleotides. These properties have translated into multiple approvals and late-stage clinical successes, while chemistry refinements (2′-sugar modifications, phosphorothioate linkages, end-cap stabilizers, and seed-region design) have mitigated metabolic liability and off-target effects.
Figure 1. Targeted mRNA and protein knockdown via GalNAc-targeted delivery of siRNA and ASO[1].
Alfa Chemistry supports this platform with specialized GalNAc building blocks and custom GalNAc conjugate delivery services—please explore our GalNAc product list:
Or refer to our GalNAc-Conjugate Delivery Platform for custom synthesis options: GalNAc Conjugate-Based Delivery Systems
Biological rationale—ASGPR as a high-capacity hepatic gateway
ASGPR is a hepatocyte-enriched lectin that binds terminal galactose/GalNAc residues on desialylated glycoproteins. Expression is largely restricted to parenchymal hepatocytes, and it is both highly abundant and rapidly recycling: classical quantitative studies estimate roughly ~5×1010 copies per hepatocyte, enabling high-capacity uptake of multivalent GalNAc ligands presented on oligonucleotides. Trivalent (triantennary) GalNAc motifs exhibit high avidity for ASGPR and are the dominant ligand architecture used clinically. After receptor-mediated endocytosis, conjugates traffic to endosomes where ligand-receptor dissociation and ligand cleavage occur; the naked oligonucleotide must then escape endosomes by mechanisms that remain incompletely defined. The net result is efficient, hepatocyte-selective uptake and dose-sparing pharmacology.
Figure 2. (A) GalNAc conjugate structural nomenclature. (B) Schematic representation of the individual ligand elements aligning with trimeric carbohydrate recognition domains of ASGPR[2].
Chemical Strategies That Made GalNAc Conjugation Clinically Viable
Two sets of chemical advances were necessary: (A) oligonucleotide chemistries that provide nuclease resistance and favorable pharmacokinetics after systemic dosing, and (B) stable yet cleavable attachment of the GalNAc cluster so that receptor recognition and intracellular release are optimized.
Key oligonucleotide chemistries
- Phosphorothioate (PS) internucleotide linkages: replacement of non-bridging oxygen by sulfur increases nuclease resistance and plasma protein binding, improving circulation half-life and cellular uptake.
- 2′-ribose modifications (2′-O-methyl, 2′-fluoro, 2′-O-methoxyethyl, constrained nucleic acids like LNA/cEt): these boost duplex stability and exonuclease resistance and are selected depending on modality (siRNA vs. ASO) and mechanism (RNAi vs. RNase H recruitment).
- Terminal end-protection (e.g., 5′-vinylphosphonate or vinylphosphonate analogs): stabilizes siRNA ends against exonucleases and can obviate the need for cellular phosphorylation, improving potency and duration.
- Seed-region modulation (e.g., incorporation of glycerol nucleic acid (GNA) or selective 2′ patterning): reduces off-target seed hybridization risks while preserving on-target activity.
These combined chemistries collectively enabled second-generation siRNA and ASO constructs with dramatically lower clinical dose requirements and improved safety margins.
GalNAc linker design and cleavage
Trivalent GalNAc clusters are covalently tethered to oligonucleotides via linkers that balance serum stability and intracellular cleavability. Linker choice affects receptor binding, endosomal release, and the kinetics of ligand detachment; optimized linkers allow the GalNAc moiety to be cleaved or metabolized after endocytosis so that the payload can access the RNAi machinery or RNase H. Rational linker chemistry has been an important engineering variable in platform optimization.
Figure 3. The most common chemical modification scheme for GalNAc conjugates. X and Z are typically ethylene glycol or alkyl spacers. Y is a versatile moiety that allows for cluster branching[1].
Modality differences: GalNAc-siRNA vs GalNAc-ASO
- siRNA (RNAi): typically delivered as duplexes with extensive 2′-modification patterns and PS ends. GalNAc-siRNA conjugates rely on RISC loading of the guide strand after endosomal escape. Second-generation chemistries (ESC, ESC+) have extended duration and greatly reduced human dosing (examples below).
- ASO: many ASOs are gapmer designs (central DNA region flanked by modified wings) that recruit RNase H. PS content and wing chemistry are tailored to mechanism. GalNAc conjugation to ASOs substantially increases hepatocyte delivery versus naked ASO and has produced comparable or larger potency gains for RNase H gapmers.
Figure 4. GalNAc-mediated siRNA and ASO delivery to hepatocytes[3].
Preclinical and Emerging Clinical Indications
GalNAc conjugates are in development for a wide range of liver-centric and liver-leveraging indications:
- Viral hepatitis (HBV): ASO and siRNA GalNAc constructs against HBV transcripts have shown robust knockdown of viral proteins in preclinical models, and multiple clinical candidates are in the pipeline.
- Inherited metabolic and storage disorders: liver-expressed drivers of disease (e.g., GYS2 in glycogen storage disease) are tractable targets, and preclinical models have demonstrated biochemical and histologic improvements with GalNAc-siRNA.
- NAFLD/NASH and HCC: targets for steatosis, inflammation, fibrogenesis and oncogenesis (TAZ/STK25, PNPLA3 variants) have been evaluated in animal models with encouraging results, and GalNAc-conjugated molecules maintain activity even with partial ASGPR downregulation that is seen in advanced disease models.
- Systemic diseases via secreted hepatic proteins: by targeting liver production of circulating effectors (complement factors, coagulation factors, apolipoproteins), GalNAc conjugates can be used to treat extrahepatic diseases with infrequent subcutaneous dosing, an attractive benefit compared with intravenous biologics.
Clinical Translation—Approved Drugs and Late-Stage Research Examples
GalNAc-conjugated therapeutics have progressed from the bench to the clinic, resulting in multiple approved and late-stage investigational products.
Table 1: Clinical Applications of GalNAc-siRNA or GalNAc-ASO Conjugates[1].
| Clinical Phase | Modality | Drug Name | Target | Lead Indication |
| Registered | siRNA | Givlaari | d-aminolevulinate synthase 1 | acute hepatic porphyria |
| Submitted for registration | lumasiran | glycolate oxidase 1 | hyperoxaluria type 1 |
| inclisiran | PCSK9 | hypercholesterolemia |
| Phase 3 | fitusiran | antithrombin | hemophilia/bleeding disorders |
| vutrisiran | transthyretin | TTR amyloidosis |
| nedosiran | lactate dehydrogenase | primary hyperoxaluria |
| Phase 2 | cemdisiran | complement C5 | complement-mediated diseases |
| JNJ-3989(ARO-HBV) | HNV viral proteins | hepatitis B infection |
| Phase 1/2 | ALN-AAT02 | AAT | α1 liver disease |
| ALN-HBV02 | HBV viral proteins | hepatitis B virus infection |
| Phase 1 | AMG-890 (ARO-LPA) | lipoprotein(a) | cardiovascular disease |
| ALN-AGT | AGT | hypertension |
| SLN-124 | TMPRSS6 | β-thalassaemia and MDS |
| Phase 3 | ASO | AKCEA-TTR-LRx | transthyretin | TTR amyloidosis |
| AKCEA-APO(a)-LRx | apolipoprotein(a) | cardiovascular disease |
| Phase 2 | AKCEA-AOPCIII-LRx | apoC-III | cardiovascular disease |
| IONIS-GHR-LRx | growth hormone receptor | acromegaly |
| IONIS-TMPRSS6- LRx | transmembrane protease, serine 6 | β-thalassemia |
| IONIS-FB-LRx | complement factor B | Immunoglobulin A (IgA) neuropathy/age-related macular degeneration |
| IONIS-HBV-LRx | HBV viral proteins | hepatitis B infection |
| Phase 1 | IONIS-FX1-LRx | factor X1 | thrombosis |
| IONIS-AZ4-2.5LRx | not reported | cardiovascular disease |
| ION839 | not reported | NASH |
Safety Considerations and Mitigation Strategies
Oligonucleotide toxicity arises from on-target pharmacology, hybridization-dependent off-target effects (seed region), chemical modification-related toxicity, and tissue accumulation.
For GalNAc-siRNA, carefully designed seed regions and chemical patterns have been identified as primary means of mitigating hepatotoxic potential; extensive preclinical toxicology studies in rodents and nonhuman primates remain central to candidate screening. Histopathology occasionally reveals reversible findings (hepatocyte vacuolation, single-cell necrosis, macrophage/lymph node accumulation), which generally do not present adverse effects unless accompanied by clinical chemistry signals.
Immunogenicity with GalNAc-siRNA is uncommon, but antibody responses have been observed with some GalNAc-ASO programs, with limited pharmacokinetic impact. Lessons learned from Revuxilan (a first-generation program that was discontinued due to observed mortality imbalances) have led to more stringent exposure reductions through improved chemistry and rigorous safety screening. The net effect of modern designs is increased efficacy at lower systemic exposures, thereby improving the therapeutic window.
Figure 5. Models of GalNAc-bound single- and double-stranded ASOs. (A) A double-stranded ASO, with a GalNAc moiety at the 3' and 5' ends of each chain. (B) Single- and double-stranded ASOs bound to three GalNAc moieties. (C) Single-stranded ASO with two GalNAc moieties at one end or one GalNAc moiety at both ends. (D) Double-stranded ASO with one GalNAc moiety at each end or one GalNAc moiety at both ends, and (E) One GalNAc moiety bound to a single- or double-stranded ASO[4].
Practical Guidance for Drug Discovery Teams
- Start with multiple chemical series using orthogonal modification patterns; early in vitro and in vivo screens should include seed-off-target profiling and hepatocyte-selective distribution studies.
- Prioritize chemistries that minimize required human exposure—each reduction in administered mass reduces the risk associated with chemical- or accumulation-driven toxicity.
- Use validated triantennary GalNAc motifs and test two or more linker designs for each lead sequence.
- Incorporate nonclinical models reflective of clinical disease state where ASGPR expression may be reduced (advanced fibrosis/HCC) to assess retained potency.
- Consider reversibility strategies (e.g., antidote oligonucleotides or "Reversir"-type binders) for high-risk programs.
FAQs about Oligonucleotide Synthesis
1. What exactly is GalNAc? Why is it trivalent?
GalNAc is N-acetylgalactosamine; trivalent (triantennary) presentation maximizes affinity for ASGPR, improving receptor binding and hepatocyte uptake compared to monovalent presentation.
2. Are GalNAc conjugates effective in advanced liver diseases where ASGPR may be downregulated?
Partial downregulation of ASGPR may occur in advanced NASH or HCC, but preclinical data suggest that many GalNAc conjugates retain functional uptake and activity; this requires a target-by-target evaluation during lead compound screening.
3. Are GalNAc-conjugated oligonucleotides immunogenic?
GalNAc-siRNA has demonstrated low immunogenicity in clinical programs; some ASO chemistries rarely induce anti-drug antibodies. The immunogenicity of each drug candidate needs to be evaluated.
4. Can GalNAc be used to deliver other payloads besides siRNA/ASO?
Theoretically, yes—the GalNAc motif can be linked to other small molecules, peptides, or larger oligonucleotide scaffolds to bias hepatic uptake, but each payload requires customized chemistry and regulatory hurdles.
5. How do I begin collaborating on the synthesis of a GalNAc-conjugated lead compound?
Start with Alfa Chemistry's GalNAc building blocks and discuss your sequence, desired linker chemistry, and scale. Our team offers customized conjugation workflows and analytical services to accelerate lead development.
Related Products & Services
References
- Debacker AJ., et al. Delivery of Oligonucleotides to the Liver with GalNAc: From Research to Registered Therapeutic Drug. Mol Ther. 2022, 28(8), 1759-1771.
- Holland RJ., et al. Ligand Conjugate Structure Activity Relationships and Enhanced Endosomal Escape with a Targeted Polymer Micelle in Non-Human Primates. Molecular Therapy. 2021, 29(10).
- Cui H., et al. Liver-Targeted Delivery of Oligonucleotides with N-Acetylgalactosamine Conjugation. ACS Omega. 2021, 6(25), 16259-16265.
- Schmidt K., et al. Characterizing the effect of GalNAc and phosphorothioate backbone on binding of antisense oligonucleotides to the asialoglycoprotein receptor. Nucleic Acids Res. 2017, 45(5), 2294-2306.
Our products and services are for research use only and cannot be used for any clinical purposes.