Phosphorodiamidate Morpholino Oligomer Development Platform
Introduction
Phosphorodiamidate Morpholino Oligomers (PMOs) are cutting-edge nucleic acid therapeutics with high stability, low immunogenicity, and precise RNA binding—used for genetic disorders, viral infections, and cancer treatment via targeted gene regulation.
Creative Biolabs, a global leader in nucleic acid therapy, has more than 20 years of PMO expertise. Its platform offers tailored solutions (design to IND-enabling studies) and supports 500+ clients to advance PMO candidates to preclinical/clinical stages.
Based on the unique mechanism of PMO (RNA base-pairing without activating RNase H, avoiding off-target RNA degradation) and latest clinical research insights, our PMO Development Platform provides nucleic acid-based therapeutic development services as follows:
Targeted Gene Expression Regulation
PMOs bind specifically to complementary mRNA sequences, enabling precise control of gene expression—either suppressing aberrant gene products (e.g., oncogenes like MYC in solid tumors) or restoring functional protein synthesis (e.g., correcting mis-splicing in spinal muscular atrophy). At Creative Biolabs, we optimize PMO sequence design via in silico modeling (predicting target affinity and off-target binding) and in vitro screening (using luciferase reporter assays). Our platform supports the synthesis of modified PMOs (e.g., conjugated with cell-penetrating peptides like CPP-PMO for enhanced cellular uptake) and offers validation via qPCR (mRNA level) and Western blot (protein level) to confirm regulation efficiency.
Fig.1 The mechanism by which self-transfected MMO-PMO chimeras targeting Nanog achieve gene silencing in vitro.1,3
Exon Skipping for Genetic Disorder Therapy
Exon skipping serves as a key application of PMOs in treating genetic disorders driven by frameshift mutations—such as Duchenne muscular dystrophy (DMD), where mutations in the dystrophin gene disrupt normal protein synthesis. By binding to splice sites or exonic splicing enhancers, PMOs redirect mRNA splicing to exclude mutated exons, ultimately generating truncated yet functional proteins; in DMD, this process produces mini-dystrophin, a shortened form of the protein that retains critical functionality.
We synthesize custom PMOs tailored to patient-specific mutations, including those designed for exon 51 skipping in DMD cases. We also conduct preclinical testing using disease-relevant models like mdx mice and human induced pluripotent stem cell (iPSC)-derived myotubes, alongside efficacy assessments—these include detecting dystrophin protein via immunofluorescence and measuring functional improvements through tests like grip strength. To align with regulatory standards, we further perform safety evaluations covering cytotoxicity, immunogenicity, and analysis of off-target splicing effects.
Fig.2 Exon skipping was performed using PMO, where c.d represents the structure of dystrophin after skipping.2,3
Antisense-Mediated Pathogen Inhibition
PMOs combat viral/bacterial infections by targeting pathogen-specific RNA (e.g., viral replication regions, bacterial toxin-encoding mRNA). They have low drug resistance risk, thanks to binding conserved pathogen RNA sequences.
Creative Biolabs designs PMOs for pathogens like influenza (M2 gene), Zika (NS5 polymerase), and Staphylococcus aureus (alpha-toxin mRNA). We validate activity via in vitro assays and boost stability in biological fluids with modifications like phosphorothioate backbones.
In Vivo Delivery Optimization
A critical challenge in PMO therapy is improving in vivo delivery—overcoming cell membrane barriers and achieving tissue-specific targeting. Our PMO Development Platform addresses this via integrated delivery technology:
- Ligand Conjugation: Conjugating PMOs with tissue-specific ligands (e.g., GalNAc for liver targeting, transferrin for muscle targeting) to enhance receptor-mediated uptake.
- Nanocarrier Formulation: Formulating PMOs with lipid nanoparticles (LNPs), polymeric micelles, or inorganic nanoparticles to protect against degradation and boost cellular penetration.
- Preclinical PK/PD Studies: Conducting pharmacokinetic (half-life, bioavailability) and pharmacodynamic (target engagement, protein expression) studies in animal models (mice, rats, non-human primates) to optimize delivery strategies and validate therapeutic efficacy.
What We Can Offer
End-to-End Expertise
Covering the entire PMO development lifecycle—from sequence design, synthesis, and modification to preclinical validation (efficacy, safety, PK/PD) and IND-enabling studies. No need for multiple vendors; we streamline your workflow.
Disease-Specific Experience
Deep expertise in PMO applications across therapeutic areas—we've supported DMD exon-skipping programs, antiviral PMO development, and cancer oncogene suppression projects, with a track record of accelerating candidates to clinic.
Flexible Customization
Offering tailored solutions for unique project needs—whether you need a single service (e.g., PMO synthesis) or a full turnkey program (design to preclinical validation). Our team of 100+ scientists collaborates closely with you to define optimal strategies.
Rapid Turnaround
Standard PMO synthesis (up to 20-mer) takes 5-7 business days; preclinical validation assays (e.g., in vitro efficacy) are completed within 2-3 weeks. We prioritize speed to help you meet development milestones.
FAQs
Q1: What is the minimum length of PMOs you can synthesize, and what modifications are available?
A: We synthesize PMOs ranging from 8-mer (short antisense sequences) to 40-mer (for complex splice modulation). Available modifications include CPP conjugation (e.g., R9, Tat), ligand conjugation (GalNAc, transferrin), fluorescent labeling (FITC, Cy5), and backbone modifications (phosphorothioate for enhanced stability).
Q2: How do you ensure the safety of PMOs, especially regarding off-target effects?
A: We use a two-step safety assessment: (1) In silico screening via our proprietary suite to predict off-target mRNA binding and exclude high-risk sequences; (2) In vitro validation (e.g., RNA-seq to analyze global gene expression, cytotoxicity assays in primary cells) to confirm no unintended effects. For in vivo studies, we monitor organ toxicity (liver, kidney) and immunogenicity (cytokine levels).
Q3: What animal models do you use for PMO in vivo testing?
A: We select models based on your therapeutic target: (1) Genetic disease models (mdx mice for DMD, SMNΔ7 mice for SMA); (2) Infectious disease models (influenza-infected mice, Zika-infected mice); (3) Cancer models (xenografts with human tumor cells). We also offer non-human primate studies for late-stage preclinical validation.
Q4: How long does a full PMO development program (design to preclinical validation) take?
A: Timeline depends on project scope: a standard program (sequence design - synthesis - in vitro efficacy - in vivo PK/PD) takes 12–16 weeks. For IND-enabling studies (including safety toxicology), the timeline is 6–8 months. We provide a detailed project plan with milestones upon initiation.
Contact Our Team for More Information and to Discuss Your Project
Creative Biolabs is dedicated to advancing PMO-based therapeutics by combining technical excellence, flexible solutions, and rapid turnaround. Whether you're at the early research stage or preparing for clinical trials, our PMO Development Platform can de-risk your program and accelerate time-to-clinic.
References
- Das, Ujjal, et al. "Self-transfecting GMO-PMO chimera targeting Nanog enable gene silencing in vitro and suppresses tumor growth in 4T1 allografts in mouse." Molecular Therapy Nucleic Acids 32 (2023): 203-228. https://doi.org/10.1016/j.omtn.2023.03.011
- Lee, Joshua, et al. "Antisense PMO cocktails effectively skip dystrophin exons 45-55 in myotubes transdifferentiated from DMD patient fibroblasts." PLoS One 13.5 (2018): e0197084. https://doi.org/10.1371/journal.pone.0197084.
- Distributed under Open Access license CC BY 4.0, without modification.