Hand holding a tool near a DNA helix, symbolizing gene editing technology and emerging modalaties

Emerging modalities are not extensions of existing drug classes — they represent a new era of medicine defined by molecular precision, curative intent, and biological innovation. 

ImageFX (2025)

Emerging modalities in therapeutics: A comprehensive guide

Emerging gene, RNA, and cell therapies are transforming drug discovery with new precision tools that redefine treatment, regulation, and innovation.
| 8 min read
Register for free to listen to this article
Listen with Speechify
0:00
8:00

Introduction: The new frontier in drug discovery

The field of drug discovery is undergoing a profound transformation. Traditional small-molecule and biologic therapeutics — long the backbone of modern medicine — are now joined by a new generation of emerging modalities. These include gene and cell therapies, RNA-based drugs, protein degraders, and other cutting-edge approaches designed to target disease mechanisms once considered undruggable.

The rise of these modalities represents not only a scientific revolution but also a paradigm shift in how diseases are treated, pipelines are built, and regulatory frameworks evolve. For researchers, investors, and clinicians alike, understanding the science, promise, and challenges of these new modalities is essential to navigating the next era of therapeutics.

What are emerging modalities?

Emerging modalities are novel classes of therapeutics that extend beyond traditional chemical and biologic drugs. While small molecules generally modulate protein function through binding and biologics such as antibodies act through extracellular or immune-mediated pathways, emerging modalities often target the genome, transcriptome, or proteome in fundamentally new ways.

Continue reading below...
A stylized illustration of human kidneys drawn in white outlines, set against a blue background filled with colorful abstract flowers and leaves.
WebinarsUnlocking insights into rare kidney disease through genomic data
Large-scale clinicogenomic data sheds light on the biology of rare kidney disorders and opens doors to new treatment possibilities.
Read More

These therapies are typically characterized by:

  • Novel mechanisms of action: For example, degrading target proteins rather than merely inhibiting them.
  • Precision targeting: Modulating gene expression or editing the genome directly.
  • High specificity: Leveraging engineered nucleic acids or cellular components.
  • Personalization: Often tailored to an individual’s genetic or molecular profile.

Examples include gene and cell therapies, RNA therapeutics (mRNA, siRNA, antisense oligonucleotides), gene editing technologies (CRISPR/Cas9, base editors, prime editors), targeted protein degraders (PROTACs, molecular glues), and engineered proteins or peptides.

Why emerging modalities matter

These new modalities can address the 85% of the human proteome that remains “undruggable” by conventional means. Traditional therapeutics often fail when targets are intracellular, lack binding pockets, or involve complex protein-protein interactions. Emerging modalities overcome these limitations by acting at different biological levels — DNA, RNA, or via induced protein degradation.

From a clinical perspective, these therapies hold the promise of disease modification or even cures rather than symptomatic treatment. Gene therapy, for example, offers the potential to correct the root cause of genetic disorders, while mRNA technology has demonstrated its scalability and versatility during the COVID-19 pandemic.

From an R&D standpoint, emerging modalities expand the therapeutic toolkit and open new commercial and scientific frontiers. The global market for such modalities is growing exponentially, attracting unprecedented investment and reshaping the pipelines of nearly every major pharmaceutical company.

Continue reading below...
A 3D rendering of motor neurons lit up with blue, purple, orange, and green coloring showing synapses against a black background.
WhitepaperNew approaches to studying ALS
Learn how stem cell-derived motor neurons and microglia are opening new pathways to understand ALS and explore potential therapies.
Read More

Key categories of emerging modalities

1. Gene therapy

Infographic showing key categories of emerging modalities: Gene Therapy, RNA-Based Therapeutics, Protein Degradation Therapies, Gene Editing, Engineered Proteins and Peptides

Emerging modalities represent the next leap in therapeutic precision.

Gemini (2025)

Definition: Gene therapy involves introducing, removing, or altering genetic material within a patient’s cells to treat or prevent disease.

Mechanisms:

  • Gene addition or replacement: Delivering a functional gene to replace a defective one.
  • Gene silencing: Using vectors or RNA interference to suppress harmful gene expression.
  • Gene editing: Making precise alterations using tools such as CRISPR/Cas9.

Applications:

  • Inherited disorders (spinal muscular atrophy, hemophilia)
  • Oncology (CAR-T and oncolytic viral vectors)
  • Ophthalmology (retinal dystrophies)

Key advances:
Adeno-associated virus (AAV) vectors have become the leading delivery system, but challenges remain — especially in vector manufacturing, immune responses, and long-term durability of gene expression. Next-generation systems, including nonviral delivery and self-amplifying RNA, are under active development.

2. RNA-based therapeutics

Definition: RNA therapeutics manipulate RNA molecules to modulate gene expression, protein translation, or immune response.

Main classes:

  • mRNA therapies: Deliver genetic instructions to produce therapeutic proteins.
  • siRNA and miRNA: Silence specific mRNA targets to reduce pathogenic protein production.
  • Antisense oligonucleotides (ASOs): Bind RNA transcripts to modify splicing or translation.

Applications:
RNA drugs are now approved for conditions such as transthyretin amyloidosis, spinal muscular atrophy, and hypercholesterolemia.

Impact:
The success of mRNA COVID-19 vaccines (Moderna and Pfizer-BioNTech) validated the modality at scale, accelerating investment into mRNA-based cancer vaccines, rare disease therapies, and regenerative applications.

Challenges:
Instability, off-target effects, and delivery barriers — especially to nonliver tissues — remain significant. Lipid nanoparticles (LNPs) are the leading delivery vehicles, but new polymers, peptides, and extracellular vesicle systems are under investigation.

Continue reading below...
A conceptual illustration of a drug capsule filled with microchips, representing the integration of artificial intelligence in drug discovery and development
Technology GuidesA Technology Guide for AI-Enabled Drug Discovery
Learn practical strategies for using artificial intelligence to find the best drug candidate.
Read More

3. Protein degradation therapies

Definition: Protein degraders are small molecules or chimeras that harness the cell’s degradation machinery to eliminate disease-causing proteins.

Types:

  • PROTACs (proteolysis targeting chimeras): Bifunctional molecules linking a target protein to an E3 ligase for ubiquitin-mediated degradation.
  • Molecular glues: Small molecules that stabilize the interaction between a target and an E3 ligase.

Applications:
Effective against previously undruggable targets, especially transcription factors and scaffolding proteins.

Clinical outlook:
Multiple PROTAC candidates are in Phase 2 and 3 trials, addressing cancers, neurodegenerative diseases, and inflammatory conditions. Companies such as Arvinas, C4 Therapeutics, and Nurix are leading development efforts.

Challenges:
Pharmacokinetics and tissue distribution remain hurdles. Ensuring specificity without degrading homologous proteins is another challenge.

4. Cell-based therapies

Definition: Cell therapies involve administering live cells that are engineered or expanded to perform a therapeutic function.

Types:

  • Autologous therapies: Using the patient’s own cells (for example, CAR-T).
  • Allogeneic therapies: Universal donor-derived cells engineered for broader use.
  • Stem cell therapies: Using pluripotent or progenitor cells to regenerate tissue.

Applications:
CAR-T therapies (Kymriah and Yescarta) revolutionized oncology by redirecting T cells to target tumors. Meanwhile, stem-cell-based treatments are advancing for diabetes, Parkinson’s disease, and cardiac repair.

Challenges:
Manufacturing complexity, cost, and regulatory oversight make scaling difficult. Emerging solutions include automated cell manufacturing and gene-edited universal donor lines to reduce rejection risk.

5. Gene editing

Definition: Gene editing technologies such as CRISPR/Cas9, TALENs, and zinc finger nucleases allow direct modification of DNA sequences.

Continue reading below...
A 3D illustration of two DNA strands in a transparent bubble
EbooksOvercoming barriers in gene therapy
Advanced gene editing, delivery, and analytical tools are driving better gene therapies.
Read More

Applications:

  • Correcting pathogenic mutations (sickle cell disease, beta thalassemia)
  • Enhancing cell therapies (CAR-T optimization)
  • Functional genomic screening for target discovery

Recent breakthroughs:
The 2023 FDA approval of Casgevy (exagamglogene autotemcel) marked the first CRISPR-based therapy approved for clinical use, treating sickle cell disease — a milestone that validated gene editing as a viable therapeutic platform.

Next steps:
Base and prime editing technologies aim for more precise, less disruptive DNA modifications, broadening the potential therapeutic landscape.


6. Engineered proteins and peptides

Definition: Engineered proteins, peptides, and scaffolds expand therapeutic possibilities through enhanced stability, selectivity, and delivery.

Examples:

  • Bispecific antibodies: Target two antigens simultaneously.
  • Antibody-drug conjugates (ADCs): Combine antibody targeting with cytotoxic payloads.
  • Peptidomimetics: Small peptides engineered for improved pharmacokinetics.

Market growth:
The ADC market alone is projected to exceed $25 billion by 2030, driven by oncology indications and improved linker technologies.

Emerging modalities: Market outlook and investment trends

The emerging modalities market is growing at an extraordinary pace. Analysts project that by 2030, nearly one-third of all new drug approvals will derive from advanced modalities.

Key figures (2025 estimates):

  • Global gene therapy market: $20–25 billion
  • RNA therapeutics market: $15–20 billion
  • Cell therapy market: $30–40 billion
  • Protein degradation and gene editing combined: $10–15 billion

Pharmaceutical pipelines now feature more than 2,000 active programs in advanced modalities, with a surge in preclinical RNA and gene editing assets. Major pharmaceutical companies — including Novartis, Roche, Pfizer, and AstraZeneca — have restructured their R&D divisions to prioritize these technologies, while smaller biotechs continue to drive innovation.

Continue reading below...
An illustration showing a DNA strand and scientists removing segments with tweezers, representing the CRISPR gene editing technology
Technology GuidesA Technology Guide for CRISPR Screening
Emerging CRISPR screening methods are shaping what’s possible in drug development and precision medicine.
Read More

Strategic investment partnerships such as Roche’s investment in GenEdit signal the industry’s commitment to gene editing and delivery platforms.

Scientific promise and therapeutic potential

Emerging modalities represent the next leap in therapeutic precision. By targeting diseases at their molecular root, these technologies have the potential to cure rather than manage chronic conditions.

Areas of promise:

  • Genetic disorders: Single-dose gene replacement or editing therapies.
  • Oncology: Immune-based therapies and targeted degradation for undruggable drivers.
  • Neurological diseases: RNA and protein-based tools to address complex CNS pathologies.
  • Infectious diseases: mRNA and siRNA therapeutics for rapid response.

The expansion of multiomic data, AI-driven design, and automation has accelerated discovery, enabling predictive modeling of RNA structures, guide RNA specificity, and degradation kinetics — all essential to optimizing next-generation drugs.

Manufacturing and scalability challenges

Despite their promise, emerging modalities face significant chemistry, manufacturing, and controls (CMC) challenges. Many are biologically complex, personalized, or require stringent cold-chain logistics.

Key challenges:

  • Vector and LNP production: High cost and batch variability.
  • Analytical characterization: New quality control frameworks are needed to ensure potency and safety.
  • Regulatory harmonization: Diverse global standards complicate approvals.
  • Automation and AI: Increasingly essential for process standardization and cost reduction.

The industry is moving toward modular, closed-system manufacturing to scale therapies safely and efficiently.

Regulatory and clinical landscape

Regulatory agencies worldwide are adapting rapidly. The U.S. Food and Drug Administration (FDA), European Medicines Agency (EMA), and Japan’s Pharmaceuticals and Medical Devices Agency (PMDA) have all issued frameworks for advanced therapy medicinal products (ATMPs) and novel drug modalities.

Continue reading below...
A compass on a nautical map
InfographicsCharting a cellular treasure map with spatial transcriptomics
Spatial transcriptomics technologies unveil a goldmine of biological information.
Read More

Recent developments:

Uncertainty remains in areas such as off-target editing assessment, long-term follow-up, and immunogenicity testing. Collaboration between regulators, sponsors, and industry consortia is essential for timely, safe approvals.


Ethical, safety, and societal considerations

With immense potential comes equal ethical responsibility. Emerging modalities challenge long-standing norms about what medicine should do. Gene editing, cellular reprogramming, and AI-assisted design raise questions about human intervention in biology, data governance, and equitable access.

Germline modification and heritable change

The most significant ethical debate centers on germline gene editing — altering DNA in reproductive cells or embryos in ways that can be inherited. Somatic editing (nonheritable) is accepted for severe diseases, but germline modification remains prohibited in most countries.

The 2018 case of He Jiankui, who announced the birth of CRISPR-edited babies in China, reinforced the global consensus that scientific capability outpaces ethical readiness. In response, the World Health Organization (WHO) and the International Commission on the Clinical Use of Human Germline Genome Editing established governance frameworks emphasizing scientific justification, transparency, and long-term follow-up.

Continue reading below...
A gloved researcher’s hand uses tweezers to place a biological sample into a storage box for preservation.
InfographicsBanking on biology
Biobanking gives scientists access to thousands of biological samples, moving precision medicine one step closer to reality.
Read More

Equitable access and global health disparities

Advanced therapies can cost more than $2 million per treatment, raising issues of affordability and access. Without deliberate policy interventions, they risk widening global health inequities. Initiatives like the WHO Global Observatory on Health R&D and the Bespoke Gene Therapy Consortium are developing open-access vector platforms and shared manufacturing infrastructure to reduce barriers.

Value-based and outcomes-based pricing models, tying payment to long-term patient outcomes, are also emerging as potential solutions.

Data ownership, privacy, and algorithmic bias

Emerging modalities rely on genomic and phenotypic data from global biobanks. As AI becomes central to designing CRISPR guides and RNA sequences, questions of ownership, consent, and bias intensify.

Frameworks such as the EU General Data Protection Regulation (GDPR) and the U.S. NIH Genomic Data Sharing Policy have addressed some issues, but global harmonization is lacking. Federated learning — training AI on distributed datasets without exposing personal data — offers a promising model.

Informed consent and long-term follow-up

Long-acting therapies challenge traditional informed consent. Patients must understand not only immediate risks but also long-term uncertainties, including off-target effects and durability. Regulators now require postmarket surveillance lasting up to 15 years for gene therapy recipients, supported by patient registries and real-world evidence frameworks.

Continue reading below...
An illustration of a DNA double helix with a part of the helix colored red
MilestonesScience Milestone: The evolution of gene therapy
Gene therapy has a complicated history, marked by many ups and downs and crafted by ever-advancing technologies.
Read More

Public trust and ethical governance

Public trust underpins the success of emerging modalities. Transparent communication of risks and benefits and inclusive policymaking are essential. The National Academies of Sciences, Engineering, and Medicine and the WHO’s Expert Advisory Committee advocate for multistakeholder governance, involving ethicists, scientists, and patients.

Ethical innovation is not a constraint on progress — it is the foundation of responsible scientific advancement.

The future of emerging modalities

Over the next decade, emerging modalities will converge. Hybrid approaches — such as RNA-guided protein degraders, gene-edited cell therapies, and mRNA-encoded antibodies — show that traditional modality boundaries are dissolving.

Integrated example: AI-driven nanoparticle engineering meets multiomics in precision oncology

A recent study by Sheikh and Jirvankar et al. (2024) demonstrated how AI, multiomics data, and nanoparticle design can be integrated to create personalized cancer therapeutics

Researchers used AI models trained on tumor-specific genomic, transcriptomic, and proteomic profiles to predict optimal nanoparticle formulations for drug delivery. The models identified receptor patterns unique to tumor subtypes, guiding ligand functionalization and surface charge optimization for targeted uptake.

Continue reading below...
An illustration of genetic engineering with CRISPR/Cas9, a powerful tool for gene editing that may cause off-target effects if used without molecular safety switches.
InfographicsSafety buttons for CRISPR/Cas9
New technologies keep CRISPR/Cas9 in check and minimize off-target risks.
Read More

Preclinical testing confirmed that AI-optimized nanoparticles achieved superior tumor penetration and reduced systemic toxicity compared with controls. This integration of AI, nanotechnology, and omics-based profiling exemplifies how computational, molecular, and materials science can converge to yield more precise, effective treatments.

Conclusion: The modalities that will define the future

Emerging modalities are not extensions of existing drug classes — they represent a new era of medicine defined by molecular precision, curative intent, and biological innovation. From CRISPR cures to programmable RNA vaccines, these technologies are reshaping human health.

As clinical successes accumulate and regulatory frameworks mature, the industry will continue to shift toward modality-agnostic platforms — focusing less on the molecule and more on the mechanism. For scientists and developers, the challenge is clear: harness the power of biology itself to transform medicine.

This article was created with the assistance of Generative AI and has undergone editorial review before publishing.

About the Author

  • Trevor Henderson is the Creative Services Director for the Laboratory Products Group at LabX Media Group. With over two decades of experience, he specializes in scientific and technical writing, editing, and content creation. His academic background includes training in human biology, physical anthropology, and community health. Since 2013, he has been developing content to engage and inform scientists and laboratorians.

Related Topics

Loading Next Article...
Loading Next Article...
Subscribe to Newsletter

Subscribe to our eNewsletters

Stay connected with all of the latest from Drug Discovery News.

Subscribe

Sponsored

Portrait of Scott Weitze, Vice President of Research and Technical Standards at My Green Lab, beside text that reads “Tell us what you know: Bringing sustainability into scientific research,” with the My Green Lab logo.
Laboratories account for a surprising share of global emissions and plastic waste, making sustainability a priority for modern research.
A 3D rendering of red and yellow protein molecules floating in a fluid-like environment.
Discover approaches that shorten the path from DNA constructs to purified, functional proteins.
A 3D rendering of two DNA double helices in different colors, representing genetic diversity or molecular comparison on a light background.
By replacing conventional plasmid systems, cell-free DNA synthesis improves speed and quality in mRNA research.
Drug Discovery News September 2025 Issue
Latest IssueVolume 21 • Issue 3 • September 2025

September 2025

September 2025 Issue

Explore this issue