Kasturi Shikshan Sanstha College of pharmacy, Shikrapur, Tal. Shirur, Dist. Pune 412208
Gene therapy represents a paradigm-shifting therapeutic modality that addresses diseases at their genetic foundation by modifying, replacing, or supplementing defective genes within patient cells. This comprehensive review examines the current status and future perspectives of gene therapy, synthesizing developments from molecular mechanisms to clinical applications and market dynamics. The field has progressed dramatically from theoretical concept to clinical reality, marked by the 2023 FDA approval of Casgevy, the first CRISPR-based therapy for sickle cell disease, demonstrating 93.5?ficacy in eliminating severe vaso-occlusive crises As of 2025, approximately 250 gene therapy clinical trials are active worldwide, encompassing diverse therapeutic areas including hemoglobinopathies, cancers, metabolic disorders, autoimmune diseases, viral infections, and neurological conditions[3]. The global market has expanded from USD 9.5 billion in 2024 to an anticipated USD 11.4 billion in 2025, with projections reaching USD 58.87 billion by 2034, reflecting a robust 20% compound annual growth rate[1]. This exponential growth is driven by technological innovations in vector design, genome editing platforms (CRISPR-Cas9, base editors, prime editors), manufacturing processes, and regulatory frameworks facilitating accelerated approvals. Gene therapy employs two fundamental approaches: in vivo therapy, where therapeutic genes are delivered directly into patient tissues, and ex vivo therapy, where patient cells are genetically modified outside the body and subsequently reinfused. Viral vectors including adeno-associated virus (AAV), lentivirus, adenovirus, and retrovirus serve as the predominant delivery vehicles, each offering distinct advantages regarding cargo capacity (4.7 kb for AAV to 30 kb for HSV), tropism, immunogenicity, and integration properties[7]. Non-viral delivery methods utilizing lipid nanoparticles, electroporation, and naked DNA are emerging as safer alternatives with reduced immunogenicity. Future perspectives indicate gene therapy's integration with personalized medicine, where treatments are tailored to individual genetic profiles, and expansion into common chronic diseases beyond rare genetic disorders. Emerging technologies including base editing (single nucleotide changes without double-strand breaks), prime editing (precise insertions and deletions), RNA editing, and epigenetic editing offer enhanced precision and reduced safety risks[8]. Innovations in manufacturing, such as point-of-care production devices enabling on- demand personalized nanomedicine preparation, promise to democratize access and reduce costs[15]. The convergence of gene therapy with artificial intelligence for predictive modeling, nanotechnology for improved delivery, and multi-omics for patient stratification will likely transform gene therapy from specialized interventions into mainstream therapeutic options, fundamentally reshaping pharmaceutical care in the coming decades.
Gene therapy represents one of the most revolutionary concepts in modern medicine: the ability to treat or cure diseases by directly modifying the genetic instructions within human cells. The conceptual foundation emerged in the 1960s when researchers first proposed that introducing functional genes into cells could compensate for defective ones [11]. However, the practical realization of this vision required decades of advancement in molecular biology, virology, and genetic engineering.
Fundamental Principles: Gene therapy operates on the principle that many diseases result from genetic abnormalities— either inherited mutations or acquired genetic changes—that can be addressed by introducing, removing, or modifying genetic materials.
Gene Addition/Augmentation: Introducing a functional copy of a gene into cells that either lack the gene or possess a defective version. This approach is particularly effective for recessive genetic disorders where even partial restoration of gene function provides therapeutic benefit
Gene Editing: Directly correcting mutations in the genome using programmable nucleases such as CRISPR-Cas9, TALENs (transcription activator-like effector nucleases), or zinc finger nucleases. This approach offers the potential for permanent correction of the underlying genetic defect.
Gene Silencing/Inhibition: Suppressing the expression of disease-causing genes, particularly useful for dominant genetic disorders, infectious diseases (viral genes), and cancers (oncogenes). Techniques include RNA interference (RNAi), antisense oligonucleotides, and CRISPR-based transcriptional repression.
Gene Killing: Introducing "suicide genes" into specific cell populations (typically cancer cells) that render them susceptible to particular drugs, enabling selective destruction of diseased cells.
Delivery Systems and Vectors
The success of gene therapy critically depends on efficient delivery of therapeutic genetic material to target cells. Delivery systems are broadly categorized as viral or non-viral vectors [10].
Viral Vectors: Viruses have evolved sophisticated mechanisms for entering cells and delivering genetic material, making them ideal gene therapy vehicles when properly modified [7]. Key viral vectors include:
Retroviral Vectors: These integrate into the host genome but only transduce dividing cells. Early-generation gamma-retroviral vectors caused insertional mutagenesis leading to leukemia in several clinical trials, significantly impacting their current usage.
Non-Viral Vectors: These systems offer advantages including lower immunogenicity, easier large- scale production, and potential for repeated administration Major non-viral approaches include:
Physical Delivery Methods: Include microinjection, gene gun (particle bombardment), and hydrodynamic injection, primarily used in research settings
Current Regulatory Landscape
Gene therapies are regulated as biological products by agencies including the U.S. Food and Drug Administration (FDA), European Medicines Agency (EMA), and equivalent bodies worldwide [37]. The regulatory framework encompasses:
Preclinical Development: Extensive laboratory and animal studies demonstrating proof-of- concept, safety, and preliminary efficacy.
Clinical Trials: Phase I (safety and dosing), Phase II (efficacy and side effects), and Phase III (comparative effectiveness) studies following Good Clinical Practice guidelines
Manufacturing Standards: Good Manufacturing Practice (GMP) compliance for vector production and cell processing
Long-term Follow-up: Extended safety monitoring (often 15 years) to detect delayed adverse events including insertional mutagenesis or immune reactions.
The CRISPR Revolution
The development of CRISPR-Cas9 technology represents a watershed moment in gene therapy history [22]. CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) systems enable precise genome editing through RNAguided DNA cleavage. The Cas9 protein, directed by a guide RNA complementary to the target DNA sequence, creates double-strand breaks at specified genomic locations. Cellular repair mechanisms then correct or disrupts the targeted gene. The transformative FDA approval of Casgevy (exagamglogene autotemcel) in December 2023 marked CRISPR's clinical validation [2]. This ex vivo therapy treats sickle cell disease by editing patients' hematopoietic stem cells to reactivate fetal hemoglobin production, compensating for defective adult hemoglobin. Clinical trial data demonstrated that 93.5% of patients (29 of 31 evaluable) achieved freedom from severe vaso-occlusive crises for at least 12 consecutive months, with all patients achieving successful engraftment.
Current Status of Gene Therapy
FDA-Approved Gene Therapies (2016-2025)
The gene therapy clinical pipeline demonstrates remarkable breadth and depth. As of February 2025, approximately 250 CRISPR-based clinical trials are registered globally, with more than 150 trials actively enrolling and treating patients
Geographic Distribution and Market Dynamics
North America maintains dominance in the gene therapy market, capturing 45% of global market share in 2024 and projected to hold 65% by 2035[1] [51]. This leadership reflects:
Technical and Scientific Challenges
Immunogenicity and Immune Responses: One of the most significant obstacles facing gene therapy is the host immune response to both viral vectors and transgene products [58]. Pre-existing neutralizing antibodies to AAV serotypes (present in 30-60% of population due to natural AAV exposure) can neutralize therapeutic vectors before transduction occurs [^59]. Additionally, innate immune recognition of viral capsids triggers complements activation, cytokine release, and hepatotoxicity, while adaptive immunity against capsid antigens can eliminate transduced cells, diminishing therapeutic durability [^60]. The immune response to transgene products poses particular challenges for patients who completely lack the normal protein (null mutations), as their immune systems may recognize the therapeutic protein as foreign [^61]. Immunosuppression protocols mitigate these responses but introduce risks including opportunistic infections and malignancies.
Data Privacy and Genetic Discrimination: Gene therapy involves collecting extensive genetic information, raising privacy concerns [^74]:
Use of genetic information in biobanks and research
Regulatory Challenges
Evolving Regulatory Frameworks: Gene therapy technologies evolve faster than regulatory frameworks can adapt [^75]:
Global Regulatory Harmonization: Different regulatory standards across jurisdictions create challenges [^76]:
Delays in global access due to sequential approval processes
Long-Term Safety Monitoring: The need for extended follow-up (15+ years) poses practical challenges.
Future Perspectives
Integration with Personalized Medicine
Gene therapy represents the ultimate manifestation of personalized medicine—treatments tailored to individual genetic profiles [5]. The convergence of gene therapy with precision medicine will accelerate through several mechanisms:
Pharmacogenomics Integration: Understanding how genetic variations affect drug metabolism and response will guide gene therapy candidate selection and dosing optimization [^78]. Patients with specific genetic variants affecting vector metabolism, immune responses, or cellular uptake may require modified treatment protocols.
Patient Stratification: Advanced diagnostics including whole genome sequencing, transcriptomics, proteomics, and metabolomics will identify which patients most likely benefit from gene therapy versus alternative treatments.
Biomarker panels may predict:
Likelihood of response to specific gene therapy approaches Risk of adverse immune reactions Optimal vector serotype selection
Timing for therapeutic intervention
Individualized Vector Selection: Rather than one-size-fits-all approaches, future gene therapies may utilize patient specific vector engineering. Directed evolution techniques can generate AAV variants optimized for individual patient tissue tropism patterns [^80].
Adaptive Trial Designs: Response-adaptive randomization and basket trials evaluating therapies across genetic subtypes will accelerate personalized gene therapy development. The convergence of technological innovation, manufacturing advancement, regulatory evolution, and ethical maturation will determine whether gene therapy fulfills its transformative potential or remains a premium intervention accessible only to privileged populations. The next decade will be decisive in establishing gene therapy's role in global healthcare systems, potentially transitioning from experimental treatments for rare conditions to mainstream therapeutic modalities fundamentally reshaping pharmaceutical care and improving human health across all populations.
CONCLUSION:
Gene therapy has evolved from a visionary concept to clinical reality, representing one of the most transformative developments in pharmaceutical sciences and modern medicine. This comprehensive review has examined gene therapy's journey from pioneering trials in the 1990s through the revolutionary CRISPR era to its current status as an established therapeutic modality with multiple FDA-approved products and a robust clinical pipeline encompassing over 250 active trials worldwide [3]. The field's maturation is evidenced by landmark achievements including the December 2023 FDA approval of Casgevy, the first CRISPR-based therapy demonstrating 93.5% efficacy in eliminating severe vaso-occlusive crises in sickle cell disease patients [2], and the expanding repertoire of gene therapies addressing diverse conditions from inherited retinal dystrophy to spinal muscular atrophy to various cancers. The global gene therapy market's trajectory from USD 9.5 billion in 2024 to projected USD 58.87 billion by 2034 reflects both the therapeutic value and commercial viability of this approach [1]. Technological innovations continue driving the field forward. The progression from first- generation retroviral vectors to sophisticated AAV serotypes with engineered tropism, from conventional CRISPR-Cas9 to precision tools like base editors and prime editors, and from autologous manufacturing to potential allogeneic "off-the-shelf" therapies demonstrates the field's dynamic nature [7] [8]. The integration of artificial intelligence for predictive modeling, development of non-viral delivery systems including lipid nanoparticles and extracellular vesicles, and innovations in manufacturing such as point-of-care production devices promise to address current limitations [15] [^86]. The therapeutic scope continues expanding beyond rare genetic disorders into common chronic conditions including cardiovascular disease, neurodegenerative disorders, metabolic diseases, and infectious diseases like HIV [16] [50]. This expansion, coupled with integration into personalized medicine frameworks utilizing pharmacogenomics and multi-omics patient stratification, positions gene therapy as a cornerstone of future healthcare delivery. As we stand at this pivotal juncture in pharmaceutical sciences, gene therapy exemplifies how molecular understanding of disease mechanisms can be translated into therapeutic interventions addressing root causes rather than symptoms. The next decade will witness gene therapy's transition from emerging technology to mainstream medical practice, with profound implications for pharmaceutical care, healthcare systems, and human health globally. The challenge and opportunity for current and future pharmacy professionals is to ensure this powerful technology is developed responsibly, deployed equitably, and utilized optimally to benefit all patients who could be helped by these revolutionary treatments.
REFERENCE
Satyajeet Pawar*, Snehal Kadbhane, Dr. Vijaykumar Kale, Ashlesha Chandale, Gauri Bhange, Gene Therapy: Current Status and Future Perspectives, Int. J. Sci. R. Tech., 2025, 2 (11), 684-692. https://doi.org/10.5281/zenodo.17731503
10.5281/zenodo.17731503