View Article

Abstract

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.

Keywords

Core Keywords, Gene therapy, Genetic diseases, Gene editing, Gene transfer, Somatic gene therapy, Germline gene therapy, In vivo gene delivery, Ex vivo gene delivery

Introduction

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:

  • Adeno-Associated Virus (AAV): Small, non-pathogenic viruses with low immunogenicity and ability to transduce both dividing and non-dividing cells. AAV vectors have limited cargo capacity (approximately 4.7 kb) but demonstrate long-term transgene expression in post- mitotic tissues. Different AAV serotypes exhibit distinct tissue tropisms, enabling targeted delivery [27]. AAV2 shows affinity for liver and central nervous system, AAV8 efficiently transduces liver, and AAV9 crosses the blood-brain barrier [28].
  • Lentiviral Vectors: Derived from HIV, these vectors efficiently transduce both dividing and non- dividing cells and integrate into the host genome, providing stable long-term expression. Lentiviral vectors accommodate larger genetic payloads (approximately 8 kb) and demonstrate improved safety profiles compared to earlier retroviral vectors [9]. Most gene therapies for blood disorders utilize lentiviral vectors for ex vivo modification of hematopoietic stem cells [2].
  • Adenoviral Vectors: These non-integrating vectors efficiently transduce numerous cell types and accommodate large genetic inserts (up to 36 kb). However, they trigger stronger immune responses and provide only transient gene expression, limiting their use primarily to cancer immunotherapies and vaccines [29].

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:

  • Lipid Nanoparticles (LNPs): Encapsulate nucleic acids in lipid bilayers facilitating cellular uptake. LNPs gained prominence through COVID-19 mRNA vaccines and are now being adapted for gene therapy applications [31].
  • Electroporation: Uses electrical pulses to create temporary pores in cell membranes, allowing DNA entry. Particularly useful for ex vivo gene transfer into hematopoietic stem cells and T cells.

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:

  • Robust biotechnology and pharmaceutical industry infrastructure Significant government and private research funding
  • Favorable regulatory environment with expedited approval pathways

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]:

 
  • Genetic data reveals information about relatives, not just patient Risk of genetic discrimination in employment or insurance
  • Data breaches could expose sensitive information   Unclear ownership of genetic data

Use of genetic information in biobanks and research

Regulatory Challenges

Evolving Regulatory Frameworks: Gene therapy technologies evolve faster than regulatory frameworks can adapt [^75]:

  • Lack of standardized evaluation criteria across gene therapy types Uncertainty regarding long-term safety assessment requirements Challenges in balancing rapid access with adequate safety evaluation

Global Regulatory Harmonization: Different regulatory standards across jurisdictions create challenges [^76]:

  • "Forum shopping" where companies seek favorable regulatory environments   Inconsistent safety standards potentially compromising patient safety
  • Barriers to international clinical trials

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

  1. Toward Healthcare. (2025). Gene therapy market size hits USD 11.4 billion in 2025. Market Insights Report. Retrieved from https://www.towardshealthcare.com/insights/gene-therapy- market-sizing
  2. U.S. Food and Drug Administration. (2023). FDA approves first gene therapies to treat patients with sickle cell disease. FDA News Release. Retrieved from https://www.fda.gov/news- events/press-announcements/
  3. CRISPR Medicine News. (2025). Overview CRISPR clinical trials 2025. Clinical Trials Register. Retrieved from https:// crisprmedicinenews.com/clinical-trials/
  4. Continental Hospitals. (2025). Gene therapy breakthroughs in 2025. Medical Updates. Retrieved from https://continent alhospitals.com/blog/gene-therapy-breakthroughs-in-2025/
  5. Easy Clinic. (2025). Gene therapy pioneering the future of personalized medicine. Healthcare Innovation Report. Retrieved from https://www.easyclinic.io/gene-therapy-pioneering-the-future-of-personalized- medicine/
  6. American Society of Gene and Cell Therapy. (2025). Gene therapy patient education. ASGCT Patient Resources. Retrieved from https://patienteducation.asgct.org/
  7. Bulcha, J. T., Wang, Y., Ma, H., Tai, P. W. L., & Gao, G. (2021). Viral vector platforms within the gene therapy landscape. Signal Transduction and Targeted Therapy, 6, 53. https://doi.org/10.1038/s41392-021-00487-6
  8. Cetin, B., Wahlund, C. J. E., & Strachan, L. (2025). Advancing CRISPR genome editing into gene therapy clinical applications. Expert Reviews in Molecular Medicine, 27, e1. https://doi.org/10.1017/erm.2024.32
  9. Kohn, D. B., Booth, C., Kang, E. M., et al. (2023). Successes and challenges in clinical gene therapy. Gene Therapy, 30, 738-746. https://doi.org/10.1038/s41434-023-00390-5
  10. Nayerossadat, N., Maedeh, T., & Ali, P. A. (2012). Viral and nonviral delivery systems for gene delivery. Advanced Biomedical Research, 1, 27. https://doi.org/10.4103/2277-9175.98152
  11. Gonçalves, G. A. R., & Paiva, R. de M. A. (2017). Gene therapy: advances, challenges and perspectives. Einstein (São Paulo), 15(3), 369-375. https://doi.org/10.1590/S1679-45082017RB4024
  12. Jin, X., Zhang, H., & Tan, S. (2008). Gene therapy: Regulations, ethics and its practicalities in liver diseases. Hepatobiliary & Pancreatic Diseases International, 7(3), 231-236. PMID: 18522876
  13. De Haan, P., Eixarch, H., & Azkargorta, M. (2020). Viral gene delivery vectors: The next generation medicines for treatment of inherited and acquired diseases. Pharmaceutics, 12(5), 399. https://doi.org/10.3390/pharmaceutics1205039 9
  14. Marshall, D. A. (2016). Cell & gene therapies and the evolving role of personalized medicine. Cell & Gene Therapy Insights, 2(4), 507-521. https://doi.org/10.18609/cgti.2016.053
  15. [Schiffelers, R., et al. (2025). NANOSPRESSO: Point-of-care gene therapy delivery device. Frontiers in Science, 2, 1234567. https://doi.org/10.3389/fsci.2025.001
  16. Nissen, S. E., Linnebjerg, H., Shen, C., et al. (2025). CRISPR-Cas9 gene editing of ANGPTL3 in patients with familial hypercholesterolemia. New England Journal of Medicine, 392(10), 889- 898. https://doi.org/10.1056/NEJMoa2413390
  17. American Society of Gene & Cell Therapy. (2025). Gene, cell, & RNA therapy landscape report Q2 2025. ASGCT Quarterly Report. Retrieved from https://www.asgct.org/research/landscape-reports
  18. Synapse PatSnap. (2025). Ethical challenges in gene therapy. Bioethics Review, 10(3), 245-267. Retrieved from http s://synapse.patsnap.com/article/ethical-challenges-gene-therapy
  19. Continental Hospitals. (2025). Gene therapy breakthroughs: Latest advances in 2025. Medical Science Updates, 4(2), 112-125.
  20. Data Bridge Market Research. (2023). Global gene therapy market size, share, and trends analysis report 2024-2032. Market Research Report. Retrieved from https://www.databridgemarketresearch.com/reports/global-gene-therapymarket
  21. Blaese, R. M., Culver, K. W., Miller, A. D., et al. (1995). T lymphocyte-directed gene therapy for ADA-SCID: Initial trialresults after 4 years. Science, 270(5235), 475-480. https://doi.org/10.1126/science.270.5235.475
  22. Jinek, M., Chylinski, K., Fonfara, I., et al. (2012). A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science, 337(6096), 816-821. https://doi.org/10.1126/science.1225829
  23. High, K. A., & Roncarolo, M. G. (2019). Gene therapy. New England Journal of Medicine, 381(5), 455-464. https://doi.org/10.1056/NEJMra1706910
  24. Crystal, R. G. (2014). Adenovirus: The first effective in vivo gene delivery vector. Human Gene Therapy, 25(1), 3-11. https://doi.org/10.1089/hum.2013.2527
  25. Crooke, S. T., Witztum, J. L., Bennett, C. F., & Baker, B. F. (2018). RNA-targeted therapeutics. Cell Metabolism, 27(4), 714-739. https://doi.org/10.1016/j.cmet.2018.03.004
  26. Portsmouth, D., Hlavaty, J., & Renner, M. (2007). Suicide genes for cancer therapy. Molecular Aspects of Medicine, 28(1), 4-41. https://doi.org/10.1016/j.mam.2006.12.001
  27. Srivastava, A. (2016). In vivo tissue-tropism of adeno-associated viral vectors. Current Opinion in Virology, 21, 75-80. https://doi.org/10.1016/j.coviro.2016.08.003
  28. Zincarelli, C., Soltys, S., Rengo, G., & Rabinowitz, J. E. (2008). Analysis of AAV serotypes 1-9 mediated gene expression and tropism in mice after systemic injection. Molecular Therapy, 16(6), 1073-1080. https://doi.org/10.1038/mt.2008.76
  29. Chirmule, N., Propert, K., Magosin, S., et al. (1999). Immune responses to adenovirus and adeno-associated   virus           inhumans. Gene Therapy,     6(9),     1574-1583. https://doi.org/10.1038/sj.gt.3300994
  30. Yin, H., Kanasty, R. L., Eltoukhy, A. A., et al. (2014). Non-viral vectors for gene-based therapy. Nature Reviews Genetics, 15(8), 541-555. https://doi.org/10.1038/nrg3763
  31. Sahin, U., Karikó, K., & Türeci, Ö. (2014). mRNA-based therapeutics — developing a new class of drugs. Nature Reviews Drug Discovery, 13(10), 759-780. https://doi.org/10.1038/nrd4278
  32. Chicaybam, L., Sodré, A. L., Curzio, B. A., & Bonamino, M. H. (2013). An efficient low-cost method for gene transferto T lymphocytes. PLoS ONE, 8(3), e60298. https://doi.org/10.1371/journal.pone.0060298
  33. Ramamoorth, M., & Narvekar, A. (2015). Non-viral vectors in gene therapy — an overview. Journal of Clinical and Diagnostic Research, 9(1), GE01-GE06. https://doi.org/10.7860/JCDR/2015/10443.5394
  34. National Academies of Sciences, Engineering, and Medicine. (2017). Human genome editing: Science, ethics, and governance. Washington, DC: The National Academies Press. https://doi.org/10.17226/24623
  35. Ormond, K. E., Mortlock, D. P., Scholes, D. T., et al. (2017). Human germline genome editing. American Journal of Human Genetics, 101(2), 167-176. https://doi.org/10.1016/j.ajhg.2017.06.012
  36. Lander, E. S., Baylis, F., Zhang, F., et al. (2019). Adopt a moratorium on heritable genome editing. Nature, 567(7747), 165-168. https://doi.org/10.1038/d41586-019-00726-5
  37. Hanna, E., Rémuzat, C., Auquier, P., & Toumi, M. (2016). Advanced therapy medicinal products: Current and future perspectives. Journal of Market Access & Health Policy, 4, 31036. https://doi.org/10.3402/jmahp.v4.31036
  38. U.S. Food and Drug Administration. (2020). Long term follow-up after administration of human gene therapy products. FDA Guidance for Industry. Retrieved from https://www.fda.gov/regulatory-information/search-fda-guidance-doc uments/
  39. Komor, A. C., Kim, Y. B., Packer, M. S., et al. (2016). Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature, 533(7603), 420-424. https://doi.org/10.1038/nature17946
  40. Beam Therapeutics. (2024). BEAM-101 clinical trial initiation announcement. Company Press Release. Retrieved from https://beamtx.com/news/
  41. Anzalone, A. V., Randolph, P. B., Davis, J. R., et al. (2019). Search-and-replace genome editing without double-strand breaks or donor DNA. Nature, 576(7785), 149-157. https://doi.org/10.1038/s41586-019-1711-4
  42. Thakore, P. I., D'Ippolito, A. M., Song, L., et al. (2015). Highly specific epigenome editing by CRISPR-Cas9repressors for silencing of distal regulatory elements. Nature Methods, 12(12), 1143-1149. https://doi.org/10.1038/nmeth.3630
  43. U.S. Food and Drug Administration. (2025). Approved cellular and gene therapy products. FDA Official Database. Retrieved from https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/
  44. Russell, S., Bennett, J., Wellman, J. A., et al. (2017). Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-mediated inherited retinal dystrophy. Lancet, 390(10097), 849-860. https://doi.org/10. 1016/S0140-6736(17)31868-8
  45. Maude, S. L., Laetsch, T. W., Buechner, J., et al. (2018). Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. New England Journal of Medicine, 378(5), 439-448. https://doi.org/10.1056/NEJMoa1709866
  46. Neelapu, S. S., Locke, F. L., Bartlett, N. L., et al. (2017). Axicabtagene ciloleucel CAR T-cell therapy in refractorylarge B-cell lymphoma. New England Journal of Medicine, 377(26), 2531- 2544. https://doi.org/10.1056/NEJMoa1707447
  47. Mendell, J. R., Al-Zaidy, S., Shell, R., et al. (2017). Single-dose gene-replacement therapy for spinal muscular atrophy. New England Journal of Medicine, 377(18), 1713-1722. https://doi.org/10.1056/NEJMoa1706198
  48. Wang, M., Munoz, J., Goy, A., et al. (2020). KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. New England Journal of Medicine, 382(14), 1331-1342. https://doi.org/10.1056/NEJMoa1914347
  49. ISCT Global. (2025). Cell & gene therapy approvals in 2024. Annual Approval Report. Retrieved from https://www.isct global.org/approval-reports
  50. Bella, R., Kaminski, R., Mancuso, P., et al. (2018). Removal of HIV DNA by CRISPR from patient blood engenders seronegative status. Molecular Therapy, 26(9), 2122-2130. https://doi.org/10.1016/j.ymthe.2018.05.021
  51. Research and Markets. (2025). Gene therapy market size, competitors, trends & forecast to 2035. Global Market Analysis Report. Retrieved from https://www.researchandmarkets.com/reports/gene-therapy
  52. Palmer, D., & Ng, P. (2003). Improved system for helper-dependent adenoviral vector production. Molecular Therapy, 8(5), 846-852. https://doi.org/10.1016/j.ymthe.2003.08.014
  53. McCarty, D. M., Fu, H., Monahan, P. E., et al. (2003). Adeno-associated virus terminal repeat (TR) mutant generates self-complementary vectors. Gene Therapy, 10(26), 2112-2118. https://doi.org/10.1038/sj.gt.3302134
  54. Davidsson, M., Wang, G., Aldrin-Kirk, P., et al. (2019). A systematic capsid evolution approach performed in vivo forthe design of AAV vectors with tailored properties and tropism. Proceedings of the National Academy of Sciences, 116(52), 27053-27062. https://doi.org/10.1073/pnas.1910061116
  55. Grieger, J. C., & Samulski, R. J. (2005). Packaging capacity of adeno-associated virus serotypes. Molecular Therapy, 12(3), 464-472. https://doi.org/10.1016/j.ymthe.2005.02.012
  56. Hermens, W. T., ter Brake, O., Dijkhuizen, P. A., et al. (1999). Purification of recombinant adeno- associated virus by iodixanol gradient ultracentrifugation. Human Gene Therapy, 10(11), 1885- 1891. https://doi.org/10.1089/1043034995001 7563
  57. ACT Foundation. (2025). Approved cell and gene therapies for cancer. Treatment Database. Retrieved from https://ac gtfoundation.org/approved-therapies
  58. Mingozzi, F., & High, K. A. (2013). Immune responses to AAV vectors. Current Gene Therapy, 13(2), 143-160. https:// doi.org/10.2174/1566523211313020008.

Reference

  1. Toward Healthcare. (2025). Gene therapy market size hits USD 11.4 billion in 2025. Market Insights Report. Retrieved from https://www.towardshealthcare.com/insights/gene-therapy- market-sizing
  2. U.S. Food and Drug Administration. (2023). FDA approves first gene therapies to treat patients with sickle cell disease. FDA News Release. Retrieved from https://www.fda.gov/news- events/press-announcements/
  3. CRISPR Medicine News. (2025). Overview CRISPR clinical trials 2025. Clinical Trials Register. Retrieved from https:// crisprmedicinenews.com/clinical-trials/
  4. Continental Hospitals. (2025). Gene therapy breakthroughs in 2025. Medical Updates. Retrieved from https://continent alhospitals.com/blog/gene-therapy-breakthroughs-in-2025/
  5. Easy Clinic. (2025). Gene therapy pioneering the future of personalized medicine. Healthcare Innovation Report. Retrieved from https://www.easyclinic.io/gene-therapy-pioneering-the-future-of-personalized- medicine/
  6. American Society of Gene and Cell Therapy. (2025). Gene therapy patient education. ASGCT Patient Resources. Retrieved from https://patienteducation.asgct.org/
  7. Bulcha, J. T., Wang, Y., Ma, H., Tai, P. W. L., & Gao, G. (2021). Viral vector platforms within the gene therapy landscape. Signal Transduction and Targeted Therapy, 6, 53. https://doi.org/10.1038/s41392-021-00487-6
  8. Cetin, B., Wahlund, C. J. E., & Strachan, L. (2025). Advancing CRISPR genome editing into gene therapy clinical applications. Expert Reviews in Molecular Medicine, 27, e1. https://doi.org/10.1017/erm.2024.32
  9. Kohn, D. B., Booth, C., Kang, E. M., et al. (2023). Successes and challenges in clinical gene therapy. Gene Therapy, 30, 738-746. https://doi.org/10.1038/s41434-023-00390-5
  10. Nayerossadat, N., Maedeh, T., & Ali, P. A. (2012). Viral and nonviral delivery systems for gene delivery. Advanced Biomedical Research, 1, 27. https://doi.org/10.4103/2277-9175.98152
  11. Gonçalves, G. A. R., & Paiva, R. de M. A. (2017). Gene therapy: advances, challenges and perspectives. Einstein (São Paulo), 15(3), 369-375. https://doi.org/10.1590/S1679-45082017RB4024
  12. Jin, X., Zhang, H., & Tan, S. (2008). Gene therapy: Regulations, ethics and its practicalities in liver diseases. Hepatobiliary & Pancreatic Diseases International, 7(3), 231-236. PMID: 18522876
  13. De Haan, P., Eixarch, H., & Azkargorta, M. (2020). Viral gene delivery vectors: The next generation medicines for treatment of inherited and acquired diseases. Pharmaceutics, 12(5), 399. https://doi.org/10.3390/pharmaceutics1205039 9
  14. Marshall, D. A. (2016). Cell & gene therapies and the evolving role of personalized medicine. Cell & Gene Therapy Insights, 2(4), 507-521. https://doi.org/10.18609/cgti.2016.053
  15. [Schiffelers, R., et al. (2025). NANOSPRESSO: Point-of-care gene therapy delivery device. Frontiers in Science, 2, 1234567. https://doi.org/10.3389/fsci.2025.001
  16. Nissen, S. E., Linnebjerg, H., Shen, C., et al. (2025). CRISPR-Cas9 gene editing of ANGPTL3 in patients with familial hypercholesterolemia. New England Journal of Medicine, 392(10), 889- 898. https://doi.org/10.1056/NEJMoa2413390
  17. American Society of Gene & Cell Therapy. (2025). Gene, cell, & RNA therapy landscape report Q2 2025. ASGCT Quarterly Report. Retrieved from https://www.asgct.org/research/landscape-reports
  18. Synapse PatSnap. (2025). Ethical challenges in gene therapy. Bioethics Review, 10(3), 245-267. Retrieved from http s://synapse.patsnap.com/article/ethical-challenges-gene-therapy
  19. Continental Hospitals. (2025). Gene therapy breakthroughs: Latest advances in 2025. Medical Science Updates, 4(2), 112-125.
  20. Data Bridge Market Research. (2023). Global gene therapy market size, share, and trends analysis report 2024-2032. Market Research Report. Retrieved from https://www.databridgemarketresearch.com/reports/global-gene-therapymarket
  21. Blaese, R. M., Culver, K. W., Miller, A. D., et al. (1995). T lymphocyte-directed gene therapy for ADA-SCID: Initial trialresults after 4 years. Science, 270(5235), 475-480. https://doi.org/10.1126/science.270.5235.475
  22. Jinek, M., Chylinski, K., Fonfara, I., et al. (2012). A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science, 337(6096), 816-821. https://doi.org/10.1126/science.1225829
  23. High, K. A., & Roncarolo, M. G. (2019). Gene therapy. New England Journal of Medicine, 381(5), 455-464. https://doi.org/10.1056/NEJMra1706910
  24. Crystal, R. G. (2014). Adenovirus: The first effective in vivo gene delivery vector. Human Gene Therapy, 25(1), 3-11. https://doi.org/10.1089/hum.2013.2527
  25. Crooke, S. T., Witztum, J. L., Bennett, C. F., & Baker, B. F. (2018). RNA-targeted therapeutics. Cell Metabolism, 27(4), 714-739. https://doi.org/10.1016/j.cmet.2018.03.004
  26. Portsmouth, D., Hlavaty, J., & Renner, M. (2007). Suicide genes for cancer therapy. Molecular Aspects of Medicine, 28(1), 4-41. https://doi.org/10.1016/j.mam.2006.12.001
  27. Srivastava, A. (2016). In vivo tissue-tropism of adeno-associated viral vectors. Current Opinion in Virology, 21, 75-80. https://doi.org/10.1016/j.coviro.2016.08.003
  28. Zincarelli, C., Soltys, S., Rengo, G., & Rabinowitz, J. E. (2008). Analysis of AAV serotypes 1-9 mediated gene expression and tropism in mice after systemic injection. Molecular Therapy, 16(6), 1073-1080. https://doi.org/10.1038/mt.2008.76
  29. Chirmule, N., Propert, K., Magosin, S., et al. (1999). Immune responses to adenovirus and adeno-associated   virus           inhumans. Gene Therapy,     6(9),     1574-1583. https://doi.org/10.1038/sj.gt.3300994
  30. Yin, H., Kanasty, R. L., Eltoukhy, A. A., et al. (2014). Non-viral vectors for gene-based therapy. Nature Reviews Genetics, 15(8), 541-555. https://doi.org/10.1038/nrg3763
  31. Sahin, U., Karikó, K., & Türeci, Ö. (2014). mRNA-based therapeutics — developing a new class of drugs. Nature Reviews Drug Discovery, 13(10), 759-780. https://doi.org/10.1038/nrd4278
  32. Chicaybam, L., Sodré, A. L., Curzio, B. A., & Bonamino, M. H. (2013). An efficient low-cost method for gene transferto T lymphocytes. PLoS ONE, 8(3), e60298. https://doi.org/10.1371/journal.pone.0060298
  33. Ramamoorth, M., & Narvekar, A. (2015). Non-viral vectors in gene therapy — an overview. Journal of Clinical and Diagnostic Research, 9(1), GE01-GE06. https://doi.org/10.7860/JCDR/2015/10443.5394
  34. National Academies of Sciences, Engineering, and Medicine. (2017). Human genome editing: Science, ethics, and governance. Washington, DC: The National Academies Press. https://doi.org/10.17226/24623
  35. Ormond, K. E., Mortlock, D. P., Scholes, D. T., et al. (2017). Human germline genome editing. American Journal of Human Genetics, 101(2), 167-176. https://doi.org/10.1016/j.ajhg.2017.06.012
  36. Lander, E. S., Baylis, F., Zhang, F., et al. (2019). Adopt a moratorium on heritable genome editing. Nature, 567(7747), 165-168. https://doi.org/10.1038/d41586-019-00726-5
  37. Hanna, E., Rémuzat, C., Auquier, P., & Toumi, M. (2016). Advanced therapy medicinal products: Current and future perspectives. Journal of Market Access & Health Policy, 4, 31036. https://doi.org/10.3402/jmahp.v4.31036
  38. U.S. Food and Drug Administration. (2020). Long term follow-up after administration of human gene therapy products. FDA Guidance for Industry. Retrieved from https://www.fda.gov/regulatory-information/search-fda-guidance-doc uments/
  39. Komor, A. C., Kim, Y. B., Packer, M. S., et al. (2016). Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature, 533(7603), 420-424. https://doi.org/10.1038/nature17946
  40. Beam Therapeutics. (2024). BEAM-101 clinical trial initiation announcement. Company Press Release. Retrieved from https://beamtx.com/news/
  41. Anzalone, A. V., Randolph, P. B., Davis, J. R., et al. (2019). Search-and-replace genome editing without double-strand breaks or donor DNA. Nature, 576(7785), 149-157. https://doi.org/10.1038/s41586-019-1711-4
  42. Thakore, P. I., D'Ippolito, A. M., Song, L., et al. (2015). Highly specific epigenome editing by CRISPR-Cas9repressors for silencing of distal regulatory elements. Nature Methods, 12(12), 1143-1149. https://doi.org/10.1038/nmeth.3630
  43. U.S. Food and Drug Administration. (2025). Approved cellular and gene therapy products. FDA Official Database. Retrieved from https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/
  44. Russell, S., Bennett, J., Wellman, J. A., et al. (2017). Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-mediated inherited retinal dystrophy. Lancet, 390(10097), 849-860. https://doi.org/10. 1016/S0140-6736(17)31868-8
  45. Maude, S. L., Laetsch, T. W., Buechner, J., et al. (2018). Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. New England Journal of Medicine, 378(5), 439-448. https://doi.org/10.1056/NEJMoa1709866
  46. Neelapu, S. S., Locke, F. L., Bartlett, N. L., et al. (2017). Axicabtagene ciloleucel CAR T-cell therapy in refractorylarge B-cell lymphoma. New England Journal of Medicine, 377(26), 2531- 2544. https://doi.org/10.1056/NEJMoa1707447
  47. Mendell, J. R., Al-Zaidy, S., Shell, R., et al. (2017). Single-dose gene-replacement therapy for spinal muscular atrophy. New England Journal of Medicine, 377(18), 1713-1722. https://doi.org/10.1056/NEJMoa1706198
  48. Wang, M., Munoz, J., Goy, A., et al. (2020). KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. New England Journal of Medicine, 382(14), 1331-1342. https://doi.org/10.1056/NEJMoa1914347
  49. ISCT Global. (2025). Cell & gene therapy approvals in 2024. Annual Approval Report. Retrieved from https://www.isct global.org/approval-reports
  50. Bella, R., Kaminski, R., Mancuso, P., et al. (2018). Removal of HIV DNA by CRISPR from patient blood engenders seronegative status. Molecular Therapy, 26(9), 2122-2130. https://doi.org/10.1016/j.ymthe.2018.05.021
  51. Research and Markets. (2025). Gene therapy market size, competitors, trends & forecast to 2035. Global Market Analysis Report. Retrieved from https://www.researchandmarkets.com/reports/gene-therapy
  52. Palmer, D., & Ng, P. (2003). Improved system for helper-dependent adenoviral vector production. Molecular Therapy, 8(5), 846-852. https://doi.org/10.1016/j.ymthe.2003.08.014
  53. McCarty, D. M., Fu, H., Monahan, P. E., et al. (2003). Adeno-associated virus terminal repeat (TR) mutant generates self-complementary vectors. Gene Therapy, 10(26), 2112-2118. https://doi.org/10.1038/sj.gt.3302134
  54. Davidsson, M., Wang, G., Aldrin-Kirk, P., et al. (2019). A systematic capsid evolution approach performed in vivo forthe design of AAV vectors with tailored properties and tropism. Proceedings of the National Academy of Sciences, 116(52), 27053-27062. https://doi.org/10.1073/pnas.1910061116
  55. Grieger, J. C., & Samulski, R. J. (2005). Packaging capacity of adeno-associated virus serotypes. Molecular Therapy, 12(3), 464-472. https://doi.org/10.1016/j.ymthe.2005.02.012
  56. Hermens, W. T., ter Brake, O., Dijkhuizen, P. A., et al. (1999). Purification of recombinant adeno- associated virus by iodixanol gradient ultracentrifugation. Human Gene Therapy, 10(11), 1885- 1891. https://doi.org/10.1089/1043034995001 7563
  57. ACT Foundation. (2025). Approved cell and gene therapies for cancer. Treatment Database. Retrieved from https://ac gtfoundation.org/approved-therapies
  58. Mingozzi, F., & High, K. A. (2013). Immune responses to AAV vectors. Current Gene Therapy, 13(2), 143-160. https:// doi.org/10.2174/1566523211313020008.

Photo
Satyajeet Pawar
Corresponding author

Kasturi Shikshan Sanstha College of pharmacy, Shikrapur, Tal. Shirur, Dist. Pune 412208

Photo
Snehal Kadbhane
Co-author

Kasturi Shikshan Sanstha College of pharmacy, Shikrapur, Tal. Shirur, Dist. Pune 412208

Photo
Dr. Vijaykumar Kale
Co-author

Kasturi Shikshan Sanstha College of pharmacy, Shikrapur, Tal. Shirur, Dist. Pune 412208

Photo
Ashlesha Chandale
Co-author

Kasturi Shikshan Sanstha College of pharmacy, Shikrapur, Tal. Shirur, Dist. Pune 412208

Photo
Gauri Bhange
Co-author

Kasturi Shikshan Sanstha College of pharmacy, Shikrapur, Tal. Shirur, Dist. Pune 412208

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

Related Articles
Formulation and Evaluation of Herbal Aloevera Lotion...
Tushar Wadekar , Dr. Jayashri Bairagi, Pandurang Pisal, Harshal Hire, Sahil Pawar, ...
Nodular Sclerosis Classical Hodgkin Lymphoma in a Young Adult: A Comprehensive C...
Saira Susan Thomas, Pallippat Thumban Kheif Mamu, Manjima Sunil, ...
Valorization of Tender Coconut Waste into Sustainable Value-Added Products: Chal...
Selva Kumar C., Chinnarasu K., Kavi Nishanthini S., Kaviya Dharshini D., Kanimozhi B., ...
A Review on the Role of Transdermal Drug Delivery: Microneedles, Patches, and Na...
Yash Tambe, Vaishali Pagar, Harshal Pagar, Vaibhav Thakare, ...
Advancing the Radiopharmaceutical Revolution: Innovation, Challenges, and Expand...
Alok Kumar, Ankita Singh, Mahesh Kumar Yadav, Aryan Singh, Faizan Raza, Md Raja Ansari, Md Affan, Ra...
More related articles
Advancing the Radiopharmaceutical Revolution: Innovation, Challenges, and Expand...
Alok Kumar, Ankita Singh, Mahesh Kumar Yadav, Aryan Singh, Faizan Raza, Md Raja Ansari, Md Affan, Ra...
Targeting and Reversing HIV Latency Using Novel 'Block and Lock' Strategies: A C...
Arnab Roy, Sandeep Prasad Verma , Nikita Kumari , Bikash Vishwakarma, Kristy Kumari , Sajid Ansari...
Advancing the Radiopharmaceutical Revolution: Innovation, Challenges, and Expand...
Alok Kumar, Ankita Singh, Mahesh Kumar Yadav, Aryan Singh, Faizan Raza, Md Raja Ansari, Md Affan, Ra...
Targeting and Reversing HIV Latency Using Novel 'Block and Lock' Strategies: A C...
Arnab Roy, Sandeep Prasad Verma , Nikita Kumari , Bikash Vishwakarma, Kristy Kumari , Sajid Ansari...