Faculty of Pharmaceutical Sciences, Mewar University Chittorgarh Rajasthan, India, 312901
Although cancer remains one of the foremost causes of human death worldwide, the traditional therapies available in the field, of which chemotherapy, radiation, and surgery are but a few examples, usually manifest limited efficacy and high toxicity, while the development of resistance often renders them ineffective. One such hope in cancer therapy is the field of gene therapy targeted toward either switching off or correcting the genetic abnormality responsible for the advancement of cancer. In the past few decades, the field has experienced major advancements in gene-editing technologies, newer delivery systems, and immunogene therapy. This review provides detailed insight into current advances in cancer gene therapy and its future potentials while underlining freshly emerging technologies such as CRISPR-Cas gene editing, RNA-based medicine (siRNA, mRNA, and miRNA), and the second generation of CAR-T cell therapy. Non-viral gene delivery strategies, such as nanoparticles, liposomes, and extracellular vesicles, are put forward as safer and more effective solutions than viral vectors. Further, applications of AI and computational biology in gene therapy are described, emphasizing on improving gene-editing accuracy, prediction of therapeutic outcomes, and personalizing cancer treatment. Synthetic biology approaches such as engineered gene circuits and programmable gene switches are being targeted for their ability to increase therapeutic specificity and safety. However, even with these prospects, challenges including off-target effects, immunogenicity, scale-up, regulatory barriers, and various other ethical issues remain. This review thus reveals the fast pace that is characteristic for cancer gene therapy along with the opportunities and challenges ahead. This improves massively the future of cancer gene therapy when all present-day challenges are tackled with the help of cutting-edge technologies to better improve the outcome of patients with more accurate, long-lasting, and less invasive therapeutic interventions.
The discovery of novel therapeutic methods should go because presently, cancer remains one of the top illnesses that lead to morbidity and mortality in the world. Gene therapy has come up as a new avenue for cancer management on the basis of the therapeutic effects of genetically-induced alterations that would inhibit oncogenic signaling or enhance immune response against tumor cells (1). During the last couple of decades, there has been much progress in cancer genomics, which has vastly refined gene therapy towards demands for specificity and improved applications. Thus, gene manipulation has gained much more specificity and maximum efficiency through gene editing technologies such as CRISPR-Cas9, TALENs, and zinc-finger nucleases (2). Common effects may be wide damage and resistance in tissues among chemotherapy and radiotherapy. Gene therapy would be best suited to personalized treatments that may be curative or have potentials to target the fundamental genetic mutations that cause tumorigenesis (3). This will open up novel avenues for newer modes of delivery such as viral and non-viral vectors toward attaining improvements in stability and specificity in gene transfer mechanisms for higher therapeutic advantages with lesser side effects (4). Gene therapy for cancer has made great strides, with the promise of advancing the development of new high-tech treatments such as oncolytic virotherapy, RNAi-their gene silencing, or "cell-in-cell" therapy, which uses the immune system with "chimeric antigen receptors" (5). Undoubtedly, these remarkable novel emerging technologies would change the face of cancer therapy from a deadly adversary to a most survivable one. The future and innovative technologies relating to cancer gene therapy are addressed in this review, which also considers advances toward recent challenges and various possibilities for their clinical implications. It has also incorporated the analysis of the recent progress made thus far and indeed its implication in the field of oncology to inform on how gene therapy is likely to shape the course of cancer therapy out into the future (6).
Emerging Technologies in Cancer Gene Therapy
Owing to the onset of novel technologies, which include refinement of specificity, enhancement of effectiveness and safety, gene therapy for cancer has undergone significant improvement. CRISPR-based new genome editing, RNA therapies, oncolytic viruses, nanotechnology delivery systems, and artificial intelligence (AI) applications in gene therapy are imparting a new life to cancer treatment. These novel strategies are directed toward achieving therapeutic response enhancement combined with lesser adverse effects (3).
1. CRISPR-Based Genome Editing2. RNA-Based
CRISPR-Cas9 has had a paradigm shift for cancer gene therapy, as it provides for precise and effective genome editing. This would allow for targeted disruption of oncogenes, correction of tumor-suppressor genes, and enhancement of the immune system response. Even more advanced CRISPR strategies, base editing and prime editing, offer much more precision, also without generating double-strand breaks, and thus with minimal off-target effects. Clinical trials with CRISPR-engineered T cells in cancer patients have shown promising results-mostly for hematological malignancies (2). AI-assisted design of CRISPR guides further helps improve specificity and efficiency in genome editing for cancer treatments (7).
2. Therapies
RNA-based therapies like small interfering RNA (siRNA), microRNA (miRNA), and messenger RNA (mRNA) vaccines are powerful tools for cancer gene therapy. Theresa siRNAs and miRNAs shut down oncogenic pathways and, on the other hand, mRNA vaccines induce immune responses against tumors. COVID-19 mRNA vaccine success has accelerated applying these therapies in oncology for personalized cancer vaccine development targeting tumor-specific mutations (8). The new running clinical trials have demonstrated that RNA-based intervention controls tumor growth and enhances immune surveillance (9).
3. Oncolytic Virus Therapy
Oncolytic viruses (ONV) are viruses engineered to kill cancer cells selectively and at the same time induce immune responses against tumors. Viruses like herpes simplex virus, adenoviruses, and vaccinia viruses have been genetically engineered to enhance their tumor specificity and immunostimulatory properties. Talimogene laherparepvec (T-VEC), the product based on oncolytic herpes simplex virus, is the first U.S. FDA-approved oncolytic virus for melanoma therapy. The next-generation oncolytic viruses are being designed to encode immune-modulating genes to enhance therapeutic potency and improve patient outcomes with cancer immunotherapy (10).
4. Delivering Genes Based on Nanotechnology Ideas
Nanotechnology also plays a crucial role in improving the delivery of gene therapy into cancer cells. Some nanoparticles, namely lipid nanoparticles (LNPs), polymeric nanoparticles, and gold nanoparticles, increase the stability and bioavailability of genetic materials, including DNA, RNA, and CRISPR elements. Similarly, LNPs effectively utilized in mRNA COVID-19 vaccines are being harnessed for targeted gene delivery in cancer therapies. These nanocarriers improve cellular uptake, limit immune clearance, and enable accurate gene editing in tumors (11).
5. CAR-T Cell and TCR-T Cell Therapy
Chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of hematologic malignancies. CAR-T cells can be programmed to recognize and destroy tumor-associated antigens and, consequently, lead to substantial remissions in leukemias and lymphomas. Now there are attempts to modify and boost CAR-T therapy for applicability against solid tumors through, for example, "armored" CAR-T cells and combinatorial treatments with immune checkpoint inhibitors. Moreover, there is T-cell receptor (TCR) therapy, supported by intracellular tumor antigens and proving a great alternative for tumors lacking proper surface targets (12).
6. Synthetic Biology and Gene Circuits
Synthetic biology is making great advances toward building programmable gene circuits for cancer therapy. Such circuits enable engineered cells to find and react to specific cues from the tumor microenvironment, activating therapeutic genes only in the presence of cancer cells, for example, through "AND-gate" gene circuits, which ensure gene therapy will be activated only by a number of tumor-specific stimuli, minimizing off-target toxicity. This further enhances maximization of safety and reduction of side effects in the applications for gene therapy (13).
7. AI and Computational Methods in Gene Therapy
The artificial intelligence (AI) assimilation in the cancer gene therapy is intended to optimize gene editing and estimate patient response to personalized treatment planning. AI-based algorithms help in identifying the tumor-specific genetic mutations, high-precision designing of CRISPR guide RNA, and large-scale genomic data analysis for biomarker identification. Also, machine learning models are improving the identification of patients most likely to benefit from gene therapy, thus enhancing the success rate of clinical trials (14).
Future Directions in Cancer Gene Therapy
Gene therapy for cancer is going through such changing times right now that one might say new avenues opened up for even more precision, safety, and accessibility in this form of therapy. For instance, genome editing, customized medicine, next-generation delivery platforms, combination therapy, and AI-based approaches will provide the very seeds of the redefinition of the future of cancer treatment. Such breakthroughs promise to go beyond existing boundaries for the enhanced benefit of patients (3).
1. Developments on CRISPR and Next-Gen Gene Editing
Currently, gene therapies for cancer shine a light around new horizons opened by advances in the CRISPR field-from base editing to prime editing. These types of ingredients allow for greater specificity in that they permit nucleotide changes while avoiding double-strand brakes, vastly reducing the chances for any off-target events. Another modulation that is being explored is that of oncogene expression: CRISPR interference (CRISPRi) is an example. As is CRISPR activation (CRISPRa). Also, the epigenome editing might allow reversible alteration in cancer therapy; it works by changing the expression of genes through mechanisms that do not result in any alteration in the DNA sequence (15).
2. Precision or Personalized Gene Therapy
Some advances made in genomics and biomarker discovery have paved the way for highly individualized gene therapies. Whole-genome sequencing studies with artificial intelligence-based data analysis will pave the way to finding a patient-specific mutation for individualized treatment programs. Targeting specific genetic modification in the patient's cancer will enhance efficacy while lessening adverse effects caused by therapeutic intervention. This is consistent with the idea of "N-of-1" clinical trials that personalize therapies for each individual patient based on his or her specific genetic makeup (16).
3. Superiority of Delivery Systems in Gene Delivery
Future gene therapies should focus increasingly on the development of more potent and selective delivery vehicles. Improved engineered adenovirus and lentivirus are now on their way to develop into novel viral vectors with reduced immunogenicity and to increase tumor specificity. These novel non-viral delivery systems include lipid nanoparticles (LNPs) and extracellular vesicles, which are slowly gaining popularity as they are biocompatible and can cross biological barriers. These latest inventions will ensure the safety and efficacy of gene delivery into solid tumors (11).
4. Integration of Gene Therapy and Immunotherapy
It is the integration between gene therapy and immunotherapy that appears to be the most promising approach for improving anti-tumor responses. For solid tumors, CAR-T cell therapy is being optimized through gene modifications that increase tumor infiltration and resistance to the immunosuppressive microenvironment. Gene-editing approaches are also concerned with augmenting natural killer (NK) cells and TCR-T cells, resulting in a wider variety of immune-based therapies against cancer. Future advancements will probably feature "universal" allogeneic CAR-T that could be mass-produced and applied to multiple patients (12).
5. Synthetic Biology and Programmable Gene Circuits
Synthetic biology is accelerating gene therapy progress by assisting in the design of programmable gene circuits which are tuned to be activated under specific tumor condition. The circuits allow for well-controlled expression of genes with little toxicity and off-target effects, such as AND-gate logic systems that ensure therapeutic genes are activated only when multiple tumor-specific signals are detected. Augmenting safety and accuracy of gene therapies in the treatment of cancers is such technologies (13).
6. AI Chemical Discovery and Treatment Optimization
Well that, too, has contributed its fair share in improving cancer gene therapy by predicting gene therapy targets, designing CRISPR guides, and analyzing that of each patient response. Clinical trials are improved by AI-based models that define the patient subgroups likely to respond to given gene therapies. AI-based simulation also involves maximizing vector design and anticipates their off-targeting effects, driving safer and more efficient treatment development (14).
7. Extension of Gene Therapy for Solid Tumors
While gene therapy has been successfully applied in the treatment of hematologic malignancies, solid tumors are far more complex due to their microenvironments and the heterogeneous nature of these tumors. Future strategies may include combinations of gene therapy together with tumor-targeting nanoparticles, increased tumor penetration of engineered immune cells, and gene-editing technologies to reprogram the tumor microenvironment itself. These hurdles will be essential in furthering the application of gene therapy to many more cancers (17).
8. Ethical and Regulatory Issues for Widespread Application
Wider use of gene therapies must have progressed along ethical and regulatory lines to ensure safe and equitable access. Future policy will have to start addressing germline editing, monitoring safety long term, and cost. Tighter regulatory frameworks will also be necessary for rapid approval of any new gene therapies to ensure full safety compliance (18).
Challenges and Ethical Considerations
However, even with considerable advancements in research and development related to cancer gene therapy, many barriers stand against it being employed in everyday clinical practice. Inherent amongst these barriers are technical and immune responses to the treatment, poor-cost delivery methods, and regulatory complications. There are also issues of ethics that include gene editing, permission from patients, and equity for access. The development of these factors will render gene therapy more secure, effective, and preferable (3).
1. Technical and Scientific Challenges
One of the remarkable challenges in cancer gene therapy is accurate and effective gene editing. Though CRISPR-Cas9 and other genome editing technologies have created groundbreaking changes in gene therapy, they are not without the risk of potential off-target activity, mutation accidents, and genomic instability. Off-target activities may activate oncogenes in undesired circumstances or inactivate tumor-suppressor genes with undesirable results. Advances in base editing, prime editing, and computer-aided guide RNA design are currently under investigation to enhance specificity and reduce unwanted mutations (15).
2. Immune Response and Safety Issues
Gene therapy usually uses viral vectors like adenoviruses and lentiviruses for delivery of genes. These vectors are, however, very immunogenic, resulting in an inflammatory state, toxicity, or reduced therapeutic effect. In rare patients, severe immune reactions could be quite low, which diminishes the success of gene therapy. Attempts are being made to further minimize these risks by immunosuppression, genetically engineering viral vectors to carry fewer immunogenic components, and by utilizing nonviral delivery systems such as lipid nanoparticles (LNPs) (19).
3. Target Specificity and Delivery Challenge
The successful delivery of gene therapy to solid tumors remains difficult in the ever-complex microenvironment of tumors. Poor penetration into solid tumors has emerged as an important problem affecting several gene therapies in their activity as well. Also required is targeted delivery of genetic alteration to the cancer cells, in such a way that normal cells are spared from the unwanted effects, thus potentially reducing toxicity. This will be developed through tumor-targeting nanoparticles, synthetic gene circuits, and tissue-specific promoters that engineers improve targeted delivery of gene therapy (11).
4. Exorbitant Prices and Unaffordability
The costs associated with developing cancer gene therapy and their delivery are so inflated that they have become a prohibitive cost for most patients. For example, CAR-T cell therapies, which have succeeded quite spectacularly in the treatment of blood cancers, are on average ranging hundreds of thousands of dollars per patient. This is almost entirely a function of the complexity of manufacturing, the personalization of treatments, and regulatory validation thrown into the mix. Cutting costs in production, expanding the scale of manufacturing, and introducing value-based pricing are essential in making gene therapy within reach and affordable (20).
5. Regulatory and Approval Complexities
The regulatory process associated with gene therapy is itself complicated and requires stringent safety and efficacy testing. Bodies such as the FDA and EMA enforce high standards to ensure that when products reach the stage of clinical development, immense safety tests have already been well and truly done. Unfortunately, these overly protective laws may serve to delay the release of drugs that could save lives. Also, to help with the accelerated clinical uptake of cutting-edge gene therapies, streamlining of the regulatory legal framework with assurances of safety shall become an absolute prerequisite. Such long-term monitoring is intended to reveal risks of long-term adverse effects in patients (21).
6. Ethical Issues in Gene Editing
The ethical standing of gene editing, in particular germline intervention, is a debated tangle. While somatic gene therapy would ethically treat only the affected cells, germline modification will have heritable effects that raise questions on its long-term implications. Adding insult to injury are the grafts of gene editing that may be used for nontherapeutic purposes directed more at cosmetics than curing an illness. Internationally accepted standards and rules are being crafted now to set the framework for the ethical application of gene editing (18).
7. Informed Consent and Patient Autonomy
Gene therapy normally comprises complex procedures whereby patients must give informed consent. However, many patients may not have a clear understanding of the risks, possible benefits, or long-term effects since a great many of the gene therapies are still experimental. This is why patients must be allowed to know information that is clear and transparent so that they may participate in ethical decision-making. Additional ethical questions arise in instances where gene therapy is tested among vulnerable populations, highlighting the necessity of stringent ethical oversight (22).
8. Equitable Access and Global Disparities
Most assessment notes that gene therapies are usually available in high-income countries creating global disparities in the access to modern cancer therapies. Most of the balloons do not have the infrastructure, expertise, and funds needed to initiate gene therapy programs in developing regions. Therefore, worldwide cooperation and investment in healthcare infrastructure, as well as the establishment of policies that favor equitable delivery of gene therapy innovations, will be required to overcome such disparities (23).
Cancer gene therapy has emerged to be an exciting new horizon in oncology with the potential of providing new ways to target genetic defects behind cancer development. The incorporation of the newly engineered gene-editing technologies, like CRISPR-Cas, TALENs, and prime editing, into precision medicine has enabled the most humane and, in practical terms, precise form of genetic alteration with minimized off-target effects. Next to this, RNA therapeutics have made a considerable impact, including but not limited to the application of siRNA, miRNA, and mRNA-based therapies, all of which target gene expression and improve therapeutic outcomes. In spite of these advances, challenges like delivery efficiency, immunogenicity, and off-target mutations will remain some of the key hurdles to be crossed in maximizing any tangible clinical gain. Advanced non-viral gene delivery technologies such as lipid nanoparticles (LNPs), extracellular vesicles, and polymer-based vectors emerge as more effective and safer alternatives than conventional viral vectors. These advances make gene therapy safer by stimulating limited immune responses while being accurately focused on sites of cancer. In addition, second-generation CAR-T cell and TCR-T cell therapies are being refined for the treatment of solid tumors and overcome the problems of ineffective tumor infiltration and immunosuppressive tumor microenvironment. The next generation cancer gene therapies could further exploit these new delivery platforms to have an increased level of specificity, lower toxicity, and enhanced efficacy in the long term. The application of AI and computational biology to gene therapy has dramatically accelerated the discovery and optimization of gene-editing targets. AI algorithms are used for accurate CRISPR guide generation, off-target effect prediction, and gene expression network simulation. These advances are helping to improve therapeutic specificity and enable targeted treatment plans customized to each patient's cancer profile. AI is also optimizing clinical trial design to bring new gene therapies fast into patient care. Further studies will certainly extend the investigations into AI applications in gene therapy strategy optimization as well as improvements in patient-specific treatment planning. Gene circuit design and programmable gene switches are approaches associated with synthetic biology that offer a dynamic means of regulating gene expression in the oncotherapy. These techniques facilitate condition-dependent activation of professed therapeutic genes in conjunction with the detection of certain tumor markers, thus reducing systemic toxicity while enhancing therapeutic efficacy. Owing to developments in synthetic biology, the regulation of cancer gene therapy promises to evolve toward highly controlled and self-sustaining therapeutic agents. Some challenges have to be resolved if the technology is to be used widely in the clinics in due time: scalability and regulatory issues. However, enormous challenges still need to be surmounted for large-scale application of cancer gene therapy. Ethical considerations surrounding germline gene editing, and the legal standing of patient autonomy and access, must heavily temper any movement to safety in clinical application. High pricing of gene therapies is one major impediment because it restricts the access of most patients on a global scale. Simplifying the procedures with which they are manufactured, optimizing regulatory protocols, and bringing into the clinic cheaper treatment approaches will be critical to extend the use of gene therapy. Future studies must concentrate on making gene therapies less expensive and more accessible so that safety and efficacy will not be jeopardized. On the whole, the future for cancer gene therapy is to depend on major technological advances, such as precision genome editing, RNA-based biology, advanced delivery platforms, artificial intelligence incorporation, and synthetic biology breakthroughs. Although issues do arise, those problems can be closed by concerted research and innovations, thus paving the way for gene therapy to become a revolution in the treatment of oncology. Realizing the breakthroughs from present-day challenges by employing contemporary scientific technologies makes it a versatile approach to the treatment of cancer, aimed at providing patients with precise, long-standing, and low-intrusive variants of therapy worldwide
REFERENCE
Pankaj Chasta*, Asrar Ahmad Sheikh, A Review on Future Directions and Emerging Technologies in Cancer Gene Therapy, Int. J. Sci. R. Tech., 2025, 2 (4), 38-44. https://doi.org/10.5281/zenodo.15150472