Call for Abstract

14th International Conference and Exhibition on Advanced Cell and Gene Therapy, will be organized around the theme “Global Collaborations and Future Directions in Gene Therapy”

Cell Therapy 2024 is comprised of keynote and speakers sessions on latest cutting edge research designed to offer comprehensive global discussions that address current issues in Cell Therapy 2024

Submit your abstract to any of the mentioned tracks.

Register now for the conference by choosing an appropriate package suitable to you.

The Evolution of Cell and Gene Therapy  explores the remarkable journey of these fields from their inception to their current status as transformative forces in medicine. This session delves into the historical milestones, pioneering research, and ground-breaking discoveries that have shaped the development of cell and gene therapies. Beginning with the early experiments that laid the foundation for these fields, the session traces the key advancements that have propelled cell and gene therapy forward. From the first successful gene therapy trials to the development of CRISPR-Cas9 technology, attendees will gain insight into the pivotal moments that have defined the evolution of these therapies. Speakers will discuss the challenges and hurdles that researchers and clinicians have overcome, highlighting the perseverance and dedication that have driven progress. The session will also explore the regulatory and ethical considerations that have shaped the landscape of cell and gene therapy, emphasizing the importance of responsible innovation. 

Stem cell therapy has continued to advance, bringing hope to cure diseases that were once considered incurable. The concepts underlying the use of stem cells in therapy depend on their inherent capacity for regenerating the original tissues of the body. Additionally, stem cells can be altered to provide powerful drugs or nanomaterial’s and has an ability to modulate the immune system. Moreover, innovative advances continue in immunotherapy with allogeneic cells and their progress toward clinical use. The T cell immunology field has focused on cytotoxic T lymphocytes, which play an essential role in the immune defence against viral infections and malignancies. The most widely adopted stem cell therapy is the transplantation of hematopoietic stem cells to treat haematological malignancies and disorders of the immune system and blood.  Other ongoing clinical trials involving stem cell therapies have already generated impressive results, such as when patient-derived induced pluripotent stem cells (iPSCs) were induced to differentiate into pigment epithelial cells of the retina when transplanted into patients with macular degeneration, greatly improving the patient's sight. Furthermore, in a recent world-first, iPSC-derived mesenchymal stromal cells have successfully been used to treat patients with acute steroid-resistant graft vs. host diseases.

Gene therapy is emerging as a transformative approach for treating a variety of ophthalmic diseases, offering the potential to correct genetic defects, halt disease progression, and restore vision. Ophthalmic diseases, such as inherited retinal dystrophies, age-related macular degeneration (AMD), and glaucoma, can significantly impact quality of life, and gene therapy provides a promising avenue for long-term treatment and possible cures. Employing RNA interference (RNAi) or antisense oligonucleotides to silence the expression of harmful genes. This is useful for conditions caused by gain-of-function mutations or toxic protein production. Gene therapy research for RP involves various approaches, including gene augmentation, gene editing, and optogenetics, targeting different genetic mutations associated with the disease. Gene therapy for glaucoma focuses on neuroprotection and lowering intraocular pressure. This includes delivering genes that protect retinal ganglion cells from degeneration or enhance the outflow of aqueous humor to reduce pressure.

Regenerative medicine is a broad field that includes tissue engineering but also incorporates research on self-healing – where the body uses its own systems, sometimes with help foreign biological material to recreate cells and rebuild tissues and organs.  The terms “tissue engineering” and “regenerative medicine” have become largely interchangeable, as the field hopes to focus on cures instead of treatments for complex, often chronic, diseases. This field continues to evolve. In addition to medical applications, non-therapeutic applications include using tissues as biosensors to detect biological or chemical threat agents, and tissue chips that can be used to test the toxicity of an experimental medication. Tissue engineering evolved from the field of biomaterials development and refers to the practice of combining scaffolds, cells, and biologically active molecules into functional tissues. The goal of tissue engineering is to assemble functional constructs that restore, maintain, or improve damaged tissues or whole organs.

This session focuses on the latest advancements in immunotherapy for cancer treatment, including novel approaches, clinical trials, Immunotherapy has emerged as a ground-breaking approach in cancer treatment, offering new hope to patients by leveraging the power of the immune system to combat cancer. Unlike traditional treatments that directly target cancer cells, immunotherapy enhances the body's natural defences to identify and destroy cancer cells more effectively. One of the key strategies in immunotherapy is checkpoint inhibitors, which block inhibitory pathways in the immune system that cancer cells exploit to evade detection. By releasing these brakes, checkpoint inhibitors unleash the immune system to mount a more robust attack against cancer.Another promising approach is adoptive cell therapy, such as CAR-T cell therapy, which involves genetically modifying a patient's T cells to recognize and kill cancer cells. This personalized therapy has shown remarkable success, particularly in certain types of blood cancers.

Cell-based therapies have emerged as a promising approach for treating neurological disorders, offering the potential to repair damaged tissues, restore lost function, and improve quality of life for patients. These therapies involve the transplantation or manipulation of cells to replace or support damaged neurons or neural tissue. Stem cells, including embryonic stem cells and induced pluripotent stem cells (iPSCs), have the ability to differentiate into various cell types, including neurons and glial cells. These cells can be used to replace damaged or lost cells in the central nervous system (CNS). Neural stem cells (NSCs) are a type of stem cell found in the CNS that can differentiate into neurons, astrocytes, and oligodendrocytes. Transplantation of NSCs has shown promise in conditions such as Parkinson's disease, spinal cord injury, and stroke. Mesenchymal stem cells (MSCs) are multipotent cells found in various tissues, including bone marrow and umbilical cord tissue. MSCs have immunomodulatory and anti-inflammatory properties, making them a potential treatment for neuroinflammatory disorders such as multiple sclerosis

Ethical considerations in genome editing are paramount due to the profound implications of altering the genetic code of living organisms. Genome editing technologies, such as CRISPR-Cas9, offer tremendous potential for treating genetic disorders, enhancing agricultural productivity, and addressing environmental challenges. However, they also raise ethical questions and concerns that must be carefully considered and addressed. One of the primary ethical considerations is the potential for off-target effects, where unintended changes to the genome could occur, leading to unforeseen consequences. Researchers and clinicians must take precautions to minimize these risks through rigorous testing and validation of genome editing techniques. Another key ethical consideration is the use of genome editing in human embryos or germ line cells, which could result in heritable genetic changes. 

Clinical trials in cell and gene therapy are critical for advancing these innovative treatments from the laboratory to the clinic. These trials are designed to evaluate the safety, efficacy, and potential benefits of cell and gene therapies in patients with various diseases and conditions. Clinical trials in cell and gene therapy are carefully designed to ensure patient safety and to provide meaningful data. They typically follow a phased approach, starting with phase 1 trial to assess safety, followed by phase 2 trials to evaluate efficacy, and finally phase 3 trials to confirm effectiveness and monitor long-term safety. Patients participating in cell and gene therapy trials are selected based on specific criteria, such as the type and stage of their disease, previous treatments, and overall health. Informed consent is a critical aspect of these trials, ensuring that patients fully understand the risks and potential benefits of participating. During clinical trials, patients are closely monitored for any adverse effects or changes in their condition. Data collected from these trials is rigorously analysed to determine the safety and efficacy of the therapy.

Vector development is a crucial aspect of gene therapy, as vectors are used to deliver therapeutic genes into target cells. Vectors can be viral or non-viral, each with its advantages and limitations. Viral vectors, such as lent viruses, adenoviruses, and adeno-associated viruses (AAVs), are often used in gene therapy due to their ability to efficiently deliver genes into cells. However, they can raise safety concerns, such as the risk of immune responses or insertional mutagenesis.Non-viral vectors, such as liposomes or nanoparticles, offer a safer alternative but are less efficient at gene delivery. Vector development aims to optimize vectors for specific applications, such as targeting specific cell types, reducing immune responses, or increasing gene delivery efficiency. This involves modifying the vector's structure, surface properties, or cargo to improve its performance.

Drug delivery strategies for gene therapies are critical for ensuring that therapeutic genes are effectively delivered to the target cells and tissues. These strategies involve the use of various delivery vectors and techniques to enhance the efficiency, specificity, and safety of gene therapy. AAVs are widely used due to their low immunogenicity and ability to deliver genes to both dividing and non-dividing cells. They are suitable for long-term gene expression and have been used in treating genetic disorders like hemophilia and retinal diseases. Lentiviral vectors can integrate into the host genome, allowing for stable and long-term expression of therapeutic genes. They are commonly used in ex vivo gene therapies, such as CAR-T cell therapy for cancer. Adenoviral vectors can deliver large genetic payloads and achieve high levels of gene expression. However, they can provoke strong immune responses, making them more suitable for short-term therapies

Chimeric Antigen Receptor T-cell (CAR-T) therapy represents a revolutionary advancement in cancer treatment, particularly for hematologic malignancies. CAR-T cell therapy involves engineering a patient’s T cells to express a receptor that specifically targets cancer cells, enhancing the body's ability to combat the disease. Numerous clinical trials are ongoing to evaluate the safety and efficacy of CAR-T cell therapy in other types of cancers, including solid tumors. Trials are also exploring combinations of CAR-T therapy with other treatments, such as checkpoint inhibitors, to enhance therapeutic outcomes. Cytokine release syndrome (CRS) and neurotoxicity are significant adverse effects associated with CAR-T cell therapy. Managing these toxicities is critical for patient safety.

Patient advocacy and engagement are critical components of successful clinical trials. They ensure that patient perspectives are integrated into the research process, enhance the relevance and quality of clinical studies, and improve patient recruitment, retention, and outcomes. Patient input can lead to the development of more patient-centered study protocols, including more relevant endpoints, feasible study schedules, and acceptable procedures. Engaged patients are more likely to participate in clinical trials and remain involved throughout the study, leading to more robust and generalizable data. Patient advocacy groups help safeguard patient rights, ensuring that informed consent is thorough and that patients are aware of their rights and the trial's risks 

Cell therapy has emerged as a promising approach for treating cardiovascular diseases (CVDs), offering potential for repairing damaged heart tissue, improving cardiac function, and enhancing patient outcomes. Cardiovascular diseases, including heart failure, myocardial infarction (heart attack), and ischemic heart disease, are leading causes of morbidity and mortality worldwide. Cell therapy aims to address these conditions by using various types of cells to regenerate heart tissue and restore its function. MSCs are multipotent cells derived from bone marrow, adipose tissue, or umbilical cord tissue. They have anti-inflammatory and immunomodulatory properties and can differentiate into various cell types, including cardiomyocytes (heart muscle cells) and vascular endothelial cells. MSCs have been shown to improve cardiac function and reduce scar tissue after myocardial infarction.

Bioinformatics plays a crucial role in the development and optimization of cell and gene therapies. By leveraging computational tools and techniques, researchers can analyze and interpret large datasets, enhance the precision of genetic modifications, and improve the overall efficacy and safety of therapies. Bioinformatics tools are used to predict and minimize off-target effects in CRISPR-Cas9 and other gene-editing technologies. Algorithms can design guide RNAs with high specificity to target genes, reducing unintended modifications. Computational models help design and optimize viral and non-viral vectors for efficient gene delivery. This includes identifying optimal promoter regions, enhancers, and other regulatory elements to ensure high levels of gene expression. Bioinformatics pipelines process and analyze WGS data to identify genetic mutations and variations. This information is crucial for developing personalized gene therapies tailored to an individual’s genetic makeup. RNA sequencing (RNA-seq) data is analyzed to understand gene expression patterns in different cell types and conditions. 

Cell and gene therapies are transforming the landscape of medical treatment, offering hope for curing previously untreatable conditions. However, the manufacturing of these therapies poses significant challenges. Addressing these challenges is crucial for the widespread adoption and commercialization of cell and gene therapies. Cell and gene therapies often involve complex processes such as cell isolation, genetic modification, expansion, and quality control. Each step requires precise conditions and advanced technologies. Scaling up from laboratory to clinical and commercial production is challenging due to the personalized nature of many therapies and the variability of biological materials. Ensuring consistent quality and potency of the final product is difficult due to biological variability and the complexity of the manufacturing process. Robust quality control measures are essential but can be challenging to implement. The manufacturing of cell and gene therapies presents unique challenges due to the complexity and variability of biological systems

The regulatory landscape for cell and gene therapies is evolving rapidly to keep pace with the scientific advancements in these fields. Regulatory agencies worldwide are working to establish frameworks that ensure the safety, efficacy, and quality of these innovative therapies while facilitating their development and approval. The FDA’s Center for Biologics Evaluation and Research (CBER) oversees the regulation of cell and gene therapies. Key regulatory pathways include Investigational New Drug (IND) applications, Biologics License Applications (BLA), and Fast Track, Breakthrough Therapy, and Regenerative Medicine Advanced Therapy (RMAT) designations. The EMA’s Committee for Advanced Therapies (CAT) is responsible for the assessment of advanced therapy medicinal products (ATMPs), including cell and gene therapies. The centralized marketing authorization procedure is mandatory for ATMPs in the EU. In the U.S., a BLA is submitted to the FDA for the approval of biologic products, including cell and gene therapies. This application includes comprehensive data from clinical trials and manufacturing processes. The regulatory landscape for cell and gene therapies is complex and rapidly evolving, reflecting the innovative nature of these treatments. 

Gene editing, particularly with the advent of CRISPR-Cas9 and other advanced technologies, holds transformative potential for public health. It offers unprecedented opportunities to prevent and treat a wide array of genetic disorders, infectious diseases, and even some forms of cancer. However, these advancements also raise significant ethical, social, and regulatory challenges. Gene editing can potentially cure genetic disorders such as cystic fibrosis, sickle cell anemia, and muscular dystrophy by correcting mutations at the DNA level. Gene drives can be used to reduce or eliminate populations of disease-carrying insects, such as mosquitoes, thereby reducing the incidence of diseases like malaria and dengue fever. Gene editing can enable the development of personalized medical treatments based on an individual’s genetic makeup, improving efficacy and reducing adverse effects. Editing the germline (changes that are heritable) raises profound ethical questions about the potential for unintended consequences and the long-term impact on human evolution. Gene editing holds immense promise for improving public health by providing new ways to prevent and treat diseases. However, realizing these benefits requires careful consideration of ethical, social, and regulatory challenges

Gene therapy is emerging as a transformative approach for treating a variety of ophthalmic diseases, offering the potential to correct genetic defects, halt disease progression, and restore vision. Ophthalmic diseases, such as inherited retinal dystrophies, age-related macular degeneration (AMD), and glaucoma, can significantly impact quality of life, and gene therapy provides a promising avenue for long-term treatment and possible cures. Employing RNA interference (RNAi) or antisense oligonucleotides to silence the expression of harmful genes. This is useful for conditions caused by gain-of-function mutations or toxic protein production. Gene therapy research for RP involves various approaches, including gene augmentation, gene editing, and optogenetics, targeting different genetic mutations associated with the disease. Gene therapy for glaucoma focuses on neuroprotection and lowering intraocular pressure. This includes delivering genes that protect retinal ganglion cells from degeneration or enhance the outflow of aqueous humor to reduce pressure

Cellular reprogramming and the generation of induced pluripotent stem cells (iPSCs) represent significant breakthroughs in regenerative medicine, offering potential applications in disease modeling, drug discovery, and cell-based therapies. This field holds promise for treating a wide range of conditions by reprogramming somatic cells to a pluripotent state, enabling them to differentiate into various cell types. Cellular reprogramming involves converting differentiated somatic cells into a pluripotent state, allowing them to give rise to any cell type in the body. This process fundamentally changes the identity of the cell, enabling new therapeutic applications. iPSCs are generated by introducing specific transcription factors into somatic cells, effectively reprogramming them to a pluripotent state. These factors, known as Yamanaka factors (Oct4, Sox2, Klf4, and c-Myc), were first identified by Shinya Yamanaka in 2006. iPSCs can differentiate into specific cell types, such as neurons, cardiomyocytes, and hepatocytes, offering potential for cell replacement therapies in conditions like Parkinson's disease, heart disease, and liver failure.