Call for Abstracts is Open Now!
Deadline for Submission: January 15, 2024
The TERMIS 2024 World Congress will feature an outstanding scientific program bringing together the international community of scientists, clinicians, industry leaders, post-doctoral fellows, graduate students, entrepreneurs, and representatives of government funding agencies engaged in the fields of tissue engineering and regenerative medicine.
Abstract Consideration
Experts from academia, industry and government are invited to present timely research to successful implementation of technologies in all areas of tissue engineering/regenerative medicine.
Note: All abstract submitters including presenting authors are expected to pay their own travel expenses and conference registration fee(s).
Timeline
The below timeline has been put in place for submission, review, and status notifications.
Call for Abstracts Open (Abstracts Accepted) | September 13, 2023 – January 15, 2024 |
Abstract Review Period | January 29, 2024 – February 19, 2024 |
Abstract Notifications Sent | March 11, 2024 |
Categories
Below is a list of the sessions within each category. Abstracts will be submitted to a specific session.
Click on the session title to see the description.
Biofabrication
Biofabrication Strategies for Advanced 3D In Vitro Models
Biofabrication technologies have geared to a level of complexity that today enables the generation of functional human 3D in vitro models that could be used for different purposes, spanning from more fundamental studies to understand pathological mechanisms, to the screening of new therapies and drugs. Examples of such models are already in place for several tissues and organs such as kidney, liver, heart, neuromuscular junctions, bone, and glands. In this special session, we would like to bring together different keynote speakers advancing this field of application of biofabrication technologies, across the 3 continents. The proposal is endorsed by the ISBF as part of the strategic alliance between TERMI and ISBF.
Light-based Bioprinting
In the quest to capture the complex environment of living organs within lab-made tissues,light emerged as a uniquely powerful stimulus for enabling dynamic and spatio-temporal control over cell and biomaterial properties, opening new avenues in regenerative medicineand tissue engineering. Light-responsive moieties permit to non-invasively trigger mechanical actuation and shape-changes in cell-laden constructs, to modulate stiffening or softening of the extracellular milieu, and to enable spatio-temporal control over cell behavior, optogenetics, drug and growth factor release. Moreover, photocrosslinkable and light-sensitive hydrogels are key for bioprinting. Cell-laden building blocks can be sculpted with unprecedented geometrical freedom into high resolution architectures, via stereolithography, multiphoton bioprinting, and volumetric tomographic bioprinting. This symposium offers a forum to discuss i) novel photoresponsive systems and biomaterials, ii) the impact of light stimuli on cell response (including tissue functions beyond viability), and iii) the integration of such discoveries in light-based biofabrication, bioprinting technologies, and regenerative healthcare.
Translational and Clinical Applications of 3D Bioprinting
3D Bioprinting is an emerging technology that aims to enable the fabrication of engineered living constructs via the delivery of cell-laden biomaterials into predetermined 3D positions. While tissue engineering as a field tends to be driven by ambitious long-term objectives such as delivering fully functional tissue or solid organs, it also benefits significantly from 3D bioprinting’s incremental contributions. This session highlights the value of translational and clinical applications that are achievable at steps along the advance of our technical capabilities. This process not only provides vital funding for labs, but it also communicates broadly the value of the tissue engineering paradigm today as a productive means to address specific healthcare and life science challenges. This session will focus on identification and solution of bottle necks in translation of bioprinting to clinical use.
Tissue Engineering and Biomanufacturing Advancements in Space
This session on biomanufacturing in space has the potential to make significant scientific contributions to biotechnology, space exploration, and medical research. By exploring the unique challenges and opportunities associated with biomanufacturing in a space environment, researchers and scientists could uncover new insights and develop innovative solutions that may lead to breakthroughs in medical applications and space technology. The session will bring together experts from various fields to discuss the latest advancements in biomanufacturing technology and explore the potential of these technologies in space. The discussions will focus on stem cell engineering, tissue engineering and regenerative medicine, 3D bioprinting, and space-based manufacturing of pharmaceuticals and other biologics. The symposium will provide a platform for researchers to share their findings and collaborate on new ideas and approaches to advance the field of biomanufacturing in space. Overall, the session has the potential to drive advances in biomanufacturing, leading to the development of new therapies, medical products, and space technologies that could significantly impact society.cription will be added soon
Biofabrication of Complex Tissue Engineering Construct and Spatial Signaling
Spatial signalling cues and growth factor gradients are important during human development for maintaining the stem cell niche and orchestrating tissue repair by regulating cell growth, differentiation and tissue patterning. This session will feature (1) the key role that these spatial signalling cues and gradients play in controlling cell fate and directing new tissue formation, (2) novel modeling platforms to capture the spatial biology of human tissues, including advanced in vivo models from Zebra fish to mice, and (3) in vitro and ex vivo models engineered through the use of advanced biofabrication tools, including 3D bioprinting technologies, for the generation of spatially and hierarchically structured tissue interfaces recapitulating organ complexity. These models are key for opening new avenues to identify complex and integrated functions of biological signals for directing stem cell specification, tissue regeneration and repair. Examples include but not limited to tendon/ligament-to-bone interface, cartilage-bone interface, neuro/vascularized tissues, lung and gastrointestinal system, etc.
Emerging Biofabrication Technologies
Biofabrication involves technologies that aim to create scaffolds for use in tissue engineering, cancer research and as structural reinforcements of constructs. Emerging technologies include, volumetric bioprinting, 4D bioprinting, and multi-materials/multimodal printing that have shown remarkable improvements experimentally in printing efficiency, accuracy, and biomimicry. Melt Electrowriting (MEW) is another high-resolution printing technology that enables the creation of highly complex scaffolds, allowing a better approximation of the natural tissue ultrastructure. Furthermore, MEW can be combined with other biofabrication techniques, such as 3D bioprinting, extrusion 3D printing, and light-based manufacturing, to create unique tissue constructs that surpass the capabilities of each individual technology. This session encompasses a broad range of applications where scaffold fabrication technologies can be employed across various tissue engineering, biofabrication, and 3D cell culture paradigms, and aims to provide a platform for knowledge exchange and fostering new collaborations to advance this important field.
Biofabrication of Complex Tissue Engineering Construct and Spatial Signaling
Spatial signalling cues and growth factor gradients are important during human development for maintaining the stem cell niche and orchestrating tissue repair by regulating cell growth, differentiation and tissue patterning. This session will feature (1) the key role that these spatial signalling cues and gradients play in controlling cell fate and directing new tissue formation, (2) novel modeling platforms to capture the spatial biology of human tissues, including advanced in vivo models from Zebra fish to mice, and (3) in vitro and ex vivo models engineered through the use of advanced biofabrication tools, including 3D bioprinting technologies, for the generation of spatially and hierarchically structured tissue interfaces recapitulating organ complexity. These models are key for opening new avenues to identify complex and integrated functions of biological signals for directing stem cell specification, tissue regeneration and repair. Examples include but not limited to tendon/ligament-to-bone interface, cartilage-bone interface, neuro/vascularized tissues, lung and gastrointestinal system, etc.
Enabling Technologies for Production of Cultivated Meat
Animal agriculture presents major sustainability challenges. Cultivated meat, also commonly called lab-grown meat, addresses the vital concerns associated with the animal agriculture such as the animal welfare, water pollution, food security, antimicrobial resistance, and greenhouse gas emissions. Novel tissue engineering approaches have been used for cellular agriculture and cultivated meat, but challenges in the high cost, scalability, nutritional composition and taste still remain. Novel biomaterials and biofabrication methods specifically tailored for cellular agriculture may provide solutions for the cultivated meat industry. This session will feature the new technological development in the field of cultivated meat such as the development of cost-effective growth media, new bioreactor design, and the development of mammalian, fish, and insect cell lines that can efficiently and sustainably produce meat.
Scale up, Standards and Automation in TERM Biofabrication
Tissue engineering and regenerative medicine therapies have been gearing towards clinical translation, showing safety and efficacy. To provide increased standardization and upscale manufacturing, biofabrication technologies and automation hold the potential to impact the field, on one side by enabling a closer biomimicry of tissue complexity while on the other side controlling and monitoring production. Learning from past failed implementation of automation in the tissue engineering field, we have been able today to produce large-scaled tissues that are currently under clinical trial investigation. In this symposium/session, we aim to present and discuss up-to-date approaches in biofabrication and automation that have resulted in the manufacturing of viable and functional tissue constructs. In parallel, robotic systems have enabled to develop more complex biological constructs. The adoption of these robotic-based biofabrication approaches are starting to surface the TERM field and offer an opportunity to further improve regenerative therapies. We will discuss promises and challenges, spanning from technological development and integration to clinical trial cases.
Software Development, Machine Learning and AI in 3D Bioprinting
The widespread application of 3D bioprinting in tissue engineering has been hindered by challenges in three key areas: the development of biomaterials/bioinks prior to bioprinting, the optimization of the bioprinting process, and the efficient assessment of printed constructs after bioprinting. In response to these challenges, machine learning and relevant software have been developed and integrated with 3D bioprinting to enhance its capabilities across the entire process in tissue biofabrication. This innovative strategy has demonstrated significant potential to reshape the paradigm of 3D bioprinting. The session will cover various topics related to the integration of machine learning/AI and bioprinting, with a particular emphasis on advancing the development of bioinks pre-bioprinting, optimizing the process during bioprinting, and enabling robust evaluation of bioprinted constructs post-bioprinting.
New Biofabrication and Organoid-based Strategies for Regenerative Healthcare – ISBF meets TERMIS
The field of Biofabrication has become an integrated discipline in Tissue Engineering and Regenerative Medicine (TERM) as key-enabling technology to generate highly advanced 3D engineered constructs to better recapitulate the complexity of native tissues. As such, the convergence of Biofabrication, via bioprinting and bioassembly, and TERM is currently expediting clinical translation of engineered tissues by addressing some of the key challenges in the field (i.e., vascularization, scalability, multicellularity). Focusing on these advances, this symposium aims to highlight works on i) new bioprinting and bioassembly technologies, ii) the emergence of multi-technology biofabrication platforms, where multiple techniques (such as extrusion, light-based, magnetic and acoustic), interact together to recreate the native hierarchical composition of native tissues, and iiI) the role of biofabrication techniques in providing geometrical and biochemical cues to guide the self-assembly of cellular aggregates and organoids, and their applications in regenerative medicine. The symposium is also endorsed by the International Society for Biofabrication (ISBF).
Biofabrication as a Method for Development and Implementation of Clinically Useful Products
The session “Biofabrication as a Method for Development and Implementation of Clinically Useful Products” is aimed at showcasing tissue engineering, product development, and clinical application within health care systems adopting biofabrication. The goal of this session is to present the current developments in the field to advance the decentralized personalized medicine at the point of care, for the benefit of patients worldwide.
This Symposium is thought to be useful to researchers, clinicians, or industry partners involved in planning, development or marketing of medical solutions made by tissue engineering or biofabrication methods, intended to be implemented in the clinical site. Any person at any role in the product development pipeline (from early idea stage to regulatory market strategy) should benefit from attending. The session will prioritize topics such as: product development for clinical solutions, scaling-up in manufacturing of personalized products, required regulation or standardization for biofabrication, new technology adoption, collaborative projects, and socially underrepresented patient’s benefit.
Strategies to Improve the Clinical Implementation of 3D-printed Patient-Specific Scaffolds
Clinical translation in tissue engineering, which involves bringing tissue-engineered products and therapies from the laboratory to clinical practice, faces several significant challenges. These challenges can vary depending on the specific tissue or organ being engineered and the intended clinical application. Some common challenges in the clinical translation of tissue engineering include: Biocompatibility: Ensuring that the engineered tissue or implant is biocompatible, meaning it does not trigger an immune response or adverse reactions when introduced into the patient’s body. Achieving this compatibility can be complex, as the tissue must integrate with the recipient’s existing biological systems. Regulatory Approval: Complying with strict regulatory requirements, including demonstrating safety and efficacy, is a major hurdle. Clinical translation often necessitates meeting the standards set by regulatory bodies like the FDA in the United States or the EMA in the European Union. Scale-up and Standardization: Transitioning from small-scale laboratory production to large-scale manufacturing of tissue-engineered products while maintaining quality and consistency is a significant challenge. Developing standardized protocols and quality control measures is vital. Vascularization: For larger and thicker tissue constructs, ensuring adequate vascularization (blood vessel formation) is crucial to supply nutrients and oxygen and remove waste. Achieving this vascularization in engineered tissues is an ongoing challenge. Long-term Functionality: Tissue-engineered products should ideally provide long-term functionality, which can be challenging due to issues like degradation of biomaterials, immune responses, and changes in tissue structure over time. Immunological Compatibility: Immune reactions to the engineered tissue are a potential concern. Strategies to minimize immune responses, such as immune evasion or immunomodulation, are areas of active research. Cost and Scalability: The cost of tissue engineering and the availability of resources can be barriers to clinical translation. Reducing costs and making these therapies accessible to a wider patient population is a significant challenge. Ethical and Societal Considerations: Ethical considerations regarding the source of cells or tissues for engineering, potential harm to donors, and the social acceptance of tissue-engineered products can be obstacles to clinical translation. Long-term Safety Monitoring: Ensuring the long-term safety of engineered tissues after they have been implanted is an ongoing concern. Post-market surveillance and monitoring for potential complications are essential. Clinical Trials: Conducting rigorous clinical trials to evaluate the safety and efficacy of tissue-engineered products can be time-consuming and resource-intensive. Interdisciplinary Collaboration: Effective translation often requires close collaboration between scientists, engineers, clinicians, regulatory experts, and industry partners. Overcoming communication and coordination challenges among these diverse groups can be difficult. Market and Reimbursement: Addressing market acceptance and reimbursement challenges, including convincing healthcare systems and insurance providers of the value of tissue-engineered therapies, is a critical hurdle. This session will address these challenges as advances in tissue engineering hold great promise for regenerative medicine, transplantation, and the treatment of various medical conditions. Overcoming these obstacles often requires interdisciplinary research and collaboration, along with sustained funding and regulatory support.
Organoids, Bioreactors & Organ-on-Chip
Organs-on-Chips and Microbioreactors in Tissue Engineering
The development in advanced technologies such as organs-on-chips (OoCs) and microbioreactors has led to exciting progress in musculoskeletal tissues engineering in the past decade. Musculoskeletal tissues engineered with such advanced technologies have demonstrated a level of tissue architecture, multi-tissue communications, biomechanical microenvironment, and physiological relevance not achievable with conventional in vitro culture methods. This has greatly enhanced our capability in regenerating diseased musculoskeletal tissues and modeling orthopaedic disorders. In this Symposium, the Invited Speaker and other presenters will highlight the latest developments in engineered musculoskeletal tissues using emerging enabling technologies and discuss the potential of such technologies in advancing regenerative medicine, disease modeling, drug discovery, and personalized medicine. As research into artificial musculoskeletal tissues and organs is trending upward and has led to exciting outcomes in scientific research and industry, the Symposium is expected to attract many conference participants in the field of musculoskeletal sciences.
New Biofabrication and Organoid-based Strategies for Regenerative Healthcare – ISBF meets TERMIS
The field of Biofabrication has become an integrated discipline in Tissue Engineering and Regenerative Medicine (TERM) as key-enabling technology to generate highly advanced 3D engineered constructs to better recapitulate the complexity of native tissues. As such, the convergence of Biofabrication, via bioprinting and bioassembly, and TERM is currently expediting clinical translation of engineered tissues by addressing some of the key challenges in the field (i.e., vascularization, scalability, multicellularity). Focusing on these advances, this symposium aims to highlight works on i) new bioprinting and bioassembly technologies, ii) the emergence of multi-technology biofabrication platforms, where multiple techniques (such as extrusion, light-based, magnetic and acoustic), interact together to recreate the native hierarchical composition of native tissues, and iiI) the role of biofabrication techniques in providing geometrical and biochemical cues to guide the self-assembly of cellular aggregates and organoids, and their applications in regenerative medicine. The symposium is also endorsed by the International Society for Biofabrication (ISBF).
Bioassembly and Formation of Spheroids and Organoids for Tissue Engineering
Biofabrication of human organs is still a challenge spite of recent advances in bioprinting approaches. In tissue engineering, the classical approach includes cell suspension seeded onto or into scaffolds, however, the organ microarchitecture is not reproduced. In this context the use of spheroids and organoids emerges. The start-of-art is based on the intrinsic capacity of spheroids and organoids to fuse (building blocks) resulting in organ microarchitecture. The proposal of this session is to bring the light recent studies about self-assembly and maturation of spheroids and organoids and biofabrication approaches to assembly these building blocks (fusion) including the development of bioinks and vascularization units.
Human Myocardium Models to Recapitulate Heart Physiology and Pathology
The symposium aims to bring together leading researchers, scientists, and clinicians in the field of regenerative medicine and cardiac biology. The symposium will provide a platform for in-depth discussions and knowledge exchange on the latest advancements in developing human myocardium models that accurately recapitulate the complex physiology and pathology of the heart. The human heart is a remarkable organ with intricate structure and function. Understanding its physiology and pathology is crucial for advancing treatments for cardiovascular diseases. The symposium aims to share insights into the design, fabrication, and characterization of human myocardium models. These models aim to capture the functional and structural properties of native heart tissue while incorporating disease-specific features. Various approaches, including 3D bioprinting, organoids, and microphysiological systems, will be explored, highlighting their potential to recapitulate heart physiology and pathology in vitro. Moreover, discussions will focus on the integration of advanced imaging techniques, genetic engineering tools, and computational modeling to enhance the fidelity and predictive power of these models. The symposium will provide a platform for participants to exchange ideas, establish new partnerships, and shape the future directions of research aimed at recapitulating heart physiology and pathology using human myocardium models.
Bioassembly for Tissue Engineering and Biofabrication
Bioassembly is recently regarded as a critical alternative biofabrication technical route to bioprinting by the International Society of Biofabrication, as it can directly manipulate millions of live cells to form spatially defined multicellular structures with close intercellular proximity. The close intercellular proximity can improve contact-dependent cell communication and promote the emergence of tissue-specific functions. Various bioassembly techniques have been demonstrated by exploring the interactions between bioparticles and physical fields, such as acoustic, optical, and magnetic fields. Compared to bioprinting, bioassembly takes advantage of high tunability, biocompatibility, and efficiency. Thus, bioassembly has been increasingly used to generate functional tissues from primary and hPSC-derived cells. In this proposed symposium, we invite top experts in the field of bioassembly to present their recent advances in bioassembly and discuss the future direction of bioassembly to construct high-fidelity and transplantable human tissue substitutes.
Cancer
Bioengineered Models of Metastasis
Advances in 3D cell culture technologies, biomaterials, and biofabrication methodologies have led to development of 3D tumor model platforms, such as tumor organoids, engineered 3D tumor constructs, and microfluidic device-based tumor-on-a-chip systems. The inclusion of different cell types, present in the tumor, as well as 3D organization, allow many of these models to be physiologically representative of in vivo tumor and tumor microenvironment (TME) biology, an essential requirement for anti-cancer treatment evaluation ex vivo. These 3D systems can mimic pathological and physical characteristics of tumors, which supports preservation of mutational profiles in cancer cells. Furthermore, they have advantages over animal models, being made from primary human tumor cells and easily controlled and manipulated in the laboratory to attain desired tumor tissue characteristics. Notably, human cancer patients largely do not die from primary tumors, but rather from metastases. In this session, we will highlight cutting edge bioengineered 3D in vitro models of tumor progression and metastasis, including integration of treatment testing and drug development, and personalized chemotherapy and immunotherapy precision oncology. Bioengineered metastasis model technologies continue to develop rapidly for applications in academic, clinical, and pharmaceutical settings.
Models for Investigating Therapeutic Approaches for Cancer
This session will feature advances in engineered tumor models for testing cancer therapeutics, including organoid-based cultures, microfluidic-based platforms and tumor explant cultures. Emphasis will be placed on models that recapitulate different aspects of the tumor microenvironment for the assessment of stromal-targeting drugs (such as immunotherapies).
Use of Biomaterials and Microfluidic Devices for Tissue Engineered Cancer Models
Researchers working in drug development, cancer drug screening platforms, and tissue engineering and regenerative engineering will be able to familiarise themselves with the current methodologies and technologies for developing cancer disease models and early diagnosis. While traditional therapeutic methods may be effective in many cases, they may not be suitable for highly metastatic cancers. When they have already spread and are harder to treat, it exacerbates the challenge of managing this disease. As a result, there is a growing interest in developing complementary tissue-engineered approaches for early cancer diagnosis and treatment to enhance patient recovery. This session will discuss realistic 3D in vitro tumour models for anticancer therapeutic screening. The models will deal with disease microenvironment dynamics, angiogenesis, and cell migration to study and manipulate different cancer treatments. Recent advanced strategies in 3D bioprinting using bio-ink and microfluidic devices allow us to investigate in-depth tumour cell invasion, intravasation, and extravasation) and tissue growth in the 3D microenvironment. A biomimetic biomaterial is expected to provide biochemical and biophysical cues that mimic the in vivo extracellular matrix. This symposium will expose the audience to fabrications of 3D scaffolds and smart hydrogels of synthetic and natural biomaterials, cancer cells and cancer-associated fibroblasts, and 3D cancer cell culture to create disease models. In addition, in vitro tissue-engineered models of the cancer microenvironment and its evolution, preclinical models, engineering of cancer organoids, and cancer lab–on–a–chip for drug development and screening, including the cancer theranostics paradigm. Organizing a session related to treatment constraints will allow us to investigate the cancer microenvironment and innovative treatments deeper using in vitro- and, finally, in vivo models is the demand of the practical uses for cancer therapies through emerging techniques.
Multicellular Models of the Tumor Microenvironment
Recently, it has become increasingly evident that biomechanical, biophysical, biochemical, topographical and cellular components of the cancer tissue environment interact in complex ways, leading to the progression of the disease and affecting the treatment response and the metastatic potential. To that end, cancer models are of paramount importance in understanding the disease mechanisms, in new therapy development as well as for personalised treatment screening. Tissue Engineering has revolutionized the field by enabling the generation of sophisticated and intricate models.
3D in vitro Tissue-engineered Tumor Modelling
Researchers in regenerative medicine, cell and molecular biology, drug screening, and tissue engineering will get an opportunity to learn about the current methodologies and technologies for developing and using biomaterial-based matrices for 3D cellular and cancer disease models. The session will discuss realistic 3D in vitro tumour models for anticancer therapeutic screening and models for disease microenvironment dynamics, including cell migration and metastasis, to study and manipulate cancer and other diseases. Advanced strategies in 3D bioprinting using bio-ink, micro-patterning, and microfluidic devices allow us to investigate in-depth tumour cell invasion, intravasation, extravasation and tissue growth in the 3D microenvironment. Therefore, organizing a session discussing the state-of-art emerging technologies to research cancer and innovative treatments deeper using in vitro models is the demand of the current edge and practical uses for cancer therapies. This symposium will also invite researchers using the latest techniques to fabricate 3D scaffolds and hydrogels using different biomaterials, cancer cells and cancer-associated fibroblasts, and 3D cancer cell culture to create disease models. These innovative approaches to cancer theranostics, mechanism of action, current stage of development, and potential advantages and limitations will provide insights into the next steps of research.
Cardiovascular/Angiogenesis/Blood
Vascularization in Tissue Engineering
A major challenge in engineered tissues with three-dimensional (3D) architecture has been the generation of functional microvasculature to deliver oxygen and nutrients and to remove waste. Strategies include patterning of vessel structures, seeding of cells that promote neovascularization, promotion of angiogenesis from the host, cell self-assembly, and in vivo tissue engineering. Additionally, vascularization is needed to advance 3D in vitro tissue models, which have emerged as valuable tools for longitudinal studies on tissue development, function, and disease progression. This session will focus on new and emerging research to vascularize a variety of engineered tissues both in vitro and in vivo. Studies testing functional interactions between engineered vessels and engineered tissues are especially encouraged. It will also explore cutting-edge research and breakthroughs in the field of vascularization within 3D in vitro models, including strategies for recapitulating vascular complexity, the role of vascularization in tissue development and functionality, and the use of vascularized models in studying diseases and testing therapeutic interventions.
Cardiopulmonary Tissue Engineering
Cardiopulmonary tissue engineering holds tremendous potential for the development of innovative therapies to treat cardiovascular and respiratory diseases. This session aims to highlight the latest advancements in the field, focusing on the use of biomaterials and translational strategies to engineer functional cardiac and pulmonary tissues. The aim will be to highlight cutting-edge research, discuss clinical applications, and address challenges in bringing these transformative approaches to patients.
How to Bridge the Chasm of Scar-free Tissue Healing Towards Personalized Therapeutics?
Per se, all tissues have the endogenous potential for healing. However, the biological potential of endogenous scar-free healing can be different across tissue types and further hampered by comorbidities or if injury size is perturbated beyond a certain limit. Thus, despite years of research, success in steering regeneration by TERM principles to empower in situ repair is still quite limited. Understanding the patient’s healing capacity through novel technologies (e.g., omics or single-cell technologies) opens the possibility of developing novel, personalized therapeutics. While the application of single-type omics or multi-omics approaches has led to identifying distinct cell populations involved in injury repair or disorders, it remains unclear how much this knowledge will help us overcome current medical needs. Scientists are confronted with technological challenges and limitations when robustly applying omics-based approaches to tissue regeneration research or prospectively using them in routine patient care. Once cell fate decisions are better understood, their control through TERM strategies to engineer niches, morphology, and time kinetics of healing cascades to direct spatial and temporal regeneration contains many unsolved challenges. In this special symposium, we intend to discuss novel strategies provided by the speakers and address the remaining limitations in making solutions for scar-free tissue regeneration a reality. We will illustrate the potential and challenges of state-of-the-art analytical technologies to gain a holistic understanding of scar-free healing.
Tissue Engineered Heart Valves and Vascular Grafts
This session will focus on novel designs for heart valves and vascular grafts based on regenerative materials guided by computational analysis and/or knowledge of vascular biology and thrombotic pathways.
Biomimetic In Vitro Models to Advance Cardiovascular Tissue Engineering
New devices and regenerative strategies are being developed for engineering cardiac and vascular tissues, and careful consideration of the complex cell – microenvironment interactions is necessary for these strategies to meet clinical requirements and exhibit long-term viability. Further, new in vitro models that better mimic the in vivo environment are being developed to reduce the number of animals needed for medical devices and regenerative medicine therapies. Therefore, this session will highlight both these more biomimetic in vitro models and the important scientific results obtained with these models to shed light on the orchestrated interplay between cells and matrix that guides cardiovascular tissue formation and maturation. This includes investigating the crosstalk among parenchymal cells and stromal cells, better ways to model the inflammatory response, more biomimetic matrix interactions, and interactions in both physiological and pathological conditions.
Computational Approaches for Tissue Engineering
Computational Modelling for Tissue Engineering
Computational modeling and machine learning can enhance our ability to design and evaluate new tissue engineering strategies for a variety of applications. This is especially important in contexts such as cardiovascular or lung TE, where the myocardium and lungs undergo immense variations in blood pressure and volume, leading to significant variations at cellular level microenvironment that is relevant to any tissue engineered product in vivo. This session will explore computational approaches and tools for tissue engineering, including predictive modeling for bioprinting/biofabrication, evaluating complex data from in vitro and in vivo studies, and predicting the performance of therapeutics in different environments. Examples of these may include fluid mechanics and bio-transport models of drug/protein delivery, patient-specific biomechanical modeling, models of protein-protein and protein-material interactions, statistical modeling for therapeutic optimization, machine learning for the design, quality controlled fabrication and analysis of tissue engineering products, machine learning for histological analysis, and bioinformatics-based platforms for analyzing complex data, including RNA sequencing and other -omics approaches.
Controlled Delivery of Therapeutics for Tissue Engineering
Biomaterials for Controlled Delivery of Therapeutics
This session brings together experts and trainees in the field of biomaterials, tissue engineering, and pharmaceutical science to delve into the latest innovations in advanced strategies for the controlled delivery of therapeutics. With a comprehensive scope, the session will encompass a diverse range of therapeutic modalities, including nucleic acid-based therapeutics, cell-based therapies, small molecules, and more. The primary objective of this session is to showcase recent breakthroughs in the development of micro- and nano-scale carriers that enable controlled and targeted drug delivery. The session will be structured to highlight various types of delivery systems, including: (1)Nanoscale Delivery Systems: Emphasizing lipid nanoparticles and their efficacy in therapeutic delivery. (2) Microscale Carriers: Spotlighting next-generation 3D printed micro- particles and devices designed for encapsulating therapeutic molecules. (3) Emerging Technologies: Showcasing other innovative approaches like hydrogels, ingestible devices, and injection-free systems. Attendees will gain insights into the development of biomaterials, engineered devices, and delivery systems designed to enhance therapeutic outcomes and pave the way for personalized medicine in tissue engineering and regenerative medicine.
Biophysical Stimuli and Stimuli-responsive Materials for Drug Delivery and Tissue Regeneration
The session will describe the most recent advances and will discuss the open issues and future perspectives of biophysical stimulation tools and stimuli-responsive materials. Biophysical stimuli can be exploited for delivering energy in different forms (e.g., light, electrical or magnetic fields, ultrasound, etc.) to cells and tissues, directly promoting their healing/regeneration. Stimuli-responsive hydrogels and stimuli-responsive nanomaterials are further emerging tools that can be exploited to engineer cells and tissues. Indeed, their responsiveness to various physical (temperature, light, electric and magnetic fields, ultrasound), chemical (pH, glucose, and ionic strength), or biological (enzymes and antigens/antibodies) stimuli allows them to amend their configuration, biodegradability, biological moieties, and different physicochemical characters, thus delivering temporal and spatial cues to stem cells, enhancing their regenerative capability over time or enabling innovative targeted therapies. Indeed, the interplay between exogenous energy sources and stimuli-responsive materials can be exploited for releasing drugs and other therapeutic molecules in situ and on-demand, acting in a selective way and minimizing side effects. The session will describe cutting-edge results in this field, exploring the variety of exogenous stimuli for promoting tissue regeneration and drug delivery, in synergy with endogenous ones, and the different materials that can be devised for maximizing the effectiveness of this paradigm.
Gene Therapy Approaches in Tissue Engineering
The key issue of this symposium will be a critical discussion concerning the cutting-edge technology on gene delivery for gene therapy and the establishment of efficient and safe gene delivery in vitro/vivo by a number of new techniques and concepts in targeted or controlled delivery of genes. Also, this symposium highlights the importance of the development of new systems for in vivo delivery of the latest technologies to correct disease-causing mutations.
RNA Therapeutics and Gene Editing for Regenerative Medicine
RNA therapeutics and gene editing are emerging technologies that revolutionize the biotechnology world. The primary objective of this symposium is to bring researchers in the RNA therapeutics and gene editing fields together to present their cutting-edge research for the application of stem cell engineering and tissue regeneration. This symposium welcomes presentations in the fields of CRISPR, CRISPR activation (CRISPRa), CRISPR inhibition (CRISPRi), RNA editing, RNAi, microRNA, long non-coding RNA and all relevant technologies and their applications to control cell differentiation and tissue regeneration.
Dental & Craniofacial
Strategies for Dental & Craniofacial Regeneration
Session description will be added sThis session will feature the state of the art in stem cell and tissue engineering-based research focused towards regenerating dental, oral, and craniofacial structures.oon
Translating Tissue Engineering Advances: From Lab to Bedside in Dental and Craniofacial Applications
Tissue engineering has emerged as a promising field in the realm of dental and craniofacial applications, offering innovative solutions for the regeneration and restoration of damaged or lost tissues with less pain and morbidity. However, the journey from laboratory-based discoveries to practical implementation at the bedside presents numerous challenges that necessitate thorough exploration and discussion. This session aims to go into the intricate process of translating tissue engineering advances from the lab to the clinical setting, providing a comprehensive overview of cutting-edge techniques, biomaterials, and biofabrication methods employed in dental and craniofacial tissue engineering clinical trials. The Key topics of discussion will include scaffold design, cell-based therapies, biomimetic approaches, and the integration of biocompatible materials in conjunction with cells and clinical trials. Additionally, the session will highlight critical factors influencing successful clinical translation, such as regulatory considerations, scalability, and long-term safety and efficacy assessments. Researchers in the field will present their findings, share insights, and engage in thought-provoking discussions, shedding light on the challenges, opportunities, and future directions in translating tissue engineering advances for dental and craniofacial applications. Attendees will gain an understanding of the multidisciplinary nature of this field, fostering collaboration and knowledge exchange among scientists, clinicians, and industry professionals.
Strategies for Enhanced Osseointegration and Prevention of Peri-implantitis
Dental implants are increasingly placed to aid patients that are (partially) edentulous toward regaining chewing function and aesthetics. Different components of dental implants interact with different tissue and require specific surface optimisation to e.g. facilitate osseointegration or prevent bacterial accumulation. This session will highlight the latest innovations in surface modifications for the different components of dental implant systems to improve their longevity.
Advances in Tissue-engineering the Oral Mucosa
The notion that tissue-engineered three-dimensional human constructs provide a more valuable experimental system than monolayer cultures is now widely accepted. Unlike skin, tissue-engineering the oral mucosa poses much more of a challenge as its structure is diverse. The oral mucosa is also subjected to a myriad of diseases from microbial-mediated periodontitis, which is the most prevalent disease in the world to conditions such as oral lichen planus and oral cancer that cause much morbidity and mortality. Here, advanced tissue-engineered models of oral mucosa to mimic these diseases have become a valuable tool not only to decipher molecular mechanisms of pathogenesis but also in the development of novel treatment modalities, novel diagnostic instrumentation and new modes of drug delivery. Furthermore, tissue-engineering the oral mucosa in regenerative medicine has emerged as a translational strategy with a significant role, offering the potential of a revolutionary paradigm shift in tissue replacement and tissue regeneration for dental science. This session will bring together researchers who have led the advancement of oral mucosal constructs and will feature presentations and discussions on cutting-edge research and translational advancements. The primary objective is to foster collaboration and inspire innovative solutions to the challenges in dental soft tissue tissue-engineering. Therefore, the symposium will pave the way for the development of innovative therapeutic strategies in the field, ultimately leading to improvements in clinical outcomes.
Imaging & Assessment
Imaging & Assessment in TERM
This session will highlight research that utilizes the assessment of tissue-engineered constructs’ structure and function through non-invasive imaging techniques. The session aims to explore the applications of advanced imaging modalities such as electron paramagnetic resonance oxygen imaging (EPROI), magnetic resonance imaging (MRI), MR elastography (MRE), computed tomography (CT), single photon emission CT (SPECT), molecular and functional optical imaging techniques, and ultrasound, etc. in evaluating the structure and functional properties of cells and tissue constructs. By highlighting research with non-invasive imaging techniques, this session aims to advance the field of tissue engineering and regenerative medicine by providing valuable insights into characterization, optimization, and tissue growth.
Non-invasive Imaging Applications for Regenerative Medicine and Tissue Engineering
This session will highlight research that utilizes the assessment of tissue-engineered constructs’ structure and function through non-invasive imaging techniques. The session aims to explore the applications of advanced imaging modalities such as electron paramagnetic resonance oxygen imaging (EPROI), magnetic resonance imaging (MRI), MR elastography (MRE), computed tomography (CT), single photon emission CT (SPECT), molecular and functional optical imaging techniques, and ultrasound, etc. in evaluating the structure and functional properties of cells and tissue constructs. By highlighting research with non-invasive imaging techniques, this session aims to advance the field of tissue engineering and regenerative medicine by providing valuable insights into characterization, optimization, and tissue growth.
Emerging Applications of Ultrasound in Regenerative Medicine
We propose a session on the applications of ultrasound in tissue engineering and regenerative medicine. Ultrasound has historically been used in diagnostic imaging and tissue ablation (e.g., kidney stones) but in the past few years, exciting new applications of ultrasound have emerged. For example, the pressure fields that ultrasound creates exerts controlled radiation forces that can remotely pattern cell populations for the assembly of engineered tissues. Ultrasound can also be used to trigger catalysis, instigate gelation, and stimulate cellular processes important for tissue regeneration. Ultrasound has also been used to image gene expression using acoustic reporter genes and harnessed to stimulate site-specific drug and gene delivery, often aided by ultrasound contrast agents (e.g., microbubbles). Our session featuring this burgeoning field will incorporate a keynote talk from a leader with selected abstracts from those communities outlined below.
Immunoengineering
Immunomodulation to Improve Tissue Regeneration
The immune system plays an indispensable role in the process of tissue regeneration following damage as well as during homeostasis. Successful regeneration requires a balanced immune cell response, with the recruitment of accurately polarized immune cells at desired time points in an appropriate quantity. This session comprises research that not only investigates the interaction between the immune system and tissue regeneration and potential molecules required for successful regeneration, but also aims to inhibit, promote, alter, or monitor this interaction to enhance tissue remodeling and regeneration.
In Vitro Models to Study the Immune System and Organ Interactions
This session aims to describe advancements in recent years to include immune-competent models and components in engineering human tissue models. Over the last decade, organs-on-a-chip and engineered tissues have emerged as new tools to study human biology; however, many of these models have not included the complex immune system in their development. This session will highlight recent advances in engineering model systems with integrated immune components, whether as tissue-resident immune populations or infiltrating communicators from circulatory or immune organ models. These immune-competent models may better advance the complexity of tissue engineered systems, as well as help model complex diseases in the body.
Smart Biomaterials for Immunomodulation and Tissue Regeneration
Inflammation control is one of most fundamental parts in biomaterial-based tissue engineering. This is due to the nature of biomaterials as alien objects when recognized by the host immune system and inevitably trigger inflammation. An inflammatory microenvironment around the biomaterial is detrimental for tissue regeneration by hindering tissue cell differentiation and maturation. Ideally, a tissue engineering biomaterial is expected to modulate the local immune environment into the one beneficial for tissue regeneration. This can be achieved by using drug delivery systems to release anti-inflammatory chemicals/biomolecules, however, due to the difference between individuals (e.g., age, gender, and disease conditions such as obesity, diabetes, cancer, immune deficiency, etc.), the basal level of inflammation/immune cell activation varies among individuals, which making it difficult to optimize the drug-loading amount and release timing for each patient. To solve this problem to achieve a personalized immune modulation, recent advances in nanotechnology have endowed drug delivery systems with the capacity of environmental-responsive drug release. Such a “smart material” can sense the levels of inflammatory factors/changes (e.g., pH value, reactive oxidative species, cytokines, surface markers, etc.) and release certain amount of anti-inflammatory factors in response to different inflammatory levels, therefore ensures a personalized immune modulation to optimize the local environment for tissue regeneration. Hence, this proposed symposium aims to summarize the cutting-edge technologies on smart biomaterial design/development for personalized immunomodulation and inflammation control. The program will include 1-2 keynote speakers and 3-4 speakers to discuss the latest advances in this area, which is expected to attract audiences such as scholars in the fields of tissue engineering, regenerative medicine, immunology, material science, clinical practitioners, and biomedical industry partners. It will provide an excellent platform to discuss future strategies for tissue engineering biomaterial development and translation, which is relevant to the conference main topic (biomaterials for the future of healthcare).
Neuro-immuno Modulated Musculoskeletal Tissue Regeneration
The immune system, especially the monocyte-macrophage cell lineage, plays a crucial role in the acute inflammatory response to biomaterial implants and subsequent bone regeneration. These cells can react to environmental signals that regulate bone homeostasis within the tissue microenvironment. Recently, the concept of “interoception” in neuroscience has shed light on how the central nervous system (CNS) controls the body’s internal state, including immune response and bone homeostasis. The CNS receives signals from various physiological systems inside the body, which are transmitted by ascending neural pathways to the brain for processing. The interoceptive information is then sent back to the peripheral organs through descending neural pathways to regulate them. Therefore, this symposium aims to investigate how biomaterials trigger or mediate musculoskeletal tissue regeneration by utilizing these specific pathways.
Musculoskeletal
Scalability of Cartilage and Bone Tissue Production: Where Size Matters
Cartilage and bone defects present significant clinical challenges, driving the need for effective regenerative strategies. Recent progress in tissue engineering technologies offers the opportunity to generate large three-dimensional (3D) constructs. To generate centimeter-scale tissues, several scalability challenges arise: expanding billions of cells, generation and maintenance of complex shapes, nutrient supply, intense manual labor, and reproducibility. This symposium will cover key approaches to dealing with scalability of 3D tissue production, particularly bone and cartilage. Emerging technologies that address scalability include biofabrication, robotics, bioreactor systems, scaffold/construct design, and vascularization. A special emphasis will be on bottom-up approaches that use microtissue building blocks to realize large structures. In conclusion, upscaling microtissue production and controlling their assembly into large complex structures holds immense potential in regenerative medicine.
Bone Tissue Engineering
Bone tissue engineering can provide transformative solutions to a wide range of musculoskeletal disorders, from fractures and bone defects to degenerative diseases like osteoporosis and osteoarthritis, ultimately improving patients’ quality of life. In this symposium, we cover recent advances in various aspects of bone tissue engineering, from the development of advanced biomaterials, such as biodegradable scaffolds and hydrogels, to biomimetic functional interfaces and micro/nano-structured coatings. The symposium also highlights personalized implant solutions, such as those created through additive manufacturing techniques, including 3D bioprinting. Other topics of interest include, but are not limited to, bioinspired constructs, vascularization strategies, mechanical stimulation, in vitro models for evaluating engineered bone tissue constructs, and clinical translation considerations, including the development of regulatory pathways for bone tissue engineering products.
Cartilage and Osteochondral Tissue Engineering
Osteoarthritis is a degenerative joint disease, typified by the loss in the quality of cartilage and bone at the interface of a synovial joint, resulting in pain, stiffness and reduced mobility. The current surgical treatment for advanced stages of the disease is joint replacement, where the non-surgical therapeutic options or less invasive surgical treatments are no longer effective. The objective of this symposium is (1) to discuss the current regenerative treatment options for cartilage and osteochondral (cartilage and underlying subchondral bone) defects in the articulating joints; (2) to report the scaffolds clinical requirements and performance of the currently available osteochondral scaffolds that have been investigated both in animal studies and in clinical trials; (3) to identify the main hurdles in osteochondral scaffold development for achieving satisfactory and durable regeneration of osteochondral tissues. The evolution of the osteochondral scaffolds – from monophasic to multiphasic constructs – will be included and the osteochondral scaffolds that have progressed to clinical trials will be examined with respect to their clinical performances and their potential impact on the clinical practices in this symposium.
Skeletal Muscle Bioengineering and Volumetric Muscle Loss (VML) Repair
Volumetric muscle loss (VML) is the loss of muscle tissue which exceeds the body’s capacity for self-repair, resulting in impaired muscle function, and in many cases, physical deformity. While many musculoskeletal traumas may be endogenously repaired over time through the myogenic potential of satellite cells, more severe cases of VML overwhelm this native repair mechanism, creating a need for intervention. To date, the treatments for VML including the muscle flap or graft are not effective and are hindered by limited tissue availability and donor site morbidity. Thus, there is a critical need for new technologies to address VML. There are several labs that are diligently working on scaffold and cell-based technologies for engineering skeletal muscle to address the unmet need for a novel intervention to repair VML. This symposium will inform the participants about promising tissue engineering strategies currently being deployed both in vitro and in vivo that, in aggregate, have tremendous potential to identify improved strategies for the repair of volumetric muscle loss.
Neuro-immuno Modulated Musculoskeletal Tissue Regeneration
The immune system, especially the monocyte-macrophage cell lineage, plays a crucial role in the acute inflammatory response to biomaterial implants and subsequent bone regeneration. These cells can react to environmental signals that regulate bone homeostasis within the tissue microenvironment. Recently, the concept of “interoception” in neuroscience has shed light on how the central nervous system (CNS) controls the body’s internal state, including immune response and bone homeostasis. The CNS receives signals from various physiological systems inside the body, which are transmitted by ascending neural pathways to the brain for processing. The interoceptive information is then sent back to the peripheral organs through descending neural pathways to regulate them. Therefore, this symposium aims to investigate how biomaterials trigger or mediate musculoskeletal tissue regeneration by utilizing these specific pathways.
Regenerative Strategies for Fibrous Connective Tissue Healing
Injuries and disorders of fibrous connective tissues, such as tendons, ligaments as well as their junctions with bones and muscles, represent one of the major reasons for practice consultations for musculoskeletal pain, impaired function and often require the need of surgery. Both, the general population and athletes are affected resulting in enormous related healthcare costs. Moreover, their treatments are often prone to long rehabilitation times, incomplete functional recovery, and secondary complications after surgery. This session will furnish an overview of the scientific discoveries in the area of these fibrous tissues pathology, mechanobiology, adaptive and innate immune responses to design innovative therapies such as tissue engineering scaffolds for their more effective structural and functional regeneration. The keynote will address the broad field of fibrous tissues regenerative approaches using biomaterials, cells, and morphogens. The abstracts will be chosen such that a broad spectrum of regenerative solutions is represented.
Advances and Challenges of Tendon Engineering and Regeneration
Tendon injuries are among the most common musculoskeletal diseases, which can lead to significant functional impairments. Recent advances in tendon regeneration and engineering have shown promising results in improving healing outcomes and restoring tendon functionality. However, challenges persist in achieving optimal tendon regeneration. The complex hierarchical structure of tendons, with aligned collagen fibers and tendon-to-bone integration, remains difficult to replicate. Additionally, the mechanical properties and functionality of engineered tendons often fall short compared to native tissues. Importantly, the complex cellular and molecular mechanisms of tendon healing are far from being completely understood. Addressing these challenges requires interdisciplinary collaborations among scientists, engineers, and clinicians. This thematic session will bring experts from different fields and foster discussion on further research needed not only to better understanding the underlaying mechanism of tendon diseases, but also to optimize biomaterial design, cellular therapies, and tissue engineering techniques to overcome these hurdles and enable successful tendon regeneration and functional recovery.
Neural & Spine
Neuroengineering Strategies in TERM
Modeling and regeneration of the nervous system remains a unique challenge for biomedical scientists and engineers stemming from need to replicate or restore intricate architecture and functional complexities. This session will highlight recent advances in neural tissue engineering approaches for the peripheral and central nervous systems, including innovations at the convergence of advanced biomaterials, stem cell technologies, and innovative fabrication techniques. Discourse is intended to unveil novel avenues in addressing challenges posed by neurodegenerative diseases and neural traumas, paving the way for new therapeutic strategies.
Regenerative Rehabilitation – Combining Regenerative Medicine with Rehabilitation Approaches for Spinal Cord Injury
There are two separate approaches for promoting functional recovery after spinal cord injury (SCI): regenerative medicine and rehabilitation. However, each approach individually has not yet resulted in full functional recovery after SCI, specifically after the most severe injuries. There is a need for synergistic combination of both approaches to maximize functional recovery after SCI by first regenerating damaged spinal cord tissue and then promoting functionality. This session will focus on introducing regenerative rehabilitation for SCI and highlight research that integrates the two approaches for enhanced functional recovery after SCI. We propose this symposium session to encourage collaboration between regenerative medicine and rehabilitation disciplines for SCI. The proposed session will have 2 invited speakers: Dr. Sing Yian Chew, Nanyang Technological University, Singapore, and Dr. Anthony Windebank, Mayo Clinic (each 30 min, 25+5 min). Then there will be 2 speakers from selected abstracts (each 15 min, 12+3 min). Dr. Chew has confirmed and we have another invite sent out to Dr. Windebank. Although, if either of the invited speakers were to drop out, we would increase from 2 to 4 speakers for the 15-minute presentations.
Functionalized Biomaterials for CNS and Imaging
CNS diseases pose a particular therapeutic challenge, as their outcomes are trailing the improvements in the treatments of other disorders. Therefore, there is an urgent need and unique opportunity for more advanced solutions for CNS diseases, and biomaterials such as nanoparticles, hydrogels, and others can contribute to filling this therapeutic gap. However, CNS is hidden behind the blood-brain barrier; therefore, traditional methods of therapy monitoring through blood testing and low-cost optical and sonographic have limited applicability. Consequently, we will focus on tissue permeable imaging systems such as MRI and PET/CT. The symposium will include a variety of labeling approaches for biomaterials. This effort will be well aligned with image acquisition methods, consisting of not only labeled biomaterials, but also advances in imaging methods capable of capturing the inherent contrast of selected biomaterials. Chemical exchange saturation transfer (CEST) is an excellent MRI method based on the physicochemical properties of biomaterials.
Intervertebral Disc Regeneration
Cellular Therapies and non-cellular therapies including extracellular vesicles (EVs/exosomes) carrying micro (mi)-RNA are on the rise. This seminar focuses on cellular and non-cellular regeneration approaches or a combination thereof. The intervertebral disc (IVD) is a difficult target with limited number of cells, which are adapted to high pressure together with low glucose, oxygen and pH. Here, it is exciting to see how advanced cell therapy or novel acellular approaches could overcome this harsh microenvironment and be used to restore IVD metabolism and mechanical properties.
Innervating Tissue Engineered Models for Health and Disease
Advances in tissue engineering have contributed to the development of highly sophisticated models that recapitulate the structural and functional aspects of human tissues. However, the integration of innervation into these models remains a critical aspect that requires further attention. This session will highlight the importance of innervation for tissue function, discuss cutting-edge strategies for incorporating neural networks into tissue engineered constructs, and shed light on the promising applications of innervated tissue models for neurodegenerative diseases, cancer progression, wound healing, and/or organ regeneration. Importantly, the session will bring together multidisciplinary experts at multiple career stages to foster collaborative discussions on next-generation tissue models.
Combining Different Approaches for Translational Nerve Tissue Engineering and Regeneration
Damage to peripheral nerves can cause debilitating consequences to patients such as lifelong pain and disability. At present, the only treatment option is microsurgical and even in the best-case scenario, recovery is still incomplete and unsatisfactory. Worldwide, research groups are using multidisciplinary approaches to develop therapies to repair peripheral nerves. The aim of this session is to bring these researchers, clinicians, and industry partners together to disseminate the current advancements in the field, build networks and ignite discussions and international collaborations. Within this session the research presented will span a broad range of disciplines ranging from biomaterials, model development, drug therapeutics and cell and gene-based therapies; all-encompassing the tissue engineering and regenerative medicine focus of the TERMIS meeting. We would also like to use this session to highlight the clinically translatable research that is being conducted across labs and what the future holds for this field.
Ophthalmologic
Ophthalmic Tissue Regeneration and Bioengineering
The human eye consists of tissues that have distinct and unique properties that is distinct from other human tissues, for example cornea and lens are the only transparent tissue in our body, and retina being a thin layer consists one of the most complex layers of neuron and non-neuron cell network. A wide spectrum of modern bioengineering technique has been researched and developed to reconstruct/regenerative these unique ophthalmologic tissues including using electronic stimulus for neuron regeneration in retina, new transparent biomaterials and scaffolds for corneal transplantation, in-situ printing for ocular wound healing, microfluidic technologies for drug delivery and organ on chip, and technologies on reconstruction of the whole eye. This session focuses on showcasing emerging technologies applied to overcome challenging questions faced in ophthalmologic research and clinical applications such as corneal transplantation, treat retinal degeneration, new drug/cell delivery methods for ocular diseases and many more.
Tissue Engineering Approaches in Ocular Regenerative Medicine
This session will bring together experts working in the field of ocular regenerative medicine. Speakers will present cutting edge research in tissue, cell, and genome engineering designed to prevent vision loss and restore sight to the blind.
Respiratory, Urologic & Gastrointestinal
TERM x Delivery: Breaking Barriers in Therapeutic Delivery to Mucosal Tissues
Respiratory, gastrointestinal, and urogenital organs all contain mucosal surfaces, making them unique access points for therapeutic delivery both locally and for systemic absorption. No matter the therapeutic — cells, exosomes, micro- and nanoparticle carriers, drugs and biologics, gene therapeutics — all are up against similar barriers to successful delivery: mucus layer turnover, epithelial tight junction integrity, surface area distribution, scalability. This session will cover fresh solutions to the common delivery problems faced by all those who seek to engineer and treat mucosal surfaces. With a scope including a diversity of developments, from cell replacement therapy to enhancing gene delivery, we hope to expose common paradigms of delivery problems and solutions. We welcome submissions from those developing regenerative medicine therapeutics for mucosal surfaces as well as those studying drug delivery via tissue-engineered platforms. Together, we will generate new insights at the intersection of TERM x Delivery.
Engineering and Regeneration of Airway, Digestive, or UroGenital Systems
In this session, we will be discussing recent advances in the tissue engineering and regenerative medicine of the airways and of the digestive and urogenital systems, identifying key lessons that could be leveraged across structures and functions, while highlighting the unique challenges of each physiological environment.
Biopreservation Technologies for Extended Storage of Organs and Tissues
Biopreservation is an essential process for storage, transport and distribution of tissue engineered products that will facilitate widespread application of the end product. The process involves keeping biospecimens at low temperatures to reduce or suspend their physiochemical and metabolic activities and then recovering them to physiological states at a desired future time. Biopreservation has been critical in a number of areas of regenerative medicine such as reproductive cells and tissues for assisted reproductive technology, tissues and organs for transplantation, or stem cells, blood cells or genetically engineered cells for cell based medicine. The goal of this session is to bring together investigators who focus their work in studying biopreservation for extended storage of tissue engineered products (e.g., organoids, microphysiological systems, tissues and whole organ constructs) including but not limited to respiratory, urologic and gastrointestinal systems. Topics of interest include cooling and rewarming technologies, strategies for protection of biospecimens from cold injury, biology at cold temperatures and other related subjects.
Scaffolds, Matrices, and Biomaterials
Sustainable Materials in TERM
Tissue engineering faces challenges in translation of biomaterials into 3D constructs that can mimic the physical, mechanical, chemical, and biological features of native tissues. Some of the traditional approaches are sophisticated and involve extensive material processing and expensive fabrication procedures. While there has been significant success in biomaterials discovery, functional and manufacturing limitations have led to the innovation of novel biomimetic materials that we can borrow from nature and human-made commodities. This session will explore tissue engineering strategies that involve unconventional biomaterials for improved scalability, sustainability, cost-efficiency, and functionality. Unconventional biomaterials are obtained from globally accessible resources and can serve across a range biomedical applications. Using non-traditional materials such as plants, silk, paper, eggshells, textiles, marine organisms, and edible products can provide unique solutions to existing challenges. With the increased use of abundant and sustainable resources, tissue engineering technologies can reach a global scale.
Bioelectronics for Studying Cell/Material Hybrid Interfaces
Achieving well-tolerated and chronic biological interfaces remains an ongoing challenge for the deployment of biomedical and bionic devices. The field of tissue engineering and regenerative medicine offers a potentially powerful approach to overcoming or controlling host foreign body reactions and subsequent degradation of device functionality, via improved integration between the ‘bio’ and ‘electronic’. By leveraging our understanding of how to design material systems that can support cell growth and function, we can begin to pursue seamless integration across the abiotic/biotic interface. Our proposed invited speaker, Dr Damiano G. Barone is well placed to speak to the integration of cell-based functional devices as a neurosurgeon and clinical lecturer who has recently published on a hybrid flexible electrode array cultured with induced pluripotent stem cells to form a living interface. It is hoped that this session highlights how our understanding of tissue engineering and regenerative medicine could be used as a toolbox and promote discussion into what challenges need to be overcome in order to translate these transformative technologies to the clinic.
Strategies to Improve the Clinical Implementation of 3D-printed Patient-Specific Scaffolds
Clinical translation in tissue engineering, which involves bringing tissue-engineered products and therapies from the laboratory to clinical practice, faces several significant challenges. These challenges can vary depending on the specific tissue or organ being engineered and the intended clinical application. Some common challenges in the clinical translation of tissue engineering include: Biocompatibility: Ensuring that the engineered tissue or implant is biocompatible, meaning it does not trigger an immune response or adverse reactions when introduced into the patient’s body. Achieving this compatibility can be complex, as the tissue must integrate with the recipient’s existing biological systems. Regulatory Approval: Complying with strict regulatory requirements, including demonstrating safety and efficacy, is a major hurdle. Clinical translation often necessitates meeting the standards set by regulatory bodies like the FDA in the United States or the EMA in the European Union. Scale-up and Standardization: Transitioning from small-scale laboratory production to large-scale manufacturing of tissue-engineered products while maintaining quality and consistency is a significant challenge. Developing standardized protocols and quality control measures is vital. Vascularization: For larger and thicker tissue constructs, ensuring adequate vascularization (blood vessel formation) is crucial to supply nutrients and oxygen and remove waste. Achieving this vascularization in engineered tissues is an ongoing challenge. Long-term Functionality: Tissue-engineered products should ideally provide long-term functionality, which can be challenging due to issues like degradation of biomaterials, immune responses, and changes in tissue structure over time. Immunological Compatibility: Immune reactions to the engineered tissue are a potential concern. Strategies to minimize immune responses, such as immune evasion or immunomodulation, are areas of active research. Cost and Scalability: The cost of tissue engineering and the availability of resources can be barriers to clinical translation. Reducing costs and making these therapies accessible to a wider patient population is a significant challenge. Ethical and Societal Considerations: Ethical considerations regarding the source of cells or tissues for engineering, potential harm to donors, and the social acceptance of tissue-engineered products can be obstacles to clinical translation. Long-term Safety Monitoring: Ensuring the long-term safety of engineered tissues after they have been implanted is an ongoing concern. Post-market surveillance and monitoring for potential complications are essential. Clinical Trials: Conducting rigorous clinical trials to evaluate the safety and efficacy of tissue-engineered products can be time-consuming and resource-intensive. Interdisciplinary Collaboration: Effective translation often requires close collaboration between scientists, engineers, clinicians, regulatory experts, and industry partners. Overcoming communication and coordination challenges among these diverse groups can be difficult. Market and Reimbursement: Addressing market acceptance and reimbursement challenges, including convincing healthcare systems and insurance providers of the value of tissue-engineered therapies, is a critical hurdle. This session will address these challenges as advances in tissue engineering hold great promise for regenerative medicine, transplantation, and the treatment of various medical conditions. Overcoming these obstacles often requires interdisciplinary research and collaboration, along with sustained funding and regulatory support.
Functionalized Biomaterials for Tissue Engineering Applications
Session description will be The use of biomaterials as therapeutics has emerged as a revolutionary approach within the field of tissue engineering, aiming to highlight their significant role in advancing regenerative medicine and accelerating clinical applications. In this session, we explore the design, fabrication, and functionalization of biomaterials, with a focus on enhancing their therapeutic potential and promoting tissue regeneration.added soon
Advances in Decellularization and Recellularization of Complex Tissues
Decellularization and recellularization techniques have emerged as powerful tools in tissue engineering, enabling the creation of complex functional tissues for transplantation and regenerative medicine applications. This session aims to explore the latest advancements and breakthroughs in decellularization and recellularization strategies, focusing on their application in the development of complex tissue constructs.
Naturally Derived Biomaterials for Sustainable TERM
Session descriptResearchers working using natural biomaterials for tissue engineering and regenerative medicine, drug screening and drug delivery will get an opportunity to learn about the current methodologies and techniques for developing sustainable, innovative scaffolds and biomimetic-based matrices for cell culture, including hydrogels. This will include novel 3D cell culture models that are aimed at improving the patient’s quality of life. Sustainable natural-based biomaterials will be investigated to further develop different 3D regeneration models for therapies and disease screening in microenvironment dynamics, including nanomaterial fabrications. These latest advances will provide a means to engineer the in vitro (and even in vivo) cellular microenvironment and cell migration to study and manipulate various hard and soft tissues. Advances made with 3D (bio) printing using hydrogel as bio-ink, micropatterning, and microfluidic devices for the study of tissue growth, stem cell differentiations for soft- and hard tissue engineering, including optical and bioelectronic properties are also within the scope of the session. Despite certain advances, developing matrices with good electrical and mechanical properties in physiological environments remains challenging. The presentations will discuss the emerging techniques to better understand cell-based tissue engineering and regenerative medicine methods in vitro and/or in vivo platforms. This symposium will explore the designing of sustainable natural biomaterials for different tissue regeneration processes.ion will be added soon
Animal Free Biomaterials for TERM
One of the key engineering challenges in TERM is the development for fully defined and ethical (non-animal derived) technologies. These range from biomaterials such as scaffolds to cell culture media . These technology underpin a growing share of TERM research and industries worldwide. The current over-reliance on poorly defined animal derived products such as Engelbreth-Holm-Swarm (EHS) mouse sarcoma solubilized basement membrane matrices or fetal bovine serum is creating a significant challenges for TERM community in term of clinical translatability and experimental reproducibility. The worldwide push toward ethical research has created a environment conducive to the growth of anima free technologies for TERM applications. This session will bring together people working in this field and promote awareness of animal free technologies to the wider TERM community to promote uptake and development.
Injectable Scaffolds
The development of injectable hydrogel scaffolds has been a focus of research in tissue engineering and regenerative medicine. These scaffolds offer unique advantages such as ease of application, site-specific action, and improved patient compliance. Injectable scaffolds also provide a uniform distribution of cells within the scaffold, creating an environment that promotes tissue regeneration. Despite significant advancements, there are still challenges in the design and development of injectable scaffold-cell systems. These include optimizing the solidification process, maintaining a conducive environment for tissue growth under mechanical stress, controlling drug/growth factor release, ensuring biocompatibility and cell viability during scaffold preparation and delivery, and understanding cell-material interaction. To address these challenges, a symposium focused on injectable scaffolds would be an ideal fit for the conference, connecting researchers from synthetic chemists to applied scientists, engineers, and clinicians. Injectable hydrogel scaffolds, formed by in situ crosslinking of aqueous gel mixture precursors, have emerged as promising biomaterials for tissue engineering applications. The symposium will cover recent advances in the development of injectable hydrogels for biomedical applications, highlighting their advantages over conventional pre-fabricated hydrogels, such as reduced cell damage, immune protection, easy manipulation, and minimal invasive procedures.
Smart Biomaterials for Immunomodulation and Tissue Regeneration
Inflammation control is one of most fundamental parts in biomaterial-based tissue engineering. This is due to the nature of biomaterials as alien objects when recognized by the host immune system and inevitably trigger inflammation. An inflammatory microenvironment around the biomaterial is detrimental for tissue regeneration by hindering tissue cell differentiation and maturation. Ideally, a tissue engineering biomaterial is expected to modulate the local immune environment into the one beneficial for tissue regeneration. This can be achieved by using drug delivery systems to release anti-inflammatory chemicals/biomolecules, however, due to the difference between individuals (e.g., age, gender, and disease conditions such as obesity, diabetes, cancer, immune deficiency, etc.), the basal level of inflammation/immune cell activation varies among individuals, which making it difficult to optimize the drug-loading amount and release timing for each patient. To solve this problem to achieve a personalized immune modulation, recent advances in nanotechnology have endowed drug delivery systems with the capacity of environmental-responsive drug release. Such a “smart material” can sense the levels of inflammatory factors/changes (e.g., pH value, reactive oxidative species, cytokines, surface markers, etc.) and release certain amount of anti-inflammatory factors in response to different inflammatory levels, therefore ensures a personalized immune modulation to optimize the local environment for tissue regeneration. Hence, this proposed symposium aims to summarize the cutting-edge technologies on smart biomaterial design/development for personalized immunomodulation and inflammation control. The program will include 1-2 keynote speakers and 3-4 speakers to discuss the latest advances in this area, which is expected to attract audiences such as scholars in the fields of tissue engineering, regenerative medicine, immunology, material science, clinical practitioners, and biomedical industry partners. It will provide an excellent platform to discuss future strategies for tissue engineering biomaterial development and translation, which is relevant to the conference main topic (biomaterials for the future of healthcare).
Harnessing Peptide and Protein-based Materials for Tissue Engineering
Bio-molecular self-assembly represents a simple and efficient route to the construction of large, complex structures across multiple length scales. This sparked evolution of numerous creative approaches for designing functional biomaterials in the tissue engineering realm, particularly from the peptide and protein-based building molecules. Specifically, this symposium will focus on the significant progress that has been made towards understanding the design rules, mechanisms and driving forces behind the design and hierarchical assembly of peptides and proteins and derivatives thereof, bringing a set of new biomaterials revolutionizing tissue engineering. Through precise molecular modifications using natural and non-natural amino acids, biomaterials spanning controlled release systems, new matrices for organoids growth and 3D cell culture, as well as components of miniaturized chips are developed.
Scaffold Functionalization with Biomimetic Factors for TERM Applications
The extracellular matrix, an assembly of multiple types of factors and molecules that surround cells in vivo, plays an important role in regulating cell functions and tissue regeneration. Highly bioactive scaffolds have been designed and developed to mimic the functions of extracellular matrices. So far, hybridization of organic/polymer compounds and inorganic compounds, immobilization of cell growth factors and proteins, grafting of biofunctional polymers, and incorporation of functional nanoparticles and bioactive molecules have been performed. In particular, growth factors and biofunctional polymers have been tethered or grafted to scaffolds by a variety of physicochemical, genetic and biological approaches. In this symposium, these approaches and functionalized scaffolds will be highlighted and discussed for their applications in stem cell research and TERM applications.
Next Generation Biomaterials for Stem Cell Culture, Differentiation and Therapy
There is a shortage of tissues and organs for patients who suffer damage or loss of their tissues and organs. Stem cells hold promise for drug discovery and regenerative medicine. We would like to propose the symposium of “Next Generation Biomaterials for Stem Cell Culture and Differentiation for Stem Cell Therapy”. The development of a fully defined microenvironment for culturing and differentiating human stem cells will have a great effect on the use of stem cells in cell therapy and tissue engineering. In this symposium, we will discuss the design and strategy of biomaterials for stem cell culture and differentiation. Especially, we will discuss biomaterial design and preparation that can not only maintain the pluripotency and stemness of human stem cells, but also guide differentiation of stem cells (adult stem cells, fetal stem cells, human embryonic stem cells and human induced pluripotent stem cells) into specific lineages of cells (osteoblasts, chondrocytes, adipocytes, mesenchymal stem cells (from human pluripotent stem cells), hematopoietic stem cells (from human pluripotent stem cells), retinal pigment epithelium (from human pluripotent stem cells) and cardiomyocytes). We will further focus on the effects of physical cues (elasticity, micropatterning, electrical field and mechanical force), together with the biomaterials used in clinical application using stem cells and the translational development of stem cell therapies in various catastrophic illnesses.
Innovative Cell Instructive Biomaterials for Regenerative Medicine
Understanding the interaction between cells and extracellular environment is a fundamental prerequisite in order to engineer functional biomaterial interfaces able to instruct cells with specific commands. Engineered cell instructive microenvironments with the ability to stimulate specific cellular responses are a topic of high interest in the fabrication and development of biomaterials for application in tissue engineering. Cells are inherently sensitive to the in vivo microenvironment that is often designed as the cell “niche.” The cell “niche” comprising the extracellular matrix and adjacent cells, influences not only cell architecture and mechanics, but also cell polarity and function. Indeed, Such advanced biomaterials might find relevant application in prosthesis design, tissue engineering, diagnostics and stem cell biology. As a consequence extensive research has been performed to establish new tools to fabricate biomimetic advanced materials for tissue engineering and regenerative medicine, that incorporate structural, mechanical, and biochemical signals that interact with cells in a controlled manner and to recapitulate the in vivo dynamic microenvironment. As a consequence of this bioactive tunable microenvironments using micro and nanofabrication have been successfully developed and proven to be extremely powerful to control intracellular signaling and cell function. The present symposium aims at discussing the most recent findings on material-induced cell responses, with a particular emphasis on their impact in tissue engineering and regenerative medicine of different tissues.
Biomaterials for Women’s Health Engineering
Women have historically been excluded at all levels of biomedical research and medical device innovation which has led to a gaping hole in the healthcare landscape to meet even the most basic needs in women’s health. To address this critical need, we need innovative biomaterial solutions to expand health technologies available to clinicians to treat women. This session will highlight advances in biomaterial approaches to address the myriad of clinical needs in women’s health. Topics include disorders in reproductive tissues (e.g. endometriosis, cancer, premature ovarian insufficiency, infertility, preterm birth), multicellular tissue-level interactions and mechanics in reproductive health, extracellular matrix dynamics and sex-related differences in chronic diseases and device design, gynecological device design, and breast cancer and reconstruction.
Bridging Gene Therapies and Implants for Tissue Repair: Gene-Activated Scaffolds in Regenerative Medicine
The challenges of regenerative medicine, such as the repair of complex tissues following disease or degeneration, require innovative solutions. This symposium aims to shed light on a growing approach for complex conditions, which combines gene therapies and biomaterial scaffolds. Gene-activated scaffolds, designed with the capacity for local, targeted delivery of genetic cargoes (including mRNA, microRNA, or pDNA), offer a potential revolutionary strategy to enhance tissue repair. Unlike growth factors and recombinant proteins, these scaffolds do not require supraphysiological dosages, as, if designed appropriately, the scaffolds can act as reservoirs for the nanomedicine, reducing the risk of adverse effects and enhancing the regenerative capacity of the scaffold.
Tissue Engineering Scaffolds in Regenerative Medicine
Tissue engineering scaffolds – that mimic the native extracellular matrix- play a vital role in providing an environment that facilitate cellular growth, differentiation, and maturation. Moreover, conjugating biomolecules and drugs to scaffolding materials actively influences cellular responses. However, designing clinically relevant scaffolds in a congenial and sustainable approach is still a critical engineering challenge. Moreover, we know little about how the mechanical properties of the scaffolds regulate various cellular processes such as cell division, stem cell differentiation, and cancer progression. This symposium will highlight cutting-edge advances in scaffold design to modulate cellular and biological outcomes. Efforts to explore the multifaceted challenges associated with the clinical translation and commercialization of tissue engineering scaffolds for regenerative medicine will also be featured. We invite experts worldwide to submit contributions that provide novel findings in scaffold-based tissue engineering and regenerative medicine.
Regenerative Rehabilitation: Connecting Basic Science and Clinic
This session will focus on introducing Regenerative Rehabilitation, a cross-disciplinary field that combines tissue engineering and regenerative medicine with applied biophysics and tissue/organ-specific rehabilitation approaches to enhance tissue repair and function outcomes. Aligning with TERMIS’s goal of advancing tissue engineering & regenerative medicine to improve patient outcomes, we imagine that this regenerative rehabilitation session will help increase communication and collaboration between the fields of tissue engineering and rehabilitation science. In this session, we will invite Dr. Fabrisia Ambrosio, who is the founding director of the Alliance for Regenerative Rehabilitation Research and Training (AR3T) and an Associate Professor at Harvard Medical School, to give an overview of the regenerative rehabilitation field, regenerative rehabilitation research, and funding opportunities. Four other speakers will be selected by the abstract reviewers to ensure that attendees will get to know the cutting-edge research in regenerative rehabilitation.
Decellularized ECM materials for TERM
This session will encompass decellularized extracellular matrices, derived from different tissues and cell populations and their diverse and wide ranging applications in TERM. The keynote will be provided by Professor Stephen Badylak, pioneer of ECM biologic scaffolds, who will provide a valuable commentary on development and clinical applications of decellularized ECM materials.
Advanced Biomaterials for Wet Tissue Adhesion
The lack of suitable tissue adhesives has resulted in a range of unmet clinical needs that current mechanical fixation methods of sutures, staples, and hooks fail to address. Tissue adhesives are needed for the facile manipulation, bonding, and placement of both natural and synthetic surfaces. The development of bioadhesive with novel bio mimicry, polymer chemistry, and/or stimuli activation attempts to fill this clinical gap. This symposium will highlight the latest developments of both preclinical and commercial tissue adhesives to usher in the replacement century-old sewing techniques. Objectives of this symposium is to bring together clinicians, academic researchers, and experienced entrepreneurs to highlight current unmet medical needs and address the technical/engineering/regulatory hurdles preventing clinical application and commercialization. Relevance is to design better healthcare materials that link patient needs, bioengineering, and biomaterials that dynamic material properties paired to tissues, and structure property relationships needed for a variety of soft tissue pathologies.
Skin Regeneration, Wound Healing and Inflammation
Advances in Skin Tissue Engineering
This session will bring the latest research on cellular, molecular, and biomechanical mechanisms involved in skin, wound healing, and inflammation, and the development and use of bioengineered skin constructs/organoids as tools to understand skin biology, perform drug screening or regenerate human skin.
Approaches for Scarless Skin Tissue Regeneration
Today’s standard of care for skin burns and large trauma is transplantation of autologous split-thickness skin, although wound healing is achieved, pain, scars and contractures can be persistent and debilitating for life. Redirecting skin regeneration is essential to address this aberrant healing from a repair to regenerative phenotype. Building on the work on the key elements of the extracellular matrix and/or stem cells from relevant cellular origins, novel approaches are required to restore the integrity of a fully functioning skin. By making use of these components, new biomaterials and skin substitutes are being developed, focused on functional hair follicles, eccrine sweat glands and sensory nerves for appropriate integration. The goal is scarless healing, matching the specific characteristics of the former intact skin. In this session, several options to reach this goal will be presented and discussed.
Skin Substitutes for Treating Infected Wounds: Bacteriophage Formulation
The session will be 90 minutes: One invited lecture: Prof. Erhan Biskin who will deliver a talk covering the points described in the symposium abstract we have already made the necessary agreement with him. His talk will be 30 mins. Then we will have 6 oral presentation – each is 10 mins – that will be selected among the abstracts submitted – all will be related using bacteriophages for skin infections. There will be also a poster session we are willing to select about 6 more abstracts that are also on bacteriophage therapy for skin wounds infections.
Stem Cell Therapies, Developmental Biology and Cell Signaling
Cell-based Materials for Regeneration and Disease Treatment
(Bio)materials based on cells and/or their derivatives (e.g., extracellular matrices – ECMs, and vesicles) have shown promising properties as tailorable structures for the delivery of bioactive agents, as well as supporting structures for tissue engineering and regenerative medicine. This Symposium addresses the exploration of cells as building blocks of scaffold-free or low-biomaterial living or devitalized materials with therapeutic potential. Also, reports on the use of extracellular elements produced by cells – including extracellular vesicles (EVs) of different dimensions and sources, and in vitro-obtained ECMs – are welcome to submit abstracts in this Symposium. Applications of the reported materials may include, but are not restricted to tissue engineering for disease modelling, cell-derived nano- to macrometric constructs for molecule delivery, tissue regeneration, and immunotherapies.
Discoveries in Regenerative Biology to Advance Regenerative Medicine
Generally, tissue engineering and regenerative medicine seek the replacement of cells, tissues or organs. However, the phenomenon of epimorphic regeneration, which achieves complete structural appendage regeneration, inspires the vision to induce complex regeneration in human tissues in the future. Salamanders and fish are well known for their ability to fully regenerate appendages lost to amputation injuries, and mammalian paradigms of epimorphic regeneration, most prominently the regenerating mouse digit tip, are subject of intense study with the goal to eventually induce structural, multi-tissue regeneration in humans, or at least improve integration of implanted cells or scaffolds. As the molecular mechanisms of wound healing, establishment of a signaling center, blastema formation and pattern formation are being uncovered in non-amniotic vertebrates and mammals, paths to translation into mammalian tissues are becoming attainable. This session focuses on ideas and approaches to activate endogenous regenerative ability in non-regenerative tissues.
Progenitor Cells for Disease Modeling and Cell Therapy
Recent advances in regenerative medicine in the respiratory, gastrointestinal, and urogenital fields have largely centered around the use of progenitor cells for direct application in vivo or through engineered tissue models. Progenitor and stem cells of primary and iPSC sources are being leveraged in cell therapy applications, to great therapeutic effect. A variety of tissue models are being developed utilizing progenitor cells for disease modeling as alternatives to traditional animal models. In some fields, such as urogenital, new models are being developed for a variety of previously understudied diseases, whereas other fields with established models, such as respiratory, are now realizing their full potential by testing myriad therapeutic interventions. This session will unify research in our respective organ systems through shared interest in the power of progenitor cells, as well as common features that contribute to model design criteria and cell-delivery considerations. Specifically, all of our systems consist of an epithelial cell barrier in contact with the external environment, which in turn closely contacts vascular and stromal cell populations, and usually possessing a robust immune component. The commonality of these design principles guide our efforts to apply cells in vivo or model these tissues ex vivo, and provides a common ground for us to unite and discuss our progress in these areas.
Stem Cell-based Therapies in Tissue Engineering
Pluripotent stem cells (PSCs) provide autologous and allogeneic replacement tissues for the treatment of potentially all degenerative diseases. Several groups are interested in developing autologous and allogeneic PSC based therapies for degenerative diseases. However, major challenges remain in translating these therapies into clinic. There are challenges to develop clinical grade PSC lines, how to quality control them and test the derived product for preclinical and clinical use. The main goal of this symposium is to discuss approaches various groups are taking to develop a regulatory compliant path to develop an PSC based cell therapy and highlight some of the major challenges that lie ahead of us.
Extracellular Vesicles in TERM
In recent years, research on cell secretome and extracellular vesicles (EVs) for diagnostic and therapeutic applications have generated great interest in the Tissue Engineering and Regenerative Medicine research community. The secretome and EVs hold an immense potential to provide novel strategies to treat a variety of diseases and to revolutionize healthcare, since they are crucial at regulating molecular key processes in health and disease. This symposium aims to provide a comprehensive overview of the current research on the applications of the secretome and EVs in tissue engineering and regenerative medicine, and their impact on personalized strategies. We welcome presentations from, but not limited to, the following topics: i) the biology, biochemistry and biosynthesis of the secretome and EVs; ii) the use of secretome and EVs in diagnostics and therapeutics; iii) the applications of EVs in bioengineering strategies, including biomaterial-secretome based approaches iv) Secretome as a source of biomarkers in personalized and regenerative medicine Target Audience: We expect that the symposium will be of interest to most of conference attendees, including basic scientists, clinicians and industry/companies. We would like to offer the whole life cycle of the use of secretome and EVs in regenerative medicine, i.e., from the bench to the bedside. Additionally, our objective is to gather researchers working on this topic to generate a critical mass and gain momentum within the tissue engineering and regenerative medicine community.
Single-cell Analysis of Engineered Tissues
Single-cell technologies have revolutionized our understanding of tissues. However, it remains difficult to compare organoids, engineered tissues, and ex vivo cultured tissues to native references. This is largely because the majority of computational tools and workflows are not designed specifically for this task. We need to research how to best compare engineered and native tissues using single-cell approaches, and how these approaches may help us to gain disease- and development-relevant biological insight. This session intends to bring together experts from across discipline, in multiple organ systems, to provide a general forum for discussion and sharing of expertise. The session will focus on research leveraging existing single-cell techniques to understand engineered tissue biology, and on pioneering work to develop next-generation computational and biological approaches that can help tissue engineering to develop as a field.
Cell Secretome: Implications in Regenerative Medicine and Aging
For this symposium we have selected a team of 2 young PIs (one male and one female) and one keynote speaker (an internationally recognized expert in the field of cell secretome and senescence), overall representing 3 different universities in 3 different European nations (all TERMIS members). Presentations and discussions would focus on: i) the identification, characterization, and functional roles of secreted factors in intercellular communication, tissue homeostasis, and disease pathogenesis; ii) extracellular vesicles (EVs), with the aim of delving into the biology, biogenesis, and functions of EVs. Topics would include the cargo carried by EVs, their roles in cell-to-cell communication, tissue regeneration, immune modulation, and their potential as diagnostic and therapeutic tools; iii) senescence, with particular focus to the molecular mechanisms underlying senescence, the impact of senescent cells on tissue homeostasis and inflammation, and potential strategies to modulate senescence for therapeutic purposes.
Preclinical Models, Commercialization & Regulation
Manufacturing and Design Considerations for Regenerative Medicine
There has been a significant increase in the number of clinical trials for cell therapies and tissue engineered treatment over the past decade. This calls for a need to design well controlled manufacturing process and product characterization to ensure safety, consistency, efficiency and accuracy of the therapy. This session will specifically focus on manufacturing challenges towards clinical translation. Specifically, the session will consist of speakers from FDA, industry, academia, and partnering government initiatives on research and experience in translating cell therapeutics from the bench to the bed-side. Topics of interest includes regulatory considerations and the development of standardized cell manufacturing methods and benchmarks.
Veterinary TERM – Preclinical and Clinical Applications
The veterinary profession has an important role to play in the TERM translational process, representing the link between basic science and human clinical applications. The TERMIS thematic subgroup “Veterinary Medicine”, with members from all three TERMIS chapters, is therefore applying for a symposium to foster translation of basic research into clinical applications in veterinary and human patients. The latest results from veterinary TERM clinical applications and basic research in large and small animals will be highlighted following the “One health one medicine concept”. In order to attract many veterinary participants, a relevant number of veterinary presentations would be required. In addition, to accommodate presentations covering small animal and large animal research, as well as research using animal models and basic veterinary science to offer a truly comprehensive overview of the field, we kindly request two symposia. If two veterinary symposia should be accepted, we would focus one on basic research and animal models and one on preclinical trails and TERM applications in veterinary patients.
Biopreservation Technologies for Extended Storage of Organs and Tissues
Biopreservation is an essential process for storage, transport and distribution of tissue engineered products that will facilitate widespread application of the end product. The process involves keeping biospecimens at low temperatures to reduce or suspend their physiochemical and metabolic activities and then recovering them to physiological states at a desired future time. Biopreservation has been critical in a number of areas of regenerative medicine such as reproductive cells and tissues for assisted reproductive technology, tissues and organs for transplantation, or stem cells, blood cells or genetically engineered cells for cell based medicine. The goal of this session is to bring together investigators who focus their work in studying biopreservation for extended storage of tissue engineered products (e.g., organoids, microphysiological systems, tissues and whole organ constructs) including but not limited to respiratory, urologic and gastrointestinal systems. Topics of interest include cooling and rewarming technologies, strategies for protection of biospecimens from cold injury, biology at cold temperatures and other related subjects.
Sex, Age, Ancestral Determinants
Biomaterials for Women’s Health Engineering
Women have historically been excluded at all levels of biomedical research and medical device innovation which has led to a gaping hole in the healthcare landscape to meet even the most basic needs in women’s health. To address this critical need, we need innovative biomaterial solutions to expand health technologies available to clinicians to treat women. This session will highlight advances in biomaterial approaches to address the myriad of clinical needs in women’s health. Topics include disorders in reproductive tissues (e.g. endometriosis, cancer, premature ovarian insufficiency, infertility, preterm birth), multicellular tissue-level interactions and mechanics in reproductive health, extracellular matrix dynamics and sex-related differences in chronic diseases and device design, gynecological device design, and breast cancer and reconstruction.
Sex, Age and Ancestral Determinants in TERM Research
As the demand for precision medicine continues to rise, the “one size fits all” approach to the design of tissue engineered devices to treat diseases and injuries is becoming increasingly outdated. Tissue engineered devices have significant potential for transforming precision medicine, and individual patient complexity often necessitates integrating multiple functions into a single device to successfully customize therapies. In this session, we seek to highlight the latest research in tissue engineering technologies that enable prueba evaluation of sex, age, and ancestry as biological variables, including implantable devices for the real-time analysis of a patient’s condition or customized devices and material chemistries that adapt to a specific patient’s biology.
Healthcare Disparities in TERM
Session description will be added soon.
Login Information
- Your login is different than your TERMIS login.
- If you have submitted an abstract to TERMIS-AM in the past, you can use the same login this year.
- If you forgot your password, select the ‘forgot your password’
- If you need to create an account, click ‘create new account’