Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 5th International Conference on Tissue Engineering and Regenerative Medicine Berlin, Germany.

Day 1 :

Biography:

Werner Mohl has graduated from the Vienna University, Medical School and received his PhD in Natural Science from the University of Vienna. He has received the Board of Surgery and the Academic License to teach Surgery. He is the Professor of Surgery at the University of Vienna. He has published more than 90 papers in reputed journals, book chapters and has been serving as an Editorial Board Member of several journals.

Abstract:

Reprogramming of cardiomyocytes require severe interference with normal signaling and cellular homeostasis. In contrast with molecular vectors and scientifically highly disputable stimulus triggered acquired pluripotency (STAP), use mechanical stress for induction which seems to induce regeneration by reiterating embryonic developmental pathways. Here, we show that epigenetic signaling via hemodynamic mechanotransduction on human venous vascular cells in cardiomyopathy hearts induces pluripotency marker Klf4, but not NANOG. Pressure controlled intermittent coronary sinus occlusion (PICSO) temporarily increases shear stress during backflow in cardiac microcirculation and via mechanical stretch of elevated pressures on vascular cells including pericytes create a pleiotropic signaling known as canonical pathway during morphogenesis of theheart. This intervention was applied in patients with end stage cardiomyopathy for 20 minutes. Sera of coronary sinus blood samples were co-cultured with human cardiomyocytes explanted from a patient with end stage cardiomyopathy during heart transplantation. Klf4 expression was measured as pluripotency marker. Sera of treated patients were able to significantly upregulate Klf4 in cultured cardiomyocytes as compared to those taken before treatment and in untreated controls. In porcine myocardial specimen subjected to acute ischemia with and without PICSO also a significant increase of Ki67 and a reduction of p53 were observed. This indicates that direct mechanotransduction as well as the release of soluble factors act in accordance in the recovery of failing hearts mimicking similar pathways known during the development of the heart. We conclude that the STAP concept might have been abolished too early and that a clinically feasible transcoronary sinus intervention may inducea paradigm change in regeneration therapy.

Biography:

Denis Barritault graduated in Physics, completed his PhD in biochemistry in Paris University. Post-doctoral in molecular immunology at Pasteur Institute and NYU as NIH Fogarty Fellow he joined INSERM unit in Paris as developmental biologist. He made the first description and patents of FGF extracted from retina in 1979 and 82 as skin and cornea healing agent, became full professor at Paris-Est University in 1985, founded and directed a CNRS Laboratory on cell and tissue regeneration until 2003. He is now President of OTR3, Emeritus professor, honorary director CRRET CNRS unit and author in over 200 publications and 30 patents.

Abstract:

Extra Cellular Matrix (ECM) microenvironment regulates locally our continuous ability to replace dead cells by new cells. This central law of all living is known as tissue homeostasis. Heparan sulfates (HS) are key elements of the ECM scaffold that store, protect and position the various Cell Communication Peptides (CCP) in the cellular microenvironment. HS play a pivotal role in the regulation of the bioavailability of CCP, cell proliferation, migration and differentiation required for tissue regeneration. Tissue injury will lead to destruction of cells and surrounding ECM. CCP released by inflammatory and circulating cells can then promote tissue repair, but with a loss of tissue quality, leaving scars or fibrosis. We have engineered biodegradable nano-polysaccharide mimicking HS, named RGTA for ReGeneraTing Agent. Introduced at the site of injury,RGTA will bind to the matrix proteins of the damaged ECM, and to the CCP produced by healthy neighboring cells, thereby restoring the ECM microenvironment and conditions for tissue homeostasis. This matrix therapy approach has considerably improved the quality of healing in various animal models with reduction or absence of fibrosis resulting in a real regeneration process. The RGTA technology has been validated in clinics and over hundred thousand of patients treated both for corneal and skin ulcers with no adverse effect. Adapted RGTA are in development for more tissue injuries extending RGTA as a new therapeutic class in the field of regenerative medicine exploiting our natural potential without need for exogenous cells. RGTA can combine with cell therapy by constructing a niche to favor homing. The future of regenerative medicine lays in a proper adjustment of the microenvironment to optimize cell colonization, expansion, replacement and recovery of their functions.

Keynote Forum

Bertha Chen

Stanford University School of Medicine, USA

Keynote: The role of regenerative medicine in Obstetrics and Gynecology

Time : 09:50-10:15

Biography:

Bertha Chen has completed her Bachelor of Science degree in Chemical Engineering from the University of California, Berkeley and Doctorate of Medicine from Stanford University School of Medicine. She is a Professor of Ob/Gyn and Urology (by courtesy) at Stanford University School of Medicine and Co-Chief of the Division of Urogynecology and Pelvic Reconstructive Surgery. Her research focus is in stem cell pluripotency, mesenchymal differentiation and stem cell based therapy for urinary incontinence. She has published more than 55 papers in reputed journals.

Abstract:

Stem cell research and regenerative medicine are integral to the field of Obstetrics and Gynecology. With the explosion of technologies directed at treatments for infertility, there is a critical need to understand properties of pluripotency and how it relates to embryo development. Evidence of cancer stem cells will drive new discoveries in the treatment of gynecologic malignancies such as ovarian and endometrial cancers. Notably, there is great interest in regeneration of aged or damaged tissues specific to the female anatomy and lower urinary tract via stem cell based technologies. In humans, damaged tissues are generally replaced by continuous recruitment and differentiation from stem cells in the body. However, the body’s ability for regeneration is reduced with aging. Examples of conditions in need of regenerative therapies are urinary and fecal incontinence resulting from sphincter deficiency and pelvic organ prolapse. These conditions are prevalent in older women and greatly affect quality of life and represent colossal health costs. Current approaches for tissue regeneration include transplantation of adult mesenchymal cells or their derivatives and implantation of engineered scaffolds seeded with these cells. Induced pluripotent stem cells are a promising source of autologous cells. Because of their property of self-renewal, large amounts of cells can be produced for transplantation. Furthermore, their pluripotent state allows for derivation of multiple cell types thus facilitating implementation in a stepwise fashion from simple cell injections to more elaborate matrices with multiple cell types that have potential for full regeneration. Here, we discuss stem cell based approaches for tissue regeneration in obstetrics and gynecology.

Keynote Forum

Pedro Morouço

Polytechnic Institute of Leiria, Portugal

Keynote: Novel scaffolds fabrication methods in regenerative medicine: The cartilage challenge

Time : 09:00-09:25

Biography:

Pedro Morouco is specialized in Biomechanics and the Head of R&D Biofabrication Group at the Centre for Rapid and Sustainable Product Development, Portugal.His research activity focuses mostly on products and process engineering, bringing the gap between the lab and in vivo applications. He has co-edited 2 books, authored and co-authored more than 100 papers published in books; 40 international journals and 55 international conferences. He is the Member of the Editorial Board in several international peer-review journals and was distinguished with the New Investigator Award 2014 from ISBS. He is the Chairman of the CDRsp Advanced Courses on Regenerative Medicine and Workshops on Direct Digital Manufacturing for Medicine.

Abstract:

Cartilage is a tissue with a huge complexity, which is present in the human body in three types: Hyaline cartilage, fibrocartilage and elastic cartilage. Apart from some resemblances, these types are quite different and play unlike roles for human functionality. For instance, the hyaline cartilage, also known as articular cartilage, has a major role in providing joints with a surface that combines low friction with high lubrication. On the other hand, fibro-cartilage (e.g. in the temporomandibular joint disc) spreads the intra-articular load, stabilizes the joints during translation and decreases the wear of the articular surface. Even though, getting a deeper knowledge on cartilage characterization and understanding, bridging the gap between anatomy and physiology, may lead the way for better implants aiming cartilage repair and regeneration. This is of even more interest as cartilage is an avascular tissue of the human body, hence with an extremely low capability for tissue regeneration. However, this ground-breaking issue can only be successful achieved with the establishment of multidisciplinary research teams. Thus, several processes have been tested to assemble tailored and optimized constructs that could endorse cartilage repair and regeneration. These processes advantages, drawbacks, results and capabilities will be presented and discussed.

  • Tissue Engineering | Tissue Repair & Regeneration
Location: Kiel 1-3

Chair

Barritault Denis

OTR3, France

Co-Chair

Pedro Morouço

Polytechnic Institute of Leiria, Portugal

Biography:

Abstract:

PIF Premise: Endogenously secreted only by viable embryos, PIF has determining immune-modulatory and transplant acceptance functions throughout pregnancy–a “perfect” regulatory scenario. Synthetic PIF replicates native peptide functions in representative preclinical/clinical non-pregnant setting. Specifically, PIF comprehensively addresses neuro-repair locally and systemically.PIF Effects: PIF exerts autotrophic effects on native stem cells. In newborn/early pregnancy PIF has neurotrophic/protective effects; the notochord initiates embryogenesis. In prematurity and multi-injury-induced brain damage (HIE), PIF reverses corpus striatum and cortex damage. In adult multiple sclerosis, (EAE) PIF promotes re-myelination while reversing chronic paralysis up-to-total resolution.PIF Mechanism of Action: Mechanistically, PIF crosses intact BBB targeting brain and spinal cord microglia, neurons and blood vessels. In HIE PIF reduces let-7 microRNA, PKC/PKA phosphorylation while increasing IL10. In adult EAE, PIF regulates brain phosphoproteins and spinal cord proteome to reduce oxidative stress and protein misfolding while promoting Na+/K+/Ca++, glucose and amino acid transport. Thus PIF promotes neuron assembly and synaptic transmission while reducing systemic inflammation. PIF promotes endogenous brain stem cells proliferation and differentiation. The effect of PIF is superior to and potentiates intra-cranially injected stem cells.Conclusion: PIF is in Fast-Track FDA Phase Ib clinical trial (NCT#02239562). Collectively, PIF high-safety and comprehensive effect on CNS and peripheral nervous system coupled with intimate interaction with endogenous stem cells support PIF’s planned phase II clinical trial for neuro-repair.

Biography:

Anne des Rieux has completed his PhD in 2006 from Université Catholique de Louvain (UCL), Belgium and preformed her Post-doctoral study at the Northwestern University, Chicago. She is an Associate Professor at UCL and is developing a team within the Louvain Drug Research Institute (LDRI). She has published more than 30 papers in reputed journals.

Abstract:

Traumatic spinal cord injuries (SCI) cause devastating neurological deficits and disabilities. Symptoms vary greatly,ranging from pain and paralysis. We focus on drug and cell delivery for spinal cord repair following traumatic SCI. We incorporated FGF-2 and VEGF in scaffolds, and GDNF and VEGF in hydrogels and studied their impact on spinal cord injury and functional recovery. We observed a functional recovery of rats treated with GDNF. In order to further stimulate spinal cord repair, our approach is to combine drug and stem cells with hydrogels, to ensure local delivery of drugs and survival of cells at the injury site. Human dental stem cells from the apical papilla (SCAP) have been selected based on their origin (neural crest) and ease of access. We first assessed the impact of SCAP encapsulation in different hydrogels. We then compared in vivo the influence of SCAP delivery with the implantation of the whole tissue they originate from (apical papilla) in a rat spinal cord hemisection model. Functional recovery was observed in the group treated with the papilla. Genomic and proteomic analysis are being performed to identify the markers involved. Second, SCAP were grown on growth factor-loaded microcarriers and embedded in hydrogel. Impact on cell survival and gene expression is being studied, after which the system will be implanted in a SCI model. In conclusion, we are developing a multidisciplinary approach based on the combination of drug and cell delivery to stimulate spinal cord repair.

Biography:

Rekha Samuel is a Pathologist working at the Centre for Stem Cell Research (CSCR), Christian Medical College, India. She did her Post-doctoral Training in Dr. Rakesh K Jain’s Laboratory at Massachusetts General Hospital, Boston, on an overseas fellowship that was funded by the Department of Biotechnology,Government of India. She is the Principal Investigator of the Vascular Biology Laboratory at the CSCR. Her current research focuses on examining the Microvascular Defects of Type 2 Diabetes using the Gestational Diabetes Mellitus Placenta. She has recently been awarded the European Foundation of Study for Diabetes/Sanofi 2015 grant.

Abstract:

Human iPSC derived vasculogenic cells may offer a long sought solution for obtaining large numbers of autologous cells sufficient for tissue engineering and opportunities to model vascular disease in a dish. We report a novel approach for the derivation of endothelial precursor cells from hips cells using triple combination of selection markers: CD34, neuropilin-1(NP-1) and KDR, and an efficient 2-D culture system for hiPS cell-derived endothelial precursor cell expansion. Functionality of blood vessels in cranial window models of severe combined immunodeficient (SCID) mice was determined by non-invasive longitudinal in vivo multiphoton laser scanning microscopy for parameters that included red blood cell (RBC) velocity, blood flow and permeability to bovine serum albumin. We successfully generated endothelial cells from hiPS cells obtained from healthy donors and formed stable functional blood vessels in vivo - lasting for 280 days in SCID mice. The RBC velocities of engineered blood vessels were comparable to normal endogenous host vessels (1.36±0.3 mm/s), and demonstrated a higher permeability as compared to endogenous vessels. We also generated mesenchymal precursor cells (MPCs) from hiPS cells in parallel. Moreover, we successfully generated functional blood vessels in vivo using these endothelial cells and mesenchymal precursor cells derived from the same hiPS cell line. In parallel, we have isolated hiPSC-derived ECs and PVCs from type-1 diabetes and maturity-onset diabetes of the young (MODY) cases. The T1D-iPS engineered blood vessels were functional for 4 months in vivo while MODY-iPS-ECs failed to form functional blood vessels. Human iPSC-derived vasculogenic cells may be an abundant source to examine vascular defects of diabetes in a dish.

Biography:

Scott L Nyberg is a trained Liver Transplant Surgeon and a Biomedical Engineer. His engineering training includes a BS degree in Chemical Engineering from MIT and PhD in Biomedical Engineering from University of Minnesota. His medical training includes an MD degree from Johns Hopkins and Transplant Surgical training from the University of Minnesota. He directs the Artificial Liver Program and Liver Regeneration Program at Mayo Clinic Rochester, where he has worked as a Transplant Surgeon since 1996. He has published over 175 papers in reputed journals, and serves on the Editorial Board of Journal of Hepatology and Liver Transplantation.

Abstract:

Acute liver failure is a serious, potentially life-ending, medical problem. Spontaneous recovery occurs in less than 50% of cases. Recovery would be more frequent if a supportive therapy were available to correct the toxic milieu of acute liver failure to prevent its extra hepatic manifestations and to assist in liver regeneration. Therefore, we have developed a novel supportive therapy, the spheroid reservoir bio-artificial liver (SRBAL), composed of 20-40% of the hepatocyte mass of a normal human liver. The greater cell dose is accomplished with anchorage-independent aggregates of primary hepatocytes (spheroids) engineered by a novel rocked mixing technique. Results of this pivotal preclinical study demonstrate that the SRBAL improved survival in an allogeneic model of acute liver failure. Survival benefit correlated with the rate of ammonia detoxification and lowering of intracranial pressure indicating a neuroprotective effect of this cell-based therapy. Plans for clinical evaluation of the SRBAL are underway. In addition, we have bioengineered a novel animal, the FAH deficient pig, to serve as an in vivo“incubator” for large scale production of primary human hepatocytes. The presentation will include progress towards a next generation “humanized” SRBAL employing human hepatocytes produced in the FAH deficient pig.

Biography:

Liudmila Leppik has completed her PhD in Molecular Biology at the Laboratory of Human Gene Structure and Function at the Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry in Moscow, Russia and Post-doctoral studies at the German Cancer Research Center in Heidelberg, Germany. She is currently a Postdoctoral Research Fellow and Assistant Director at Frankfurt Initiative for Regenerative Medicine at the Goethe-University in Frankfurt/Main.

Abstract:

Electrical stimulation has been successfully used to treat bone defects clinically for many years. Recent in vitro studies have demonstrated that ES can change cell behavior such as migration, proliferation and differentiation. The goal of this project was to combine bone tissue engineering (TE) and electrical stimulation (ES) approaches in in vivo and in vitro experiments to determine its effectiveness and characterize the underlying mechanisms, respectively. In vitro experiments were performed in a customized six well ES culture chamber, in which we exposed adipose- (AT-MSC) and bone marrow- (BM-MSC) derived MSC to ES in 2D and 3D culture. Results showed that ES changed osteogenic gene expression patterns in both AT- and BM-MSC,and that these changes differed between the two groups. We found that ES increased BMP2 and TGF-ß1 expression in MSC suggesting calcium signaling as a potential mechanism. In in vivo experiments, a rat femur critical size defect (CSD) model was used and treated with AT-MSC seeded on calcium phosphate scaffold (β-TCP) material. Femurs from three groups of rats(40 per group; control=ß-TCP alone; sham=ß-TCP+AT-MSC; experimental=ES+ß-TCP+AT-MSC) were assessed at one and eight weeks post treatment by mean of histology, gene-expression and mechanical testing. The results of this ongoing in vivo experiment will be presented at the upcoming meeting. These initial findings may support the use of ES in TE applications which could potentially improve and expand their use in the clinical setting.

Eva Sykova

Institute of Experimental Medicine ASCR, Czech Republic

Title: Stem cells and biomaterials for the treatment of spinal cord injury, stroke and ALS
Biography:

Eva Sykov is a Physician, Scientist, an internationally recognized expert in Neuroscience and Cell Therapy and was the Director of the Institute of Experimental Medicine from 2001-2016. Her research focuses on neurologic diseases and stem cells. She has worked at Charles University, Prague, Founding and Directing the Department of Neuroscience, 2nd Medical Faculty (1996-2012) and leading the Center for Cell Therapy and Tissue Repair (2000-2011). In 2008 she built the Innovative Biomedical Center, which houses a business incubator that assists in the transfer of results in regenerative medicine research into practice.

Abstract:

Stem cells alone or in combination with biomaterials have been investigated for their therapeutic potential in animal models of spinal cord injury (SCI), stroke and ALS. We compared human mesenchymal stem cells (MSCs) from bone marrow, a conditionally immortalized human stem cell line from fetal spinal cord (SPC-01) and human induced pluripotent stem cellderived neural precursors (iPS-NPs) for their capacity to migrate towards lesion sites, differentiate and induce regeneration.Transplanted cells were labeled with iron-oxide nanoparticles for MRI tracking. Animals were tested using motor and sensory tests. Animals with chronic SCI were implanted with HPMA-RGD hydrogels seeded with MSCs. In SCI, the best functional improvement was found after iPS-NP transplantation, followed by MSCs and SPC-01. MRI proved that the cells migrated into the lesion, survived for several months and differentiated into motoneurons and glia. Improved motor and sensory scores in chronic SCI were found after the implantation of biomaterials seeded with MSCs. Natural ECM hydrogels bridged the SCI lesion cavity, modulated the immune response and provided the substrate for in vivo neural tissue regeneration. Grafting of  iPS-NPs reversed stroke-induced somatosensory and motor deficits and protected the host substantia nigra from atrophy. MSC were also intrathecally applied to rat ALS-model (SOD1). Overall survival in the cell-treated group compared with the shamtreated group was prolonged, the cell-treated rats showed better motility, grip strength and greater numbers of perineuronal nets in the spinal cord. Our results demonstrate that the intrathecal application of MSCs can slow down progression of the disease.

Biography:

Enrique Brandan is a Professor and Chairman of Department of Cell and Molecular Biology, Faculty of Biological Sciences at the Pontificia Universidad Católica de Chile, Santiago, Chile. He has earned his BSc in Biology at Universidad de Chile and PhD in Cell Biology at Universidad Catolica de Chile. He was an International Scholar of the Howard Hughes Medical Institute, USA for 10 years. He is a Member of the National Academy of Sciences, Chile. He has been interested in the signaling pathways that control skeletal muscle differentiation and in the mechanisms associated to fibrosis present in several skeletal muscular dystrophies. He has been concentrating in to develop new strategies to fight fibrosis improving skeletal muscle strength and cell therapeutic approaches.

Abstract:

Duchenne muscular dystrophy (DMD) is characterized by absence of the protein dystrophin, muscle wasting and fibrosis.We have evaluated the role of the profibrotic connective tissue growth factor (CTGF/CCN2) in a genetically model (mdx-CTGF+/- mice) and in a decreasing CTGF activity model, by blocking CTGF antibodies in mdx mice. In both models, we observed a decrease of fibrosis together with a significant less muscle damage compared to mdx mice. The decrease or blocking of CTGF caused an improvement of muscle strength and improvement of wild type cell graft. Thus, CTGF could be novel therapeutic agents to treat muscle fibrosis associated to DMD and improve cell therapeutic approaches.

Biography:

W Bonani has received his PhD in Materials Science from the University of Trento in 2011 and in Mechanical Engineering from the University of Colorado in 2012. He is currently holding a Post-doctoral position at the University of Trento (INSTM research unit). During his career, he has developed and studied scaffolds for tissue engineering applications (vascular, bone, cartilage, neural), systems for drug encapsulation and delivery, extraction, manipulation and processing of biopolymers (silk, collagen, alginate) and cellularized 3D building blocks for tissue modeling. He has published more than 15 papers in peer-reviewed journals.

Abstract:

Mammary acini organization plays a fundamental role in tissue development and physiology, but also could offervaluable insights in cancer onset and progression. 3D in vitro models represent a new paradigm in biological sciences.Reconstituted Matrigel® Matrix was successfully used to model 3D tissues; however, Matrigel® encompasses a wide range of different factors, presents large batch-to-batch variation, is expensive and difficult to isolate. Here we designed tunable alginatebased substrates and assessed the assembly of MCF10A human breast-epithelial cells with a focus on the role of laminin-1 in acini maturation and cavitation. Collagen and laminin-1 were used to increase cell adhesive properties of the alginate matrix and to modulate cell activity. We exploited an overlay model consisting of an alginate-based hydrogel substrate and the addition of laminin-1 in the cell culture medium and followed mammary acini morphogenesis. The evolution of cell spheroids was followed for 21 days; immunofluorescence was performed to investigate acini maturation, cavitation and cell polarization were also analyzed in terms of area and roundness. Image analysis was performed to analyze spheroids/acini for area and roundness. We developed an efficient method for the development of 3D cell spheroids/acini in vitro and studied the effect of laminin-1 on spheroids formation (added to medium). This work established tunable, instructive and medium-throughput in vitro culture systems consisting of alginate-based hydrogel substrates and enriched culture media for the development of mammary acini. The modulation of culture substrates and signaling molecules could represent a platform to study the effects of cellular microenvironment on acinar morphogenesis.

Biography:

Andrzej Lange has graduated with a Medical degree with distinction from the Medical School in Wrocław, Poland. Currently, he is a Professor in the Institute and Founder and Head of the Lower Silesia Center of Cellular Transplantation in Wrocław. His international experience started as a Leverhulme Fellow in the Middlesex Hospital Medical School, London. During 8 year period, he used to spend three months yearly as a Visiting Scientist in the Institute of Experimental Medicine and Biology in Borstel, Germany. He has been a Short Term-Visitor and lectured in a number of European and North American scientific institutions. He is author of 239 scientific papers in peer-reviewed journals.

Abstract:

Graft vs. Leukemia effect preventing relapse in patients with hematologic malignancies receiving allogeneic hematopoietic stem cell transplantation (alloHSCT) can be augmented by infusion of donor lymphocytes (DLI) in patients at the risk of relapse. However, this procedure can ignite the graft vs. host disease in about 30% of patients post DLI. The lower rate of leukemia relapse in patients receiving alloHSCT as opposed to patients post autoHSCT prompted us to develop a new approach in DLI delivery to facilitate anti-leukemic allo-reactivity not increasing the risk of GvHD. The aim of our study is to regenerate the immune system originated from transplanted (HSCT) material especially within the lymphocyte compartmentexerting cytotoxic potential but not facilitating GvHD. The elaborated and clinically implemented approach consisted of the following steps: (1) Identification of relapse post alloHSCT with the presence of leukemic cells in the marrow or in the bones in extramedullary relapse; (2) Obtaining from the donor of mononuclear cell population containing 61%±2.34 (sem) of CD3+ cells in the lymphocyte gate with the use of cell separator; (3) The cells are divided into portions securing CD3+ cells infusion in a dose of 10E6 kg-1 BW for the first infusion, 10E7 for the second approach and 10E8 for the third infusion, frozen in the liquid nitrogen and stored in our tissue bank and (4) Donor cells are injected directly to the posterior iliac crest in patients with marrow or, to the affected bones in patients with extramedullary relapse. The 17 procedures were performed in 5 patients. To have an access to the marrow cavity or to the bones affected by leukemia, the routine trocar for bone harvesting or the bone injection gun, were employed, respectively. The procedure was safe and well tolerated and no symptoms of GvHD were noted after the procedures. In a patient with extramedullary relapse, the healing of the bone lesions was seen clinically and in the evaluated nuclear resonance images. The positive response lasted 7 months until the fatal marrow relapse emerged. In 4 patients with medullary relapse, the reduction of leukemic cells or stable disease was seen in all patients except one, after 1st and 2nd DLI procedures. The patients were under the observation from 3 to 16 months. The positive response was consolidated either by the second transplant (2 patients) or the maintenance approach with the use of Sorafenib (1 case) or anti-CD20 MoAb (one  CLL case). Notably in all marrow evaluations performed along the treatment, there was an excess of CD8+ cells in the marrow as compared to the blood especially seen within CD8+ and CD279+ cells compartment. In conclusion, an accumulation of CD8+ donor lymphocytes harboring CD279+ cells in the marrow cavity of patients relapsing post alloHSCT receiving IB-DLI associates with a positive anti-leukemic response. This observation strongly suggests the presence of T cells cytotoxic response in the marrow infiltrated by leukemia restored by IB-DLI.

Biography:

Luminita Simion Labusca is a Medical Doctor, Consultant in Orthopedics and Traumatology. She has completed her PhD in Regenerative Medicine. She has worked as Medical Practitioner, Clinical Researcher and Scientist involved in basic science in various national and international institutes in Belgium, Ireland,Denmark and as well as in South Korea. Scientifically, she has had international collaboration across multitude of EU and US. She has both EU wide and international recognition as a Scientist in Orthopedic and Traumatology, Sports Medicine, Regenerative Medicine, Cell Biology. She is a Reviewer in several international scientific journal boards. She has edited special issue of international peer reviewed journals. She is a Founding Member of Regenero, Romanian Regenerative Medicine Association.

Abstract:

The use of nano-scale materials, particularly of magnetic nanoparticles (MNP) has evolved as an increasing field of research in life sciences. In the past two decades, various iron oxide based MNPs are being explored for applications in magnetic separation, magnetic resonance imaging (MRI), drug delivery, as tracking agents within the emerging field of cell therapy, for guided tissue engineering as well as for hyperthermia (HT) applications. Our research focus on the development of proprietary Fe-Cr-Nb-B MNPs for local delivery of HT aiming the treatment of solid malignancies, for magnetic enhanced cell delivery as well as for magnetic guided tissue engineering. Medium and long term interaction of MNPs with human mesenchymal and stem cells of various origins (bone marrow, adipose tissue) was investigated in regard with their proliferative, differentiation and immune modulatory potential.Cell viabilility, proliferative as well as mesenchymal lineage differentiation potential was unaffected by the presence of bare as well as chitosan coated MNPs of clinically relevant amount (1-2 mg/mL). Particle retention inside the cell was demonstrated up to 28 days with several particularities related to cell type. We are currently investigating MNP loaded cell behavior after exposure to magnetic field.

Biography:

Marta Pokrywczynska is the Head of the Department of Regenerative Medicine at Nicolaus Copernicus University (NCU) (Bydgoszcz, Poland). She has completed her MSc and PhD in Medical Biotechnology from the NCU. Her research area focuses on “development of new tissue engineering and regenerative medicine
technologies”.

Abstract:

Background: Development of an effective method of urinary bladder regeneration is associated with identifying the pathways that play a key role in the regeneration process. Until now, the regeneration pathways in tissue engineered urinary bladder have not been determined.
Aim: The aim of this study was to analyze regeneration pathways in the tissue engineered urinary bladder.
Method: The study was performed on 40 Wistar rats. Adipose tissue was harvested from 20 rats and adipose derived stem cells(ADSCs) were isolated. After hemicystectomy, bladders were augmented with bladder acellular matrix (BAM) (n=20) or BAM seeded with ADSCs (n=20). 10 rats were sacrificed in each group after three and six months. The total RNA was isolated from reconstructed bladder wall and then quantity, purity and integrity of RNA were evaluated. Gene expression was evaluated using microarray and GeneSpring software.
Results: Isolated RNA revealed good purity, concentration and RNA integrity number above seven in all samples. Gene expression analysis indicated 711 differentially expressed transcripts in bladders reconstructed with BAM seeded with ADSCs compared to bladders reconstructed with unseeded BAM, six months after the reconstruction and 8 241 differentially expressed genes, three months after the reconstruction. A large number of differentially expressed genes were involved in a lot of pathways, including B and T cell receptor signaling pathway, IL-3, IL-6, IL-2 and IL-5 signaling pathways, inflammatory response pathway, GPCR signaling pathway and Wnt signaling pathway.
Conclusion: Microarray gene expression analysis allows to create a regeneration patterns in tissue engineered urinary bladder.

Biography:

J R Burgess runs a very successful Non-Surgical Orthopedics practice that utilizes medical fitness as part of their care for achieving better outcomes. Currently,they assembled a 28000 sq. ft. facility with well over 8000 patients who are seeing life changing results. He along with Dr. Baumgartner formed MedFit and has licensed their model to over 30 clinics around the world.

Abstract:

With the uncertainty regarding the future of the health care system, administrators, medical professionals and private practices are looking for solutions to a broken system. Today’s crisis leads to a new opportunity that will combat rising health care costs and improve patient outcomes. The obesity epidemic and the incidence of disease and injury have forced the need for a preventable, medically integrated, outcome based model of medical fitness. Exercise and proper nutrition is the foundation to all functional medicine and anti-aging treatments as well as a necessity for healing and preventing injuries in patients. As we optimize body composition through increasing lean muscle mass while decreasing the fat mass, many of the hormone imbalances that are causing chronic disease can be eliminated. As specialists in our field, we need to have the tools to educate and train our patients on proper exercise and lifelong nutrition to maximize the healthy changes in the lives of our patients. As physicians, we have the ability to influence our patients to make positive changes in their fitness and nutrition that will impact there clinical outcomes. We just need to implement those into our current practice models. Five learner objectives are: Review the current literature on to use medical fitness to change the course of history; understand the need for physicians to prescribe medical fitness and the power we have to make change; maximizing orthopedic outcomes through integrated fitness and nutritional services; compliment any medical practice model to increase cash based revenue and obtain new patients at the same time and; easy ways to implement medical fitness into your practice.

Biography:

Heba Al-Zer is a Dentist who has completed her PhD in Stem Cells Biology from the University Medical Center Hamburger-Eppendorf in Germany, 2015. She is an Assistant Professor of the Conservative Department at the School of Dentistry, University of Jordan. Her work focuses on “The properties and the potentials of the human dental pulp stem cells (DPSCs)”.

Abstract:

The peripheral nervous system (PNS) has an intrinsic ability for repair and regeneration even after severe injury. Schwann cells play a major role in PNS axon regeneration. They are able to dedifferentiate, re-enter the cell cycle and promote axonal regrowth by phagocytosis of the axonal and myelin debris, recruiting macrophages to the injury site, secreting several neurotrophic and key transcription factors and forming a unique column of cells called bands of Bungner within their endoneurial tubes for guiding the axonal sprouts from the proximal stumps. In clinical practice, nerve autografting is considered to be the “clinical golden standard” for promoting repair of segmental peripheral nerve injury and bridging a critical gap defect,but the results are still unsatisfactory. In addition, the reported outcomes of synthetic nerve conduits and allogenic nerve grafts have been voluminous and often conflicting. Therefore, cell-based therapies by transplantation of Schwann cells within appropriate scaffolds have been introduced as a promising treatment modality for peripheral nerve regeneration. The dental pulp of adult human teeth contains different stem cells populations, which show broad diversity and potentials. Here, the NC derived DPSC population is discussed in term of their culturing method and induction of differentiation into SCs. The NC derived DPSC population is to be recruited in the future for peripheral nerve injury regeneration after their induction into SCs in vitro. We recommend that these DPSC-derived SCs can be considered as a superior alternative source of SCs compared with a nerve donor source. DPSCs are feasible, cost and time efficient and are harvested without complicated surgical procedures.

  • Stem Cell Transplant | Ethical & Legal Issues
Location: Kiel 1-3

Chair

Scott L. Nyberg

Mayo Clinic School of Medicine, USA

Co-Chair

Roberta Di Pietro

Gabriele d’Annunzio University, Italy

Biography:

Claire Henchcliffe has received her Doctorate at Oxford University UK. After completing Medical Training at the College of Physicians and Surgeons of ColumbiaUniversity, New York and the New York Presbyterian Hospital, she established the Weill Cornell Parkinson’s Disease and Movement Disorders Institute, New York,where she serves as a Director. She is a Vice Chair for Clinical Research and Associate Professor of Neurology at the Weill Cornell Medical College, New Yorkand combines clinical practice and research in Parkinson’s disease. Her research interests focus on developing new therapies for Parkinson’s disease, in particularcell-based approaches and optimizing clinical trial designs.

Abstract:

Neurorestorative approaches in Parkinson’s disease (PD) based upon new stemcell technologies are imminent. As clinicaltrials are starting, it is critical to consider how to best ensure a genuinely informed consent.We therefore focused upon current barriers to such a goal and identified the following themes:
 
(1)Barriers to adequate disclosure of risks and benefits(investigator-dependent): In first-in-human trials, risks and benefits are incompletely defined. A substantial literature in fetal tissue transplant in PD is only partially applicable, and the possibility of serious risks remains, including tumor formation, graft rejection, and risks of immunosuppressive agents if administered.
 
(2)Barriers to understanding clinical research methodology(participant-dependent):Research participants may have difficulties understanding concepts integral to clinical research, such as equipoise. There is also strong evidence that the therapeutic misconception influenced participant’s consent in previous surgical trials in PD. Cognitive dysfunction, common in PD, may or may not impair understanding of informed consent information, thus complicating evaluation of capacity to consent.
 
(3) Need for extended consent process (investigator and participant-dependent): The traditional model of a single limited visit to address informed consent appears to be insufficient in upcoming clinical trials that involve complex scientific underpinnings, uncertainty in risks and benefits, and recruitment of participants with a neurodegenerative disorder that commonly affects cognition. Extended informed consent processes to consider include the use of multimedia educational materials, assignation of a “research partner”, and multiple disclosure sessions.

Biography:

Jan Kriz has completed his PhD from the Charles University in Prague, Czech Republic and Post-doctoral studies from the University of Western Ontario in London, Ontario, Canada. He is the Principal Investigator of projects focused on “improvement of pancreatic islet transplantation efficiency in the IKEM”, at leading transplant center in the Czech Republic and one of the four most active centers in transplantation of insulin producing tissue. He has published more than 30 papers in reputed journals.

Abstract:

The pancreatic islet transplantation represents a real treatment for type-1 diabetic patients suffering from hypoglycemia unawareness syndrome. The wider application of this method is limited among others by a suboptimal efficiency of islet engraftment. In clinical practice, islets are commonly infused into the hepatic portal vein, where they spontaneously settle in terminal branches. Immediately after infusion, they induce the Instant Blood Mediated Inflammatory Reaction (IBMIR),which results in considerable destruction of graft cells and subsequent non-optimal long term function. The better site for islet transplantation should predominantly provide the adequate oxygen supply without a direct contact to the recipient blood, and should be created using a minimally invasive surgery without any signs of inflammation in time of transplantation. Therefore,the most groups prefer a long term (at least four weeks) creation of subcutaneous cavities with completely finished healing process in time of islet administration. Practically, they transplant islet into the almost avascular fibrous tissue. The main goal of this study is the identification of optimal timing of islet transplantation after the insertion of polymeric macroporous scaffold under skin. We intend to detect the vascular density, blood perfusion and intensity of inflammation during individual phases of tissue healing in the close vicinity of inserted scaffolds. Using a marginal mass of islets transplanted in optimal time,we demonstrate the superior metabolic control in diabetic Lewis rats.

Thomas Skutella

University of Heidelberg, Germany

Title: Germ stem cell reprogramming
Biography:

Thomas Skutella has completed his MD from Frankfurt, Germany and Post-doctoral studies from Max Planck Institute for Psychiatry, Munich and the Institute of Anatomy, Charite, Berlin. He was the Director of the Center for Regenerative Medicine, Tübingen and is currently the Head of a Department in the Institute for Anatomy and Cell Biology, Heidelberg.

Abstract:

It has been suggested that the generation of human germline stem cells from primordial germ cells (PGCs) or spermatogonial stem cells (SSCs) may provide simple and non-controversial access to an individual cell-based therapy. These human studies are based on mouse models of germ stem cell reprogramming, in which the molecular mechanisms underlying the shift to pluripotency could be investigated in more detail. However, recent studies revealed the impossibility of a conversion of adult mouse SSCs and human PGCs into a truly pluripotent state. For the conversion of PGCs it appeared to be necessary to conduct a two-factor induction of pluripotency with Oct4 and Sox2. From the CD49f MACS- and matrix-selected fractional part of enriched human adult spermatogonia, adult human germ stem cell cultures were grown. When compared to authentic pluripotent stem cells, these human adult germ stem cell (haGSC) cultures were not fully reprogrammed. For this reason, an investigation of the molecular blockages of pluripotency in germ cells is necessary. It would be deeply interesting to improve culture conditions, to apply bolting factors, such as small molecules deterring from the molecular block that hinders a full germ stem cells (GSCs) or haGSC conversion to molecular pluripotent stem cells, and to enlarge the restricted time window of SSC to pluripotent stem cell (PSC) conversion.

Biography:

Ivana Haunerova is a Quality Assessor for biotech and advanced therapy medicinal products in the State Institute for Drug Control in the Czech Republic. She has completed her graduation in Biochemistry at the Institute of Chemical Technology, Prague. Since 2009, she is a Member of the Committee for Advanced Therapies at EMA.

Abstract:

Development of an advanced therapy medicinal products and their shift from the basic research to the clinical application is very difficult task. The legislation and regulatory requirements are in force but the research at this area is so rapid that we are facing new and new challenges. On one hand, it is necessary to define the rules so that the regulatory field is transparent,consistent and the decisions are predictable. As a consequence, there are some situations when the requirements seem to be too restrictive. On the other hand, there is a strong wish to have effective treatment on the market as soon as possible.Regulators need to balance both point of views. The national regulatory authorities and especially European Medicine Agency(EMA) offer different procedures that can be used by the product developers. These procedures such as classification of the borderline products, certification and scientific advices can be very helpful for the planning of development and saving time and resources, if used appropriately. At the beginning of the planning of cell-based product development, it is important to know what the regulatory framework for the specific product is, what are the procedures that can or should be used and what is the manufacturer goal (national hospital production or European market). The risk-based approach should be used to define risk and other important aspects but also the requirements that are not relevant for this particular case. Those findings must be taken into consideration during the whole development phase.

Biography:

Toshio Miki is an Associate Professor of Surgery at the Keck School of Medicine of USC. He has received his MD and PhD degrees from Nihon University in Tokyo, Japan. With the goal of overcoming the organ shortage faced by patients awaiting liver transplants, he started studying Xenotransplantation and Hepatocyte Transplantation. In his search for alternative cell sources for hepatocyte transplantation, he discovered stem cell-like populations in the human amnion in the year 2005. Since then, he has been best-known for his studies of placental stem cells and their clinical applications.

Abstract:

Placental stem cells are a readily available cell source for regenerative medicine, which can be procured in a non-invasive manner, and there are few ethical concerns regarding their use. The hepatic differentiation potential of human amniotic epithelial cell (hAEC) suggests usefulness in cell therapies for congenital liver metabolic disorders including ornithine transcarbamylase (OTC) deficiency. OTC deficiency is the most common urea cycle disorder, which can cause severe brain damage and death. To determine if hAEC transplantation has a therapeutic effect against OTC deficiency, we transplanted hAECs into the liver pulp of wild type (WT) and spf/ash mice and measured the following parameters: Engraftment potential,OTC enzymatic activity, levels of urine metabolite markers and behavioral response to excess ammonia. We present our findings that hAECs can engraft in immunocompetent mouse livers and provide a therapeutic effect. While OTC enzyme activity from untreated spf/ash controls produced 12.75±0.91 μM citrulline/mg protein/h, spf/ash mice treated with hAECs (spf/ash-hAEC)produced increased activity at 19.08±1.33 μM citrulline/mg protein/h (p<0.001). In addition, spf/ash-hAEC mice possessed levels of urine metabolites more closely to levels found in WT controls (p<0.05), confirming the improvement in OTC enzyme activity. Lastly, spf/ash-hAEC mice better tolerate ammonia challenge, exhibiting improved behavioral phenotypes compared to untreated controls (p<0.01). This growing body of evidence better suggests that hAECs can be used for cell replacement therapy to treat OTC deficiency.

Biography:

Željka Večerić-Haler is a Medical Doctor and Specialist of Nephrology. As a Nephrologist, she faces numerous challenges from the field of Regenerative Medicine on daily basis. She has completed her PhD from University of Ljubljana, Faculty of Medicine in 2016. Her research interests include studies on stem cell transplantation.

Abstract:

Mesenchymal stem cells (MSCs) are one of the promising tools in regenerative medicine. Non-clinical studies have shown that MSC transplantation markedly improved animal survival as well as functional and morphological parameters of various injured organs. Beneficial effects of MSCs can be attributed to different mechanisms such as their ability to secrete various immunomodulatory molecules as well as other soluble factors that have antiapoptotic, antifibrotic and antioxidative effects. Despite their beneficial effects, the outcome of the MSC transplantation is not always successful. It is suggested that one of the major problems influencing the efficacy of stem cell therapy is the poor MSCs survival following transplantation.Placing MSCs into a foreign tissue or harmful microenvironment may defeat their beneficial effects due to overload of proinflammatory cytokines or insufficient resistance of transplanted stem cells to oxidative and inflammatory stresses at the injured sites. It is interesting that most of the studies that evaluated beneficial effects of human MSCs in cisplatin-induced acute kidney injury have been so far performed on immunocompromised animals; although it is known that both innate and adaptive immune system is important contributor to the pathogenesis of acute kidney injury and can significantly affects the extent of nephrotoxicity. For instance, severly immunocompromised animals are susceptible to cisplatin nephrotoxicity, while mice without T lymphocytes, CD4+ or CD8+ T cells are protected against cisplatin nephrotoxicity. We evaluated the efficacy of MSC transplantation on mice with normal and compromised immune system and found out that difference in immunological environment affected not only the level of inflammation and oxidative stress at the site of injury but beneficial effects of MSC as well.

Biography:

Ingrid Lang has completed her Masters and her PhD degree from Karl-Franzens University, Graz. After additional studies and experience as High School Teacher in Biology, she was appointed as Assistant Professor and subsequently as Associate Professor of Histology and Embryology at the Institute of Cell Biology, Histology and Embryology, Medical University Graz. She has established a method for the isolation and characterization of fetal and adult endothelial cells from the macro and the microvasculature. She has experience in the field of “placenta research, stem cells and vascular biology”. She has published more than 60 articles in reputed journals.

Abstract:

We recently showed that placental-derived mesenchymal stem/stromal cells (PMSC) enhance endothelial cell viability and network formation of endothelial cells in vitro and induce angiogenesis in a mouse model in vivo by the secretion of paracrine factors or direct cell-cell contact. Thus, beside potential therapeutic aspects in disorders caused by insufficient angiogenesis such as chronic wounds, PMSC are promising tools for supporting vessel formation in engineered tissue constructs. Here, we assessed the impact of PMSC on the integrity and stability of endothelial cells in vascular grafts exposed to shear stress. ePTFE grafts were used to culture endothelial cells derived from the human placenta (hPEC) and PMSC in a 3D tissue culture model. hPEC were attached to the inner graft surface and a constant low (0.015 dyne/cm2) or physiological flow (0.92 dyne/cm2) was applied by a perfusion system. hPEC were supported by PMSC attached to the outer graft surface.PMSC improved the viability of hPEC exposed to 0.015 and 0.92 dyne/cm2 as shown by a decreased LDH release of 18% and 47%, respectively. Angiogenesis array analysis revealed that hPEC exposed to 0.92 dyne/cm2 secreted decreased levels of GRO,MCP-1, TIMP1, TIMP2 and angiogenin under co-culture with hAMSC. hPEC exposed to flow stimulated the migration of hAMSC. Our data demonstrate that PMSC improve adhesion, viability, and stability of endothelial cells in perfused vascular prostheses. Increased angiogenic properties and enhanced migration of cells favor endothelialization in order to provide a non-thrombogenic surface of small diameter vascular grafts in clinical use.

Biography:

Heyong Yin graduated in July 2016 with a master’s degree in surgery at Nankai University and Chinese PLA General Hospital. His researches are focus on Cartilage tissue engineering and stem cell. Since September 2016 he joins department of surgery, University of Munich (LMU) as a PhD candidates and continues his research into regenerative treatment of orthopaedics injuries to ligaments, tendon and hyaline cartilage.

Abstract:

We propose a method of preparing a novel cell carrier derived from natural cartilage extracellular matrix (ECM),designated cartilage ECM -derived particles (CEDPs). Through a series of processes involving pulverization, sieving,and decellularization, fresh cartilage was made into CEDPs with a median diameter of 263±48 μm. Under microgravity culture conditions in a rotary cell culture system (RCCS), bone marrow stromal cells (BMSCs) can proliferate rapidly on the surface of CEDPs with high viability. Histological evaluation and gene expression analysis indicated that BMSCs were differentiated into mature chondrocytes after 21 days of culture without the use of exogenous growth factors. Functional cartilage microtissue aggregates of BMSC-laden CEDPs formed as time in culture increased. Further, the microtissue aggregates were directly implanted into trochlear cartilage defects in a rat model (CEDP+MSC group). Gait analysis and histological results indicated that the CEDP+MSC group obtained better and more rapid joint function recovery and superior cartilage repair compared to the control groups, in which defects were treated with CEDPs alone or only fibrin glue, at both 6 and 12 weeks after surgery.In conclusion, the innovative cell carrier derived from cartilage ECM could promote chondrogenic differentiation of BMSCs,and the direct use of functional cartilage microtissue facilitated cartilage regeneration. This strategy for cell culture, stem cell differentiation and one-step surgery using cartilage microtissue for cartilage repair provides novel prospects for cartilage tissue engineering and may have further broad clinical applications.