Autologous approaches to tissue engineering

This is the latest version of this chapter. The previous version can be found here.

  • Beatrice Dionigia,b,
  • Dario O. Fauzaa,*

aDept. of Surgery, Boston Children's Hospital and Harvard Medical School

bDept. of Surgery, Brigham & Women's Hospital and Harvard Medical School

Stem cells have added a new thrust to tissue engineering. Their distinctive self-renewal and plasticity have not only optimized many tissue engineering developments, but also rendered feasible some applications which would otherwise be unattainable with somatic cells. This review focuses on general aspects of autologous tissue engineering based on so-called adult stem cells, which not only allow for a number of the therapeutic strategies envisioned for reprogrammed and embryonic stem cells, but also tend to be genetically more stable than the latter, not to mention less restrained by regulatory hurdles and ethical controversy. While the breadth of stem cell-based tissue engineering has yet to be defined in various large scale clinical applications, numerous experimental reports and the early human experience underwrite its potential. The field continues to advance robustly in an ever expanding number of laboratories around the world, yet much remains to be learned and developed, not only by scientists and clinicians, but also entrepreneurs and regulatory agencies. Nevertheless, given its scientific premises, the potential magnitude of its impact to society, and what has been achieved thus far, the perspective of stem cell-based tissue engineering reaching the mainstream of clinical practice has become a justifiable realistic expectation.

1. Introduction

The promise of tissue engineering has predictably gained renewed impetus with the addition of stem cells to its armamentarium. While the broader definition of tissue engineering includes a few non cell-based methods, the vast majority of its processes comprise different forms of cell therapies, for which stem cells, with their distinctive self-renewal and plasticity, are particularly suitable. In addition, stem cells allow for certain engineering strategies that would be impossible with other cell types. For example, the fact that it has been difficult to expand a sizeable proportion of somatic cells in sufficiently large numbers ex vivo for the engineering of many tissues or organ substitutes can, often times, be overcome by the use of stem cells. Indeed, the virtually limitless conceptual viability of autologous approaches to tissue engineering is one of the most direct consequences of this very principle.

While the breadth of stem cell-based tissue engineering has yet to be defined in different large scale clinical applications, numerous experimental reports and the initial clinical experience underwrite its potential. Still, perhaps the most compelling substantiation of this principle is nature's numerous precedents of tissue regeneration stemming from naturally-occurring (tissue-specific or not) stem cell activity. Indeed, the biomimetic stimulation of native stem cells has been an actively pursued strategy in regenerative medicine for some time now. This concept, however, is beyond the scope of this chapter. Other alternative approaches equally beyond this review include stem cell-mediated gene therapy; tolerance induction; embryonic stem cells; somatic cell nuclear transfer; and induced pluripotent stem cell (iPS) reprogramming as autologous approaches to stem cell-based tissue engineering – all of which are covered in other chapters. This review focuses on general aspects of tissue engineering based on so-called adult stem cells, which not only allow for a number of the therapeutic strategies envisioned for reprogrammed and embryonic stem cells, but also tend to be genetically more stable than the latter, as well as disconnected from their specific regulatory hurdles and eventual ethical controversy.

2. Cell sources

Different putative sources of adult stem cells have been identified or suggested almost regularly over the last several years. The search for alternative sites from which to isolate these cells and/or for new means to obtain them has been one of the most prolific aspects of the whole field, with an abundance of reports to date. Adult progenitor cells have been isolated from bone marrow (Pittenger, Mackay et al. 1999; Jiang, Jahagirdar et al. 2002), peripheral blood (Amos and Gordon 1995; Smith and Storms 2000), umbilical cord blood (Amos and Gordon 1995; Romanov, Svintsitskaya et al. 2003), placenta (Rubinstein, Carrier et al. 1998; Fauza 2004), amniotic fluid (Torricelli, Brizzi et al. 1993; Kaviani, Perry et al. 2001; Prusa and Hengstschlager 2002; Fauza 2004; Tsai, Hwang et al. 2006), amniotic membrane (Sakuragawa, Enosawa et al. 2000; Takahashi, Enosawa et al. 2002), Wharton jelly (Wang, Hung et al. 2004), adipose tissue (Zuk, Zhu et al. 2001; Wagner, Wein et al. 2005; Kern, Eichler et al. 2006), dermis (Toma, Akhavan et al. 2001; Young, Steele et al. 2001), hair follicle (Amoh, Li et al. 2005), synovial membrane (De Bari, Dell’Accio et al. 2001), skeletal muscle (Lee, Qu-Petersen et al. 2000; Young, Steele et al. 2001), central nervous system (Snyder, Deitcher et al. 1992), olfactory bulb (Liu and Martin 2004), retina (Coles, Angenieux et al. 2004), inner ear (Li, Liu et al. 2003), gastrointestinal epithelium (Bjerknes and Cheng 2002), fetal liver (Amos and Gordon 1995; Krupnick, Balsara et al. 2004), and likely others by the time this goes to print. Different progenitor cell types, including mesenchymal, epithelial, hematopoietic, neural, endothelial, and trophoblastic have been identified from all these sources, some of which harbor more than one type of progenitor cell. More recently, it has been shown that certain specialized stem cells can be isolated from certain sources in the presence of disease, such as for example the isolation of neural stem cells from amniotic fluid in the setting of neural tube defects (Turner, Klein et al. 2012). At the same time, however, despite the apparent surplus of reports on stem cell sources, many suffer from incomplete analyses and still require further in depth validation. Most of the more widely used cell isolation methods rely on different combinations of natural selection by the culture media with some form of direct cell isolation, typically mechanical, magnetic, or immune-based. In addition, oxygen tension manipultations, multi-stage cultures, and numerous alternative media formulations have also been explored, among many other variations of this principle.

The stem cell source can impact the algorithm for eventual clinical translation of a given tissue engineering strategy. Phenotypically comparable stem cells may behave quite differently, depending on the cell source considered (Wagner, Wein et al. 2005; Chang, Shih et al. 2006; Kern, Eichler et al. 2006; Kunisaki, Fuchs et al. 2007). Such differences can have a significant bearing not only on cell processing in vitro, but also on many relevant aspects of the engineered tissue made from comparable stem cells (Kunisaki, Fuchs et al. 2007). For example, mesenchymal stem cells (MSCs) procured from amniotic fluid proliferate significantly faster in vitro than immunophenotypically equivalent MSCs obtained from bone marrow or cord blood and lead to a very peculiar form of engineered cartilage, unusually rich in both glycosaminoglycans and α-elastin, when compared with constructs originated from these other MSCs, under equal bioreactor conditions (figure 1) (Kunisaki, Fuchs et al. 2007).

image
Figure 1.

Typical gross appearance of a tubular cartilaginous construct engineered from amniotic mesenchymal stem cells.

The goal of generating clinically relevant engineered tissue places specific requirements on the cell type/source, including minimally invasive accessibility, the ability to produce an inordinately large quantity of cells in a relatively short period of time, hardiness to often prolonged in vitro processing, and reproducible differentiation pathways, among others. These parameters have favored certain stem cell types over others. Mesenchymal stem cells are among the most broadly utilized stem cells in tissue engineering due to their diverse sources, self-renewal pattern, and multilineage potential (Bianco and Robey 2001; Barrilleaux, Phinney et al. 2006). Indeed, most of the tissues used for structural surgical repair are mesenchymal in nature. Further, in addition to all mesenchymal lineages, certain MSCs such as those from umbilical cord blood and amniotic fluid can also differentiate into cells from different germ layers, sometimes all three of them. This broadens the appeal of these cells even further. While many sites have been shown to yield MSCs, the bone marrow remains the most studied and best characterized source, thus it is often the benchmark for comparisons involving MSCs. At the same time, nonetheless, MSC isolation and expansion from the bone marrow is more difficult than from other sources and can be significantly influenced by the donor's age (Vaananen 2005; Kunisaki, Fuchs et al. 2007). Indeed, “alternative” sources of MSCs, such as the amniotic fluid and adipose tissue, have recently received increasingly more attention for tissue engineering applications, due to their better translational appeal in many clinical scenarios, when compared with bone marrow and other sources (figure 2) (Kaviani, Perry et al. 2001; Kaviani, Guleserian et al. 2003; Awad, Wickham et al. 2004; Fuchs, Kaviani et al. 2004; Dragoo, Lieberman et al. 2005; Kunisaki, Freedman et al. 2006; Kunisaki, Fuchs et al. 2006; Kunisaki, Jennings et al. 2006; Kunisaki, Armant et al. 2007; Kunisaki, Fuchs et al. 2007; Stosich and Mao 2007; Steigman, Armant et al. 2008; Steigman, Ahmed et al. 2009; Klein, Turner et al. 2010; Turner, Klein et al. 2011; Gray, Turner et al. 2012; Turner, Klein et al. 2012).

image
Figure 2.

Diagramatic representation of the concept of autologous amniotic mesenchymal stem cell-based fetal tissue engineering for the treatment of congenital anomalies.

3. Translational challenges

In order for novel therapeutic concepts and methodologies, such as those implicated in the development of autologous stem cell-based tissue engineering, to be brought to clinical fruition, many biological, technical, and regulatory hurdles must be overcome. While many of these hurdles are disease-specific, one can generalize certain aspects of the translational challenges involved.

3.1. Biological challenges

Timing is an intrinsic constraint in most tissue engineering concepts. Autologous construct-based approaches generally involve weeks, if not months, of processing ex vivo before final implantation. While the use of stem cells can often minimize this limitation, at least when compared with the use of most somatic cells, more often than not it remains a concern.

Even autologous cells may not be completely free of pathogens in culture, as the growth medium often requires xenogeneic products, such as fetal bovine serum. In addition, some cells can only propagate consistently on xenogeneic feeder layers, so that infectious risks cannot be completely eliminated.

The search for the ideal biocompatible scaffolding material(s) for a given clinical application is almost a never-ending pursuit. The possibility that newer components may prove better than what has been considered state of the art is an ever-present prospect. Many of the currently available synthetic scaffolds remain plagued by foreign body reaction in the setting of an immunocompetent environment, which in turn can lead to a reduction in the diffusion of nutrients and waste products, fibrosis, and other complications. Also, the cytotoxic effects of macrophage-generated nitric oxide can reach and destroy transplanted cells. Thus, it is not surprising that most of the scaffolds implanted in humans to date have been derived from natural sources. On the other hand, natural scaffolds are usually linked to unfavorable biomechanical properties and excessive heterogeneity. Also, chemicals used in decellularization processes often negatively affect different properties of the scaffold. Not surprisingly, the search for enhanced biocompatible synthetic biomaterials, such as elastomers, nanostructures, and others, remains a lively aspect of the development of tissue engineering, regardless of whether stem cell- or somatic cell-based. For example, scaffolds impregnated with select growth factors or specific peptide sequences may also allow for better control of the surrounding microenvironment. Undeniably, advances in synthetic materials will be instrumental in broadening the reach of tissue engineering both in the immediate and long term future.

3.1.1. Construct vascularization

The search for methods to establish and/or control the vascularization of engineered tissues or organs has been one of the major foci in the recent development of tissue engineering. Although the scaffold and three-dimensional milieu play a role, typically constructs greater than approximately 1cm in thickness cannot rely solely on vascular ingrowth from the host to remain viable in vivo (Davis, Schroeder et al. 2007). Thus, a major hurdle for clinical application of large engineered tissues and organs has been securing blood supply to the graft at the time of implantation. An appealing strategy among the many being currently explored to overcome this hindrance has been to build a microcirculation network within the engineered scaffold itself. One interesting example of such an approach has been the use of MicroElectroMechanical Systems (MEMS) (Fidkowski, Kaazempur-Mofrad et al. 2005). Advances in MEMS have enabled the development of a robust computational model of the vascular microcirculation, including variables such as fractal network topology, blood flow rheology, and the mass transfer of oxygen and nutrients across the vascular bed. This method has allowed the etching of vascular channels onto silicon wafers, which can then be transferred onto biodegradable polymer systems by stacking multiple monolayers of this architecture so as to form three-dimensional structures. Some of these strategies have already reached preclinical stage in large animal models (Hsu, Carraro et al. 2010). Another approach to organ revascularization has involved using the native vasculature of a decellularized organ for the seeding of donor endothelial cells in a bioreactor prior to anastomosis of the engineered graft, to date only attempted in rodent models (Ott, Clippinger et al. 2010). Various other approaches to establish or optimize construct vascularization, such as gene therapy, biomimetic microvascular guides and/or microfluidic networks, and scaffold-based delivery of angiogenic and/or growth factors, among others, have also been pursued (Wong Po Foo, Patwardhan et al. 2006; Brewster, Brey et al. 2007; Shen, Kastrup et al. 2008; Borenstein, Tupper et al. 2010; Loai, Yeger et al. 2010). Conceivably, many of the principles established by these developments might be also be eventually applied in facilitating the formation of other complex ancillary networks within engineered grafts, such as neural, lymphatic, and biliary systems, for example (Allmeling, Jokuszies et al. 2006).

4. Regulatory challenges

The Food and Drug Administration (FDA) has mandatory jurisdiction over cell-based therapies in The United States, including tissue engineering. An elaborate and costly infrastructure is necessary for the development and manufacture of engineered tissue amenable to FDA validation. Such regulatory clearance demands the use of so-called Good Manufacturing Practice (GMP) facilities, which not only must fulfill strict physical and operational requirements, but also be controlled by a critical mass of highly trained personnel. Another practical intricacy is the fact that certain tissues require preconditioning in complex bioreactors, which may not be readily compatible with large scaled manufacturing and shipping (Griffith and Naughton 2002). All of these underlying hurdles translate into chronic difficulties in establishing multicentric clinical trials, which are essential for the widespread application of technologies such as this.

Regulatory constraints have significantly hampered the clinical translation of many tissue engineering therapies (Lee, Arcidiacono et al. 2010). The FDA has often been criticized for slow approval processes, which include both justifiable and debatable requirements. This predicament has been attributed, at least to some extent, to the lack of clear, predictable regulatory frameworks and to uncertainties regarding the proper classification of different tissue engineered products. This is particularly evident when stem cells are to be used, in that, besides eventual ethical concerns, they typically trigger regulatory demands for unique safety data sets, more notably on genomic stability and tumorigenesis. Fortunately, it seems that the FDA is mindful of this scenario and is working to address these concerns. Meanwhile, however, many American biotechnology companies have engaged in collecting clinical data overseas, at lower costs, as most other countries have less stringent regulatory procedures (Atala, Bauer et al. 2006; McAllister, Maruszewski et al. 2009).

Regardless, these constraints, combined with strict reimbursement policies and poor business models, have typically rendered tissue engineering products commercially unsustainable in the long run (Fauza 2003). Indeed, a sizeable proportion of firms with considerable interest in tissue engineering have exited the market over the last 10 plus years. On the other hand, a number of companies still continue to invest in the development of new products (Lysaght and Reyes 2001; Vacanti 2008). It seems that a healthy partnership between academia and industry should be further explored as a means to expand tissue engineering into clinical reality on a more meaningful scale, a principle which has been proven viable in a number of circumstances (Fauza 2003; Vacanti 2008).

5. Current clinical applications

The infancy of the field, combined with the different challenges briefly discussed above, elucidate why few controlled prospective trials have validated clinical tissue engineering applications to date. Indeed, over the last decade a number of tissue-engineered products have either been abandoned following phase I/II clinical trials, or have failed in phase III clinical testing (Lysaght and Hazlehurst 2004). Very many challenges remain to be overcome by companies before “off-the-shelf” tissues can be offered commercially, including adequate sources of healthy expandable cells, the optimization of scaffolds, scaled-up bioreactors, the prevention of tissue rejection, and optimal product preservation ex vivo. Many of these challenges can be better tackled by the use of stem cells. Perhaps the best and most successful example is hematopoietic stem cell transplantation, which, first performed in humans during the 1950s, has long been firmly established clinically and will not be further explored here. Below is a brief yet illustrative sampling of some of the recent clinical experiences with tissue engineering. It is by no means purported to be a comprehensive, all-inclusive list. In that regard, we must remember that, although some still link the term “tissue engineering” solely to the paradigm of cell delivery within biocompatible scaffolds, the field is certainly much broader (Langer and Vacanti 1993; Vacanti and Langer 1999). In essence, tissue engineering technologies fall into one of four main strategies: delivery of isolated/expanded cells (simple cell transfers); tissue-inducing substances (not applicable to this review); extracorporeal and encapsulation techniques (closed systems); and cell transplantation within three-dimensional matrices (open systems).

5.1. Cardiovascular repair

End stage congestive heart failure and myocardial infarction remain the leading causes of death in the United States. The underlying process in these forms of cardiac disease seems related to boss a loss of functional cardiomyocites and to the inability of the myocardium to regenerate. The myocardium cannot repair itself at least in part because of a paucity of myocardial stem cells, not to mention the disease process and the consequential local hostile environment. The end result is fibrotic scarring, resulting in a decrease in the ventricular ejection fraction, diastolic dysfunction, and an overall decline in mechanical performance.

Over the last several years, the local delivery of autologous skeletal muscle satellite cells or bone marrow-derived stem cells, a procedure known as cellular cardiomyoplasty, has been evaluated as an experimental therapy in patients with ischemic myocardium. To date, hundreds of patients have received this therapy worldwide. This approach aims to minimize fibrotic remodeling of the injured myocardium by populating the damaged area with myogenic precursor cells. Support for this hypothesis has been based largely on animal data, which has shown that implanted cells can differentiate into multinucleated myocyte-like cells, resulting in improved global ventricular performance without the need for electromechanical coupling between donor cells and host cardiomyocytes (Murry, Field et al. 2005).

At this time, the clinical feasibility of this form of cell transfer in the management of ischemic heart disease has mostly been reported in multiple descriptive, pilot studies (Murry, Field et al. 2005). It has been shown that hundreds of millions of myoblasts can be grown from a small muscle biopsy under GMP conditions. Several uncontrolled studies have demonstrated modest improvements in ejection fraction and regional wall activity after skeletal myoblast transplantation (Herreros, Prosper et al. 2003; Menasche, Hagege et al. 2003; Siminiak, Kalawski et al. 2004). At least two small randomized trials using autologous bone marrow mesenchymal cells have also demonstrated improvements in cardiac performance, including increased myocardial fluorodeoxyglucose uptake, enhanced wall motion, a reduction in ventricular end-systolic and end-diastolic volumes, and a 14% net increase in ejection fraction, when compared with a saline-infused control group (Chen, Fang et al. 2004; Murry, Field et al. 2005).

The optimal cell type for this therapeutic application, however, remains to be defined (Elnakish, Hassan et al. 2012). At the same time, the mechanisms for the observed clinical benefits of cellular cardiomyoplasty remain unknown. Because cell survival after the procedure appears to be quite low, proponents have speculated that the improved myocardial indices may be secondary to other factors, such as increased angiogenesis, minimization of deleterious ventricular remodeling, and/or enhanced cytokine-mediated resident cell survival. Further studies are certainly needed in order to adequately assess the risks and benefits of cellular cardiomyoplasty. Also, whether or not the direct myocardial injection of donor cells may also be associated with tumors and/or an increased risk for malignant arrhythmias warrants further scrutiny.

Several products based on the principle of cellular cardiomyoplasty have been, or are currently being tested in controlled clinical trials, both in the United States and in Europe. One such therapy, marketed as MyoCell™ (Bioheart, Inc.) involves a collaboration of multiple centers and has entered phase III trials as a result of encouraging initial results (Haider, Lei et al. 2008). In this approach, autologous skeletal myoblasts are expanded ex vivo and supplied as a cell suspension to be delivered directly into the epicardium during coronary artery bypass grafting surgery. In the United States, a donor-derived bone marrow preparation, marketed as Prochymal™ (Osiris Therapeutics) is being evaluated in a phase I trial, with preliminary data suggesting that the product is safe when administered intravenously soon after myocardial infarction.

5.1.1. Congenital anomalies

Congenital heart disease is the leading cause of neonatal death from birth defects. Single ventricle anomalies are particularly threatening, often requiring multiple operations within the first few months of life in order to prevent irreversible congestive heart failure. A procedure commonly performed in these patients includes the separation between the pulmonary and systemic circulations by creating a cardiac total cavopulmonary connection using a biocompatible synthetic conduit (e.g., Dacron or Teflon). Unfortunately, these synthetic conduits are prone to thromboembolism and infection, and do not grow with the patient. As a consequence, many of these patients undergo risky re-operations to revise the conduit, in procedures associated with significant mortality and morbidity. A tissue engineered blood vessel conduit composed of autologous cells seeded on a biodegradable scaffold would avoid the many disadvantages associated with synthetic conduits. In both large animal models and humans, investigators have shown that tissue-engineered blood vessels can be made using a tubular scaffold composed of polyglycolic acid woven with either ε-caprolactone or L-lactide seeded with autologous bone marrow mononuclear cells for just 2 hours prior to implantation (Shin’oka, Matsumura et al. 2005). At least 25 children with congenital heart disease were followed for over 9 years after having received comparable tissue-engineered grafts, with encouraging results (Shin’oka, Imai et al. 2001; Hibino, Shin’oka et al. 2005; Hibino, McGillicuddy et al. 2010).

This early experience pointed to the feasibility and safety of clinical tissue engineering in pediatric cardiac surgery, but also uncovered the complication of graft stenosis secondary to neotissue formation in up to 20% of patients (figure 3) (Duncan and Breuer 2011). Fortunately, so far such complication has generally been amendable to percutaneous angioplasty. After further animal studies, the first U.S. patients were approved by the FDA to receive engineered vascular implants in a controlled trial starting in 2012. It remains to be determined whether the next generation of engineered blood vessel conduits will be associated with lower long-term morbidity after pediatric cardiac repair (Naito, Shinoka et al. 2011).

image
Figure 3.

Growth potential of human engineered vascular grafts. A) Magnetic resonance image (MRI) 9 months following implantation. B) Three-dimensional computed tomography (CT) angiogram one year after implantation. Red arrows indicate location of the implant.

Anecdotal clinical experience with heart valves engineered from autologous endothelial progenitor cells has also started to be reported, with encouraging results (Cebotari, Lichtenberg et al. 2006). This methodology benefited from the presence of a small population of CD34 positive mononuclear hematopoietic progenitor cells in human peripheral blood capable of differentiating into the endothelial lineage in culture (Asahara, Murohara et al. 1997; Assmus, Schachinger et al. 2002), as this obviates the need for vascular procurement. Nevertheless, in addition to the endothelial lining, a mesenchymal population is typically needed to maintain the extracellular matrix and overall integrity of the valvular construct, for which MSCs have been explored. The optimum cell sources and scaffold –cell interactions remain to be established. Yet another significant and somewhat unique limitation of heart valve engineering stems from the complex and dynamic demands of the constantly moving three-dimensional environment, as well as its long term stability, particularly in pediatric applications. Still, in a separate study involving 11 patients, autologous endothelial progenitor cells isolated from a forearm vein were used to seed valve allografts with good long-term results at 10 years follow up (Dohmen, Lembcke et al. 2011). At the same time, given that most conventional acellular allografts tend to fail between 15 and 20 years after implantation, longer term follow up will be necessary in order to possibly establish the clinical validity of heart valves engineered with this methodology.

5.2. Neural repair

Parkinson's disease is a prevalent and debilitating neurodegenerative disorder caused by a selective loss of mesencephalic dopaminergic neurons within the substantia nigra. Affected patients have characteristic symptoms, including bradykinesia, resting tremors, muscle rigidity, and gait disturbance. Current medical therapy, including the exogenous administration of dopamine (levodopa), can be effective in many patients but in some is associated with numerous secondary motor complications.

Another type of simple cell transfer, namely the transplantation of human fetal ventral mesencephalic dopaminergic cells, has been studied for many years as a potential treatment for Parkinson's disease. Initial observational studies suggested prolonged survival of the transplanted cells, as well as substantial clinical neurological improvements in the absence of medication, with some recovery of the nigrostriatal pathway for more than a decade following the operation. Nevertheless, at least two randomized controlled trials comparing the effectiveness of human fetal cell transplantation with sham surgery demonstrated no significant differences between the two groups, except for some modest gains in a cohort of younger patients (Freed, Greene et al. 2001; Olanow, Goetz et al. 2003). Furthermore, both the practical and regulatory limitations of human fetal cell transplantation have precluded its widespread clinical application. Moreover, the transplantation of human fetal ventral mesencephalic dopaminergic cells has also been associated with the occurrence of adverse effects in the mid to long term, such as graft-induced dyskinesias (GIDs) stemming from graft-derived striatal serotonergic hyperinnervation (Politis, Oertel et al. 2011).

A number of investigators have shifted their focus from fetal cell transplantation to approaches that employ neural stem cells, which have a high capacity for self-renewal and could conceivably supply an abundant number of specialized neurons for the treatment of Parkinson's disease. One biotechnology company, NeuroGeneration, is seeking regulatory approval for phase II clinical trials of a strategy that includes neural stem cell-derived dopaminergic cells delivered into the affected striatal structures of patients with Parkinson's Disease. Other methodologies encompassing different cell sources/types and/or delivery methods continue to be pursued, though also have yet to be validated (Braak and Del Tredici 2008; Rossler, Boddeke et al. 2010). The use of neural and other stem cells in spinal cord repair is of course another area of even greater experimental activity, not within the scope of this review.

5.3. Bone repair

Different forms of bone loss can occur in a variety of pathological entities. Fairly established bone replacement methodologies include autologous and allogeneic bone grafts, demineralized bone, and numerous natural or synthetic bone substitutes, all still fraught with substantial limitations. Thus, in addition to growth factors and cytokines such as Bone Morphogenic Proteins (BMPs), the use of cell-based methods has been explored. Mesenchymal stem cells are natural candidates for bone replacement and indeed a number of anecdotal cases and small series of MSC-based tissue engineering to that end have been reported to date. Injectable suspensions of bone marrow MSCs have been used as a means to enhance bone healing in the treatment of congenital and acquired bone defects for decades (Salama and Weissman 1978; Jackson, Scheker et al. 1981; Healey, Zimmerman et al. 1990; Kitoh, Kitakoji et al. 2004; Hibi, Yamada et al. 2006; Hibi, Yamada et al. 2006). However, the first instance of bone repair using a three-dimensional cell-seeded construct, by Vacanti et al., took place only in the late 1990’s, on a patient with traumatic avulsion of a distal phalanx, in whom partial local bone formation was documented post-operatively (Vacanti, Bonassar et al. 2001). In this case, no stem cells were used, but rather osteoblasts obtained from a periosteal biopsy. Reports of different forms of bone repair with bone marrow MSC-seeded three-dimensional constructs have followed, typically in very small series or case reports (Ohgushi, Goldberg et al. 1989; Quarto, Mastrogiacomo et al. 2001; Warnke, Springer et al. 2004; Marcacci, Kon et al. 2007). Although variable degrees of osteoregeneration have been documented in these studies, the role of stem cell-based tissue engineering in bone repair remains to be properly defined. One additional interesting development, also still in its infancy, is the use of MSCs, for example procured from fetal liver, delivered prenatally to ameliorate genetic bone disorders, such as osteogenesis imperfecta (Horwitz, Prockop et al. 1999; Horwitz, Prockop et al. 2001; Guillot, Abass et al. 2008). Also here, while the results of the first clinical applications have been encouraging, much remains to be achieved before this principle, as all as the other MSC-based therapies discussed above, can be fully validated and broadly recommended.

5.4. Skeletal muscle repair

Duchenne muscular dystrophy (DMD) is an X-linked genetic disorder caused by a defect in the encoding for dystrophin, a muscle fiber stabilizing protein. The disease results in chronic injury to skeletal myocytes, leading to a vicious cycle of myocyte degradation and fibrosis. Most affected children are wheelchair-bound by early adolescence and, by early adulthood, most patients develop severe respiratory failure and cardiomyopathy. Despite significant advances in the understanding of the pathophysiology of DMD, its management continues to be largely supportive as no effective therapy yet exists for this disease.

One experimental approach to the treatment of DMD has been to enable relocalization and expression of normal functional dystrophin transcripts within the muscle through the transplantation of muscle precursor cells from normal donors, a procedure known as myoblast transfer therapy (MTT). This approach was originally shown to have promising results in animal models using mdx mice. Initial clinical experience showed evidence of dystrophin transcript expression in DMD patients by reverse-transcriptase polymerase chain reaction (Law, Goodwin et al. 1992). However, multiple clinical trials to date have not shown any objective benefit in DMD patients injected with donor myoblasts. In one of the earlier human studies, dystrophin-positive fibers comprised up to 36 percent of the injected muscles after 1 month (Gussoni, Pavlath et al. 1992). Nonetheless, such expression has generally been undetectable by 6 months post-injection. A subsequent study employing serial injections of normal myoblasts procured from unaffected relatives over a 6 month time period also did not show any clinical benefit (Mendell, Kissel et al. 1995).

These disappointing clinical results to date have likely been due to multiple factors, including poor cell survival, immune rejection, and limited cell distribution after injection. To our knowledge, there are no clinical trials of MTT currently ongoing. Still, the past few years have witnessed a renaissance in preclinical efforts to hopefully enable MTT eventually. These studies are attempting to address some of the shortcomings of the previous human trials, using enhanced immunosuppressive regimens and alternative stem/progenitor cell sources such as muscle-derived stem cells, among other approaches (Urish, Kanda et al. 2005).

5.5. Urologic repair

Complex urologic reconstructions of the bladder, ureter, and urethra have traditionally relied on the use of heterotopic autologous grafts such as stomach, intestine, or colon. None of these structures constitute ideal urologic replacements in that they lack urothelium and are associated with significant morbidity, including urolithiasis, metabolic disturbances, and malignant degeneration. Many investigators have long been evaluating tissue engineering approaches as potential means to overcome these common complications of urologic reconstruction, leading to some clinical experience.

Initial clinical applications of tissue engineering in urology focused on bladder augmentation (cystoplasty). This procedure is usually indicated for the management of high-pressure, low compliant bladders secondary to spina bifida, spinal cord injury, or urge incontinence. Although anti-cholinergic medication and intermittent bladder catheterization can be effective, surgical reconstruction with intestine remains as the standard in severe cases. To date, engineered bladders, referred to as Neo-Bladder AugmentTM (Tengion, East Norriton, PA), have been implanted into several children with myelomeningocele (MMC) and end-stage bladder disease, albeit only in a phase I trial (Atala, Bauer et al. 2006). Despite these early feasibility results, Tengion abandoned plans for phase II trials and further work in 2011, in large part due to the limited to no clinical efficacy and the occurrence of serious adverse events. The trial was also hampered by the lack of a control group (i.e., standard enterocystoplasty), as well as the absence of data on innervation, in part because of the underlying disease.

In contrast to bladder augmentation, one area of urologic tissue engineering that has received much attention in recent years is urinary diversion for bladder cancer (Basu, Jayo et al. 2012). Bladder cancer is one of the most common malignancies worldwide, with over 20,000 urinary diversions performed in the United States and Europe annually. The current standard for urinary diversion involves utilizing a segment of intestine to transport urine through a hole in the abdominal wall into a standard ostomy bag. As of early 2012, phase I clinical trials for Tengion's tissue engineered urinary conduit (Neo-Urinary ConduitTM) were ongoing at five U.S. centers in patients requiring urinary diversion following cystectomy for bladder cancer. Neo-Urinary ConduitTM carries urine from the ureters to the skin surface using a polylacetate glycolate scaffold seeded with autologous smooth muscle cells isolated from autologous abdominal fat.

Similar techniques have been used to implant other tissue-engineered urological structures in humans, including tubularized conduits for traumatic injury of the membranous urethra (Raya-Rivera, Esquiliano et al. 2011). Also here, further controlled analyses are needed to determine the value of tissue-engineered urethral replacements as an alternative to buccal mucosa or skin in patients requiring urethroplasty. Indeed, as it is the case in virtually all other reports involving clinical application of engineered grafts to date, the clinical experience with tissue engineering in urology lacks controlled trials, and none of the products are approved by the FDA as of this writing.

5.6. Airway reconstruction

Despite the relative rarity of surgical airway disease, engineered airway conduits have been of substantial interest for the treatment of a variety of airway ailments for many years now (Macchiarini, Walles et al. 2004; Kunisaki, Freedman et al. 2006; Macchiarini, Jungebluth et al. 2008; Lange, Fishman et al. 2011; Gray, Turner et al. 2012). Except for primary repairs, tracheal reconstructions remain frequently associated with suboptimal functional results and substantial morbidity and mortality. Although the trachea is a hollow conduit, its structure and biomechanical properties are in fact quite complex and demanding. Accordingly, the ideal engineered substitute needs be airtight, nonimmunogenic, noncollapsible, well-vascularized, well-epithelialized, and conducive to sustained chondrogenesis (Baiguera, D’Innocenzo et al. 2011).

The early clinical experience with tissue-engineered tracheas has been arguably promising. To date, a number of patients in Europe have undergone tracheobronchial replacement with a cellularized engineered construct, with interesting results (Macchiarini, Jungebluth et al. 2008; Baiguera, D’Innocenzo et al. 2011). Both decellularized airway and synthetic scaffolds have been used, usually seeded with autologous bronchial epithelial cells and bone marrow-derived MSCs and/or differentiated chondrocytes.

One limitation of the current approach in tracheal engineering has been the protracted time required to fabricate these constructs. In attempts to streamline the process, more recent techniques have incorporated the use of nano-composite polymer and growth factor-induced endogenous stem cell mobilization as means to enhance engraftment and remodeling in vivo (Jungebluth, Alici et al. 2011). At the same time, however, once more long-term follow-up, along with comparisons in controlled phase III trials will be essential before more widespread application of this technology can be justified.

6. Future perspectives

Tissue engineering remains in the early phase of its developmental curve. As evidenced in many examples discussed in this chapter and others, concrete clinical benefits from this technology have accrued in the past several years, albeit still at a much slower pace than what we can expect for the future, particularly as stem/progenitor cells become increasingly more studied in these applications. In addition to the therapeutic implications of the use of stem cells in tissue engineering, related studies can also lead to unique insights into more basic aspects of stem cell biology, which in turn may bring further, as yet unsuspected therapeutic developments. Although the widespread availability of “custom-made” tissues and organs has yet to become a reality, the field is both very young and very vibrant. Much remains to be learned and developed, not only by scientists and clinicians, but also entrepreneurs and regulatory agencies. Nevertheless, given its scientific premises, the potential magnitude of its impact to society, and what has been achieved thus far, it is reasonable to envision stem cell-based tissue engineering eventually reaching conventional clinical practice, from fetal medicine to geriatrics and the entire gamut in between.

References

Allmeling, C. Jokuszies, A. et al. (2006). “Use of spider silk fibres as an innovative material in a biocompatible artificial nerve conduit”. J Cell Mol Med 10(3), 770–777. Article Abstract

Amoh, Y. Li, L. et al. (2005). “Multipotent nestin-positive, keratin-negative hair-follicle bulge stem cells can form neurons”. Proc Natl Acad Sci U S A 102(15), 5530–5534. Article Abstract

Amos, T.A. Gordon, M.Y. (1995). “Sources of human hematopoietic stem cells for transplantation–a review”. Cell Transplant 4(6), 547–569. Article Abstract

Asahara, T. Murohara, T. et al. (1997). “Isolation of putative progenitor endothelial cells for angiogenesis”. Science 275(5302), 964–967. Article Abstract

Assmus, B. Schachinger, V. et al. (2002). “Transplantation of Progenitor Cells and Regeneration Enhancement in Acute Myocardial Infarction (TOPCARE-AMI)”. Circulation 106(24), 3009–3017. Article Abstract

Atala, A. Bauer, S.B. et al. (2006). “Tissue-engineered autologous bladders for patients needing cystoplasty”. Lancet 367(9518), 1241–1246. Article Abstract

Awad, H.A. Wickham, M.Q. et al. (2004). “Chondrogenic differentiation of adipose-derived adult stem cells in agarose, alginate, and gelatin scaffolds”. Biomaterials 25(16), 3211–3222. Article Abstract

Baiguera, S. D’Innocenzo, B. et al. (2011). “Current status of regenerative replacement of the airway”. Expert Rev Respir Med 5(4), 487–494. Article Abstract

Barrilleaux, B. Phinney, D.G. et al. (2006). “Review: ex vivo engineering of living tissues with adult stem cells”. Tissue Eng 12(11), 3007–3019. Article Abstract

Basu, J. Jayo, M.J. et al. (2012). “Regeneration of native-like neo-urinary tissue from nonbladder cell sources”. Tissue Eng Part A 18(9–10), 1025–1034. Article Abstract

Bianco, P. Robey, P.G. (2001). “Stem cells in tissue engineering”. Nature 414(6859), 118–121. Article Abstract

Bjerknes, M. Cheng, H. (2002). “Multipotential stem cells in adult mouse gastric epithelium”. Am J Physiol Gastrointest Liver Physiol 283(3), G767–777. Abstract

Borenstein, J.T. Tupper, M.M. et al. (2010). “Functional endothelialized microvascular networks with circular cross-sections in a tissue culture substrate”. Biomed Microdevices 12(1), 71–79. Article Abstract

Braak, H. Del Tredici, K. (2008). “Assessing fetal nerve cell grafts in Parkinson's disease”. Nat Med 14(5), 483–485. Article Abstract

Brewster, L. Brey, E.M. et al. (2007). Blood Vessels. Principles of Tissue Engineering. Lanza, R. Langer, R. Vacanti, J.P. San Diego: Academic Press; , 569–584. Article

Cebotari, S. Lichtenberg, A. et al. (2006). “Clinical application of tissue engineered human heart valves using autologous progenitor cells”. Circulation 114(1 Suppl), I132–137. Article Abstract

Chang, Y.J. Shih, D.T. et al. (2006). “Disparate mesenchyme-lineage tendencies in mesenchymal stem cells from human bone marrow and umbilical cord blood”. Stem Cells 24(3), 679–685. Article Abstract

Chen, S.L. Fang, W.W. et al. (2004). “Effect on left ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction”. Am J Cardiol 94(1), 92–95. Article Abstract

Coles, B.L. Angenieux, B. et al. (2004). “Facile isolation and the characterization of human retinal stem cells”. Proc Natl Acad Sci U S A 101(44), 15772–15777. Article Abstract

Davis, B.H. Schroeder, T. et al. (2007). “An in vitro system to evaluate the effects of ischemia on survival of cells used for cell therapy”. Ann Biomed Eng 35(8), 1414–1424. Article Abstract

De Bari, C. Dell’Accio, F. et al. (2001). “Multipotent mesenchymal stem cells from adult human synovial membrane”. Arthritis Rheum 44(8), 1928–1942. Article Abstract

Dohmen, P.M. Lembcke, A. et al. (2011). “Ten years of clinical results with a tissue-engineered pulmonary valve”. Ann Thorac Surg 92(4), 1308–1314. Article Abstract

Dragoo, J.L. Lieberman, J.R. et al. (2005). “Tissue-engineered bone from BMP-2-transduced stem cells derived from human fat”. Plast Reconstr Surg 115(6), 1665–1673. Article Abstract

Duncan, D.R. Breuer, C.K. (2011). “Challenges in translating vascular tissue engineering to the pediatric clinic”. Vasc Cell 3(1), 23. Article Abstract

Elnakish, M.T. Hassan, F. et al. (2012). “Mesenchymal stem cells for cardiac regeneration: translation to bedside reality”. Stem Cells Int (2012). , 646038. Article Abstract

Fauza, D. (2004). “Amniotic fluid and placental stem cells”. Best Pract Res Clin Obstet Gynaecol 18(6), 877–891. Article Abstract

Fauza, D.O. (2003). “Tissue engineering: current state of clinical application”. Curr Opin Pediatr 15(3), 267–271. Article Abstract

Fidkowski, C. Kaazempur-Mofrad, M.R. et al. (2005). “Endothelialized microvasculature based on a biodegradable elastomer”. Tissue Eng 11, 1–2. 302–. Article Abstract

Freed, C.R. Greene, P.E. et al. (2001). “Transplantation of embryonic dopamine neurons for severe Parkinson's disease”. N Engl J Med 344(10), 710–719. Article Abstract

Fuchs, J.R. Kaviani, A. et al. (2004). “Diaphragmatic reconstruction with autologous tendon engineered from mesenchymal amniocytes”. J Pediatr Surg 39(6), 834–838; discussion 834–838. Article Abstract

Gray, F.L. Turner, C.G. et al. (2012). “Prenatal tracheal reconstruction with a hybrid amniotic mesenchymal stem cells-engineered construct derived from decellularized airway”. J Pediatr Surg 47(6), 1072–1079. Article Abstract

Griffith, L.G. Naughton, G. (2002). “Tissue engineering–current challenges and expanding opportunities”. Science 295(5557), 1009–1014. Article Abstract

Guillot, P.V. Abass, O. et al. (2008). “Intrauterine transplantation of human fetal mesenchymal stem cells from first-trimester blood repairs bone and reduces fractures in osteogenesis imperfecta mice”. Blood 111(3), 1717–1725. Article Abstract

Gussoni, E. Pavlath, G.K. et al. (1992). “Normal dystrophin transcripts detected in Duchenne muscular dystrophy patients after myoblast transplantation”. Nature 356(6368), 435–438. Article Abstract

Haider, H. Lei, Y. et al. (2008). “MyoCell, a cell-based, autologous skeletal myoblast therapy for the treatment of cardiovascular diseases”. Curr Opin Mol Ther 10(6), 611–621. Abstract

Healey, J.H. Zimmerman, P.A. et al. (1990). “Percutaneous bone marrow grafting of delayed union and nonunion in cancer patients”. Clin Orthop Relat Res 256, 280–285. Article Abstract

Herreros, J. Prosper, F. et al. (2003). “Autologous intramyocardial injection of cultured skeletal muscle-derived stem cells in patients with non-acute myocardial infarction”. Eur Heart J 24(22), 2012–2020. Article Abstract

Hibi, H. Yamada, Y. et al. (2006). “Distraction osteogenesis assisted by tissue engineering in an irradiated mandible: a case report”. Int J Oral Maxillofac Implants 21(1), 141–147. Abstract

Hibi, H. Yamada, Y. et al. (2006). “Alveolar cleft osteoplasty using tissue-engineered osteogenic material”. Int J Oral Maxillofac Surg 35(6), 551–555. Article Abstract

Hibino, N. McGillicuddy, E. et al. (2010). “Late-term results of tissue-engineered vascular grafts in humans”. J Thorac Cardiovasc Surg 139(2), 431–436, 436 e431–432. Article Abstract

Hibino, N. Shin’oka, T. et al. (2005). “The tissue-engineered vascular graft using bone marrow without culture”. J Thorac Cardiovasc Surg 129(5), 1064–1070. Article Abstract

Horwitz, E.M. Prockop, D.J. et al. (1999). “Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta”. Nat Med 5(3), 309–313. Abstract

Horwitz, E.M. Prockop, D.J. et al. (2001). “Clinical responses to bone marrow transplantation in children with severe osteogenesis imperfecta”. Blood 97(5), 1227–1231. Article Abstract

Hsu, W.M. Carraro, A. et al. (2010). “Liver-assist device with a microfluidics-based vascular bed in an animal model”. Ann Surg 252(2), 351–357. Article Abstract

Jackson, I.T. Scheker, L.R. et al. (1981). “Bone marrow grafting in the secondary closure of alveolar-palatal defects in children”. Br J Plast Surg 34(4), 422–425. Article Abstract

Jiang, Y. Jahagirdar, B.N. et al. (2002). “Pluripotency of mesenchymal stem cells derived from adult marrow”. Nature 418(6893), 41–49. Article Abstract

Jungebluth, P. Alici, E. et al. (2011). “Tracheobronchial transplantation with a stem-cell-seeded bioartificial nanocomposite: a proof-of-concept study”. Lancet 378(9808), 1997–2004. Article Abstract

Kaviani, A. Guleserian, K. et al. (2003). “Fetal tissue engineering from amniotic fluid”. J Am Coll Surg 196(4), 592–597. Article Abstract

Kaviani, A. Perry, T.E. et al. (2001). “The amniotic fluid as a source of cells for fetal tissue engineering”. J Pediatr Surg 36(11), 1662–1665. Article Abstract

Kern, S. Eichler, H. et al. (2006). “Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue”. Stem Cells 24(5), 1294–1301. Article Abstract

Kitoh, H. Kitakoji, T. et al. (2004). “Transplantation of marrow-derived mesenchymal stem cells and platelet-rich plasma during distraction osteogenesis–a preliminary result of three cases”. Bone 35(4), 892–898. Article Abstract

Klein, J.D. Turner, C.G. et al. (2010). “Chest wall repair with engineered fetal bone grafts: an efficacy analysis in an autologous leporine model”. J Pediatr Surg 45(6), 1354–1360. Article Abstract

Krupnick, A.S. Balsara, K.R. et al. (2004). “Fetal liver as a source of autologous progenitor cells for perinatal tissue engineering”. Tissue Eng 10(5-6), 723–735. Article Abstract

Kunisaki, S.M. Armant, M. et al. (2007). “Tissue engineering from human mesenchymal amniocytes: a prelude to clinical trials”. J Pediatr Surg 42(6), 974–979; discussion 979–980. Article Abstract

Kunisaki, S.M. Freedman, D.A. et al. (2006). “Fetal tracheal reconstruction with cartilaginous grafts engineered from mesenchymal amniocytes”. J Pediatr Surg 41(4), 675–682. Article Abstract

Kunisaki, S.M. Fuchs, J.R. et al. (2006). “Diaphragmatic repair through fetal tissue engineering: a comparison between mesenchymal amniocyte- and myoblast-based constructs”. J Pediatr Surg 41(1), 34–39; discussion 34–39. Article Abstract

Kunisaki, S.M. Fuchs, J.R. et al. (2007). “A comparative analysis of cartilage engineered from different perinatal mesenchymal progenitor cells”. Tissue Eng 13(11), 2633–2644. Article Abstract

Kunisaki, S.M. Jennings, R.W. et al. (2006). “Fetal cartilage engineering from amniotic mesenchymal progenitor cells”. Stem Cells Dev 15(2), 245–253. Article Abstract

Lange, P. Fishman, J.M. et al. (2011). “What can regenerative medicine offer for infants with laryngotracheal agenesis?”. Otolaryngol Head Neck Surg 145(4), 544–550. Article Abstract

Langer, R. Vacanti, J.P. (1993). “Tissue engineering”. Science 260(5110), 920–926. Article Abstract

Law, P.K. Goodwin, T.G. et al. (1992). “Feasibility, safety, and efficacy of myoblast transfer therapy on Duchenne muscular dystrophy boys”. Cell Transplant 1(2–3), 235–244. Abstract

Lee, J.Y. Qu-Petersen, Z. et al. (2000). “Clonal isolation of muscle-derived cells capable of enhancing muscle regeneration and bone healing”. J Cell Biol 150(5), 1085–1100. Article Abstract

Lee, M.H. Arcidiacono, J.A. et al. (2010). “Considerations for tissue-engineered and regenerative medicine product development prior to clinical trials in the United States”. Tissue Eng Part B Rev 16(1), 41–54. Article Abstract

Li, H. Liu, H. et al. (2003). “Pluripotent stem cells from the adult mouse inner ear”. Nat Med 9(10), 1293–1299. Article Abstract

Liu, Z. Martin, L.J. (2004). “Pluripotent fates and tissue regenerative potential of adult olfactory bulb neural stem and progenitor cells”. J Neurotrauma 21(10), 1479–1499. Article Abstract

Loai, Y. Yeger, H. et al. (2010). “Bladder tissue engineering: tissue regeneration and neovascularization of HA-VEGF-incorporated bladder acellular constructs in mouse and porcine animal models”. J Biomed Mater Res 94(4), 1205–1215. Article Abstract

Lysaght, M.J. Hazlehurst, A.L. (2004). “Tissue engineering: the end of the beginning”. Tissue Eng 10(1–2), 309–320. Article Abstract

Lysaght, M.J. Reyes, J. (2001). “The growth of tissue engineering”. Tissue Eng 7(5), 485–493. Article Abstract

Macchiarini, P. Jungebluth, P. et al. (2008). “Clinical transplantation of a tissue-engineered airway”. Lancet 372(9655), 2023–2030. Article Abstract

Macchiarini, P. Walles, T. et al. (2004). “First human transplantation of a bioengineered airway tissue”. J Thorac Cardiovasc Surg 128(4), 638–641. Article Abstract

Marcacci, M. Kon, E. et al. (2007). “Stem cells associated with macroporous bioceramics for long bone repair: 6- to 7-year outcome of a pilot clinical study”. Tissue Eng 13(5), 947–955. Article Abstract

McAllister, T.N. Maruszewski, M. et al. (2009). “Effectiveness of haemodialysis access with an autologous tissue-engineered vascular graft: a multicentre cohort study”. Lancet 373(9673), 1440–1446. Article Abstract

Menasche, P. Hagege, A.A. et al. (2003). “Autologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunction”. J Am Coll Cardiol 41(7), 1078–1083. Article Abstract

Mendell, J.R. Kissel, J.T. et al. (1995). “Myoblast transfer in the treatment of Duchenne's muscular dystrophy”. N Engl J Med 333(13), 832–838. Article Abstract

Murry, C.E. Field, L.J. et al. (2005). “Cell-based cardiac repair: reflections at the 10-year point”. Circulation 112(20), 3174–3183. Article Abstract

Naito, Y. Shinoka, T. et al. (2011). “Vascular tissue engineering: towards the next generation vascular grafts”. Adv Drug Deliv Rev 63(4–5), 312–323. Article Abstract

Ohgushi, H. Goldberg, V.M. et al. (1989). “Repair of bone defects with marrow cells and porous ceramic. Experiments in rats”. Acta Orthop Scand 60(3), 334–339. Article Abstract

Olanow, C.W. Goetz, C.G. et al. (2003). “A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson's disease”. Ann Neurol 54(3), 403–414. Article Abstract

Ott, H.C. Clippinger, B. et al. (2010). “Regeneration and orthotopic transplantation of a bioartificial lung”. Nat Med 16(8), 927–933. Article Abstract

Pittenger, M.F. Mackay, A.M. et al. (1999). “Multilineage potential of adult human mesenchymal stem cells”. Science 284(5411), 143–147. Article Abstract

Politis, M. Oertel, W.H. et al. (2011). “Graft-induced dyskinesias in Parkinson's disease: High striatal serotonin/dopamine transporter ratio”. Mov Disord 26(11), 1997–2003. Article Abstract

Prusa, A.R. Hengstschlager, M. (2002). “Amniotic fluid cells and human stem cell research: a new connection”. Med Sci Monit 8(11), RA253–257. Abstract

Quarto, R. Mastrogiacomo, M. et al. (2001). “Repair of large bone defects with the use of autologous bone marrow stromal cells”. N Engl J Med 344(5), 385–386. Article Abstract

Raya-Rivera, A. Esquiliano, D.R. et al. (2011). “Tissue-engineered autologous urethras for patients who need reconstruction: an observational study”. Lancet 377(9772), 1175–1182. Article Abstract

Romanov, Y.A. Svintsitskaya, V.A. et al. (2003). “Searching for alternative sources of postnatal human mesenchymal stem cells: candidate MSC-like cells from umbilical cord”. Stem Cells 21(1), 105–110. Article Abstract

Rossler, R. Boddeke, E. et al. (2010). “Differentiation of non-mesencephalic neural stem cells towards dopaminergic neurons”. Neuroscience 170(2), 417–428. Article Abstract

Rubinstein, P. Carrier, C. et al. (1998). “Outcomes among 562 recipients of placental-blood transplants from unrelated donors”. N Engl J Med 339(22), 1565–1577. Article Abstract

Sakuragawa, N. Enosawa, S. et al. (2000). “Human amniotic epithelial cells are promising transgene carriers for allogeneic cell transplantation into liver”. J Hum Genet 45(3), 171–176. Article Abstract

Salama, R. Weissman, S.L. (1978). “The clinical use of combined xenografts of bone and autologous red marrow. A preliminary report”. J Bone Joint Surg Br 60(1), 111–115. Abstract

Shen, F. Kastrup, C.J. et al. (2008). “Using microfluidics to understand the effect of spatial distribution of tissue factor on blood coagulation”. Thromb Res 122(Suppl 1), S27–30. Article Abstract

Shin’oka, T. Imai, Y. et al. (2001). “Transplantation of a tissue-engineered pulmonary artery”. N Engl J Med 344(7), 532–533. Article Abstract

Shin’oka, T. Matsumura, G. et al. (2005). “Midterm clinical result of tissue-engineered vascular autografts seeded with autologous bone marrow cells”. J Thorac Cardiovasc Surg 129(6), 1330–1338. Article Abstract

Siminiak, T. Kalawski, R. et al. (2004). “Autologous skeletal myoblast transplantation for the treatment of postinfarction myocardial injury: phase I clinical study with 12 months of follow-up”. Am Heart J 148(3), 531–537. Article Abstract

Smith, C. Storms, B. (2000). “Hematopoietic stem cells”. Clin Orthop 379(Suppl), S91–97. Article Abstract

Snyder, E.Y. Deitcher, D.L. et al. (1992). “Multipotent neural cell lines can engraft and participate in development of mouse cerebellum”. Cell 68(1), 33–51. Article Abstract

Steigman, S.A. Ahmed, A. et al. (2009). “Sternal repair with bone grafts engineered from amniotic mesenchymal stem cells”. J Pediatr Surg 44(6), 1120–1126; discussion 1126. Article Abstract

Steigman, S.A. Armant, M. et al. (2008). “Preclinical regulatory validation of a 3-stage amniotic mesenchymal stem cell manufacturing protocol”. J Pediatr Surg 43(6), 1164–1169. Article Abstract

Stosich, M.S. Mao, J.J. (2007). “Adipose tissue engineering from human adult stem cells: clinical implications in plastic and reconstructive surgery”. Plast Reconstr Surg 119(1), 71–83; discussion 84–75. Article Abstract

Takahashi, N. Enosawa, S. et al. (2002). “Transplantation of amniotic epithelial cells into fetal rat liver by in utero manipulation”. Cell Transplant 11(5), 443–449. Abstract

Toma, J.G. Akhavan, M. et al. (2001). “Isolation of multipotent adult stem cells from the dermis of mammalian skin”. Nat Cell Biol 3(9), 778–784. Article Abstract

Torricelli, F. Brizzi, L. et al. (1993). “Identification of hematopoietic progenitor cells in human amniotic fluid before the 12th week of gestation”. Ital J Anat Embryol 98(2), 119–126. Abstract

Tsai, M.S. Hwang, S.M. et al. (2006). “Clonal amniotic fluid-derived stem cells express characteristics of both mesenchymal and neural stem cells”. Biol Reprod 74(3), 545–551. Article Abstract

Turner, C.G. Klein, J.D. et al. (2012). “Craniofacial repair with fetal bone grafts engineered from amniotic mesenchymal stem cells”. J Surg Res. Article Abstract

Turner, C.G. Klein, J.D. et al. (2011). “Preclinical regulatory validation of an engineered diaphragmatic tendon made with amniotic mesenchymal stem cells”. J Pediatr Surg 46(1), 57–61. Article Abstract

Turner, C.G. Klein, J.D. et al. (2012). “The Amniotic Fluid as a Source of Neural Stem Cells in the Setting of Experimental Neural Tube Defects”. Stem Cells Dev. Article Abstract

Urish, K. Kanda, Y. et al. (2005). “Initial failure in myoblast transplantation therapy has led the way toward the isolation of muscle stem cells: potential for tissue regeneration”. Curr Top Dev Biol 68, 263–280. Article Abstract

Vaananen, H.K. (2005). “Mesenchymal stem cells”. Ann Med 37(7), 469–479. Abstract

Vacanti, C.A. Bonassar, L.J. et al. (2001). “Replacement of an avulsed phalanx with tissue-engineered bone”. N Engl J Med 344(20), 1511–1514. Article Abstract

Vacanti, J.P. (2008). “Tissue engineering: from bench to bedside via commercialization”. Surgery 143(2), 181–183. Article Abstract

Vacanti, J.P. Langer, R. (1999). “Tissue engineering: the design and fabrication of living replacement devices for surgical reconstruction and transplantation”. Lancet 354(Suppl 1), SI32–34. Article Abstract

Wagner, W. Wein, F. et al. (2005). “Comparative characteristics of mesenchymal stem cells from human bone marrow, adipose tissue, and umbilical cord blood”. Exp Hematol 33(11), 1402–1416. Article Abstract

Wang, H.S. Hung, S.C. et al. (2004). “Mesenchymal stem cells in the Wharton's jelly of the human umbilical cord”. Stem Cells 22(7), 1330–1337. Article Abstract

Warnke, P.H. Springer, I.N. et al. (2004). “Growth and transplantation of a custom vascularised bone graft in a man”. Lancet 364(9436), 766–770. Article Abstract

Wong Po Foo, C. Patwardhan, S.V. et al. (2006). “Novel nanocomposites from spider silk-silica fusion (chimeric) proteins”. Proc Natl Acad Sci U S A 103(25), 9428–9433. Article Abstract

Young, H.E. Steele, T.A. et al. (2001). “Human reserve pluripotent mesenchymal stem cells are present in the connective tissues of skeletal muscle and dermis derived from fetal, adult, and geriatric donors”. Anat Rec 264(1), 51–62. Article Abstract

Zuk, P.A. Zhu, M. et al. (2001). “Multilineage cells from human adipose tissue: implications for cell-based therapies”. Tissue Eng 7(2), 211–228. Article Abstract

This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

*To whom correspondence should be addressed. E-mail: dario.fauza@childrens.harvard.edu

Last revised August 28, 2012. Published December 10, 2012. This chapter should be cited as: Dionigi, B. and Fauza, D.O., Autologous Approaches to Tissue Engineering (December 10, 2012), StemBook, ed. The Stem Cell Research Community, StemBook, doi/10.3824/stembook.1.90.1, https://www.stembook.org.