8

Roadmap to Organ Printing

Abstract

Tissue engineering has been a promising field of research, offering a means to bridge the gap between organ shortage and transplantation needs; however, construction of three-dimensional (3D)-vascularized whole organs remains the main technological barrier to be overcome. Organ printing, which is defined as computer-aided additive biofabrication of 3D cellular constructs, has advanced this field into a new era. Organ printing takes advantage of bioprinting technology to print cells, biomaterials, and cell-laden biomaterials individually or in tandem, layer by layer, directly creating 3D organ constructs. This chapter presents the roadmap to organ printing technology step-by-step, detailing a set of interrelated processes from isolation of stem cells to posttransplantation monitoring technologies and discusses the current challenges and trends toward fabricating living organs for transplant in the near future.

Keywords

Bioreactors; Organ printing; Organ transplantation; Stem cells; Vascular tissue printing

8.1. Introduction

Despite the increase in donors, organ shortage continues to be problematic. For example, in 2008, more than 28,000 patients received transplants in the United States and 50,463 new patients were added to transplantation wait list 1 year later; nearly half of them received a transplant but quarter of them died while waiting for a suitable organ (Saidi and Hejazii Kenari, 2014). The long-term solution to this problem, as with the solutions to other grand engineering challenges, requires building or manufacturing living organs from an individual’s own cells. For the past three decades, tissue engineering has emerged as a multidisciplinary field involving scientists, engineers, and physicians, for the purpose of creating biological substitutes mimicking native tissue to replace damaged tissues or restore malfunctioning organs (Langer and Vacanti, 1993). The traditional tissue engineering strategy is to seed cells onto scaffolds, which can then proliferate and differentiate, and remodel three-dimensional (3D) functional tissues. Tissues and organs that are anatomically thin or avascular, such as skin, cartilage, bone, bladder, etc., have been successfully engineered (Fisher and Mauck, 2012). Although significant progress has been made in the past decade both in research and clinical applications, it is obvious that complex 3D organs require more precise multicellular structures with vascular network integration, which cannot be accomplished by traditional methods (Ozbolat, 2015).
3D bioprinting processes have emerged to deposit living cells for 3D tissue and organ fabrication using extrusion-based bioprinting (EBB) (Ozbolat and Hospodiuk, 2016), droplet-based bioprinting (DBB) (Gudapati et al., 2016), and laser-based bioprinting (LBB) (Piqué, 2011), as detailed in Chapters 46, respectively. Bioprinting offers great precision for the spatial placement of cells, rather than merely providing scaffold support. Although still in its infancy, this technology appears to be a promising avenue for advancing tissue engineering toward organ fabrication, ultimately mitigating organ shortage and saving lives. Fig. 8.1 demonstrates the concept of futuristic 3D direct organ printing technology, where multiple living cells with supportive media stored in cartridges are printed layer by layer using DBB technology. It offers a controllable fabrication process, which allows precise placement of various biomaterial and/or cell types simultaneously according to the natural arrangement within the target tissue or organs. Multiple cell types, including organ-specific cells from both stroma (i.e., fibroblasts, smooth muscle, and endothelial cells) and parenchyma (i.e., hepatocytes, stellate, Kupffer, and liver sinusoidal endothelial cells for liver) constitute the entire organ. Although the concept seems to be trivial considering the complexity and functionality of the parts that can be manufactured using contemporary 3D printing technology, several challenges impede the evolution of organ printing (Ozbolat and Yu, 2013).
image
Figure 8.1 Organ Printing Concept.
3D functional organs are printed bottom-up using living cells in a supportive medium (Image courtesy of Christopher Barnatt, www.explainingthefuture.com).
Despite the progress in tissue engineering, several issues must be addressed for organ printing to become a reality. The most critical challenge in organ printing is the integration of a vascular network, a problem that faces the majority of tissue engineering technologies. Without vascularization, engineered 3D thick tissue or organs cannot get enough nutrients or efficiently exchange gasses, and remove waste, all of which are needed for tissue maturation during perfusion. This results in low cell viability and malfunction of artificial organs. Systems must be developed to transport nutrients, growth factors, and oxygen to cells while extracting metabolic waste products such as lactic acid, carbon dioxide, and hydrogen ions so the cells can grow and organize to form the organ. Cells in a large 3D organ structure cannot maintain function without a transport system traditionally provided by blood vessels. Blood vessels are an intraorgan branched vascular tree that is a part of the circulatory system in the human body. Fluid and media transport as well as oxygenation takes place at the capillary level. Bioprinting technology, on the other hand, currently does not allow organ fabrication where bifurcated vessels are manufactured with capillaries to mimic natural vascular anatomy. Although several researchers have investigated the development of vascular trees using computer models (Mondy et al., 2009), only a few attempts have been made toward fabricating bifurcated or branched channels so far (Norotte et al., 2009). Successful tissue maturation with a functional mechanically integrated bifurcated blood vessel network is still not a reality.

8.2. State-of-the-Art in Organ Printing

Bioprinting of organs of the clinically relevant dimensions has yet to be performed (Ozbolat, 2015). Up to now, there are two major strategies followed for organ printing including scaffold-free and scaffold-based approaches.
Using scaffold-free approach, Forgacs and his coworkers at the University of Missouri, Columbia, bioprinted heterocellular tissue spheroids, producing scaffold-free vascular constructs (Norotte et al., 2009). Upon fusion of tissue spheroids followed by a tissue maturation process of 3 days postbioprinting, the support material was removed manually to generate the lumen. Multiple cell types, including human umbilical vein smooth muscle cells and human skin fibroblast cells, were printed together to fabricate multicellular constructs. The same group also demonstrated bioprinting of cell pellets, instead of tissue spheroids, for fabrication of blood vessels (see Fig. 8.2A1-A3) and nerve conduits using a similar approach, where the pellet was deposited between strands of agarose, which were inert to cell adhesion facilitating aggregation of cells in 3D (Norotte et al., 2009; Owens et al., 2013). The bioprinting platform used in this study, called Novogen MMX Bioprinter™, has been specialized for bioprinting a broad array of cell types to create functional 3D tissues that can recapitulate in vivo biology for human disease research, drug discovery and development, and toxicology testing. One of the major challenges of this technology is the use of a mold, which restricts the geometry of the tissues into thin tubular shapes. In this regard, the author’s group recently used tissue strands as building blocks for larger-scale cartilage patches (Yu et al., 2016). Using 8-cm-long cartilage tissue strands, cartilage patches that were histologically close to native bovine cartilage, were 3D bioprinted without the need for a mold thus allowing scale-up bioprinting (see Fig. 8.2B1–B4). As cartilage is avascular, the use of tissue strands produced large cartilage patches that can be used for human joint repair in the future. Despite these efforts, further research and development is needed to scale-up the constructs to clinically relevant volumes. When building large-scale organ constructs, mechanical integrity as well as the integration of the vascular network with the rest of the organ seems to be the major obstacle to expanding the technology for further clinical applications.
image
Figure 8.2 Examples of Organ Printing.
(A1) Step-by-step demonstration of scaffold-free bioprinting of cell pellet along with agarose support, (A2) where the bioprinted construct facilitated aggregation of pig smooth muscle cells in 3 days (A3) followed by removal of the agarose support (Reproduced/adapted with permission from Norotte et al. (2009)). (B1) Multilayer bioprinting of free-standing cartilage tissue strands facilitated complete fusion of strands in a week followed by (B2–B3) implantation on a bovine osteochondral explant with a 4  mm × 4 mm defect. (B4) Further cultivation of the bioprinted patch resulted in cartilage that was histologically close to the native bovine cartilage (Reproduced/adapted from Yu et al. (2016)). (C1–C2) A computer-aided design (CAD) model of a human mandible bone generated for a defect captured using CT images. (C3) Toolpath plan was generated for the cell-laden bioink, polycaprolactone (PCL), and fugitive Pluronic F-127, and (C4) 3D printing was performed accordingly. (C5) Alizarin Red staining demonstrated osteogenic differentiation of human amniotic fluid derived stem cells (hAFSCs) in a long-term cultured mandible bone construct (Reproduced/adapted with permission from Kang et al. (2016)).
Recently, scaffold-based bioprinting approach has been utilized to generate larger-scale tissue constructs (Kang et al., 2016). Using polycaprolactone (PCL) as a 3D-printed thermoplastic frame along with Pluronic as a fugitive ink, mechanically strong and stable cell-laden tissue constructs were printed with a porous architecture prior to implantation. The concept was successfully tested for various tissue types such as cartilage, bone, and muscle on murine models. A similar concept was previously attempted (Pati et al., 2014), where decellularized matrix components of various tissue types, such as adipose and cartilage, were bioprinted along with PCL fibers to fabricate larger-scale tissue constructs. Although larger-scale constructs were 3D printed with better structural and mechanical integrity, use of PCL in tissue construct remains a major drawback due to its slow degradation rate that may interfere with soft tissue regeneration.

8.3. Roadmap to Organ Printing

Organ printing is a computer-aided process in which cells and/or cell-laden biomaterials are placed according to a blueprint model that serves as building blocks that are further assembled into 3D constructs and matured toward functional organ formation. It is an automated approach that offers a pathway for scalable, reproducible mass production of engineered living organs, where multiple cell types can be positioned precisely to mimic their natural counterparts. Developing a functional organ requires advances in integration of three types of technology (Ozbolat and Yu, 2013; Lanza et al., 2007): (1) cell technology, which addresses the procurement of functional cells at the level needed for clinical applications, (2) biofabrication technology, which involves combining the cells with biomaterials in a functional 3D configuration, and (3) technologies for in vivo integration, which addresses the issue of bioprinted organ immune acceptance, in vivo safety and efficacy, and monitoring of organ integrity and function postimplantation. To successfully realize organ printing in practice at the clinical level, robust automated protocols and procedures should be established. Fig. 8.3 illustrates the roadmap to organ printing, which is composed of three major steps including (1) preorgan printing stage, (2) organ printing stage, and (3) postorgan printing stage.
In preorgan printing stage, the required raw materials consisting of patient-specific cells, nonimmunological biomaterials, growth factors, and cytokines for the organ printing process are prepared. Patient-specific cells can be obtained from stem cells such as bone marrow stem cells. Ideally, stem cells should be harvested with minimum invasion; for example, skin fibroblasts can be reprogrammed into induced pluripotent stem cells (iPSCs). Once stem cells are isolated, the next step is to differentiate stem cells into organ-specific cells that are essential to reconstruct the highly heterocellular nature of complex organs. After expansion of cells in sufficient numbers for the scale-up organ printing mission, cells can be cultured in two-dimensional (2D) or 3D depending on the organ to be printed. For example, in the case of the pancreas, islets of Langerhans need to be fabricated in the form of spheroids (Fennema et al., 2013; Bernard et al., 2012). Next, the bioink material, with the cell density comparable to the cell density in natural organs, is prepared using one of the approaches presented in Chapter 3.
image
Figure 8.3 Roadmap to organ printing process (Image courtesy of Elsevier for stem cell isolation image Lanza et al. (2007); Christopher Barnatt from www.explainingthefuture.com for bioprinter image; Elsevier for organ transplant image).
In organ printing stage, anatomically correct models of organs should be acquired for the target organ. Currently, there exist several noninvasive medical imaging techniques, as discussed in Section 2.3, to capture the 3D geometry of human organs. Then, the CAD blueprint model can be prepared using one of the appropriate blueprint modeling techniques, discussed in Section 2.4, and imported to machine control software (see Section 7.2.1) to run the bioprinter. At this step, the bioprinter needs two major pieces of information including (1) what to deposit, and (2) where and when to print. During the organ printing process, both the parenchymal and stromal part of the organ should be printed in tandem along with any support or temporary structural elements needed.
After the organ printing process, the bioprinted construct is highly fragile and not structurally coherent or integrated at a sufficient level to facilitate transplantation. Therefore the postorgan printing stage is critical to obtain functional, mechanically stable, and innervated organs for transplantation. The cultivation period necessitates proper bioreactor technologies to enable mechanical and chemical stimulation and signaling to regulate organ remodeling and growth. Upon sufficient maturation and testing of organs, the organs can be transplanted to the patient, and the functionality and in vivo safety parameters monitored for a significant period of time. The following section discusses in detail each of the three major stages for the roadmap to organ printing.

8.3.1. Isolation and Differentiation of Stem Cells

Successful organ printing relies heavily upon advancements in stem cell technology as native tissues and organs are heterocellular. Acquisition of functional primary cells from patients with visceral dysfunction such as liver or heart failure or other devastating diseases such as type I diabetes as autologous beta (β) cells is not possible. Stem cells, which are found in several tissues in human body, can self-renew to produce more stem cells and have the remarkable potential to differentiate into diverse specialized cell types to form various organs. A variety of cell types can be used for this application, such as embryonic stem (ES) cells (Thomson et al., 1998), adult stem cells (ASCs) (Baglioni et al., 2009), iPSCs (Takahashi et al., 2007), human adipocyte stem cells (ADSCs) (Gimble et al., 2007), and tissue-specific cell lines (Pan et al., 2009). Although ES cells are pluripotent, their availability has been hampered by controversial ethical concerns due to their derivation from early embryos (Wert, 2003). Induced pluripotent stem cells have eliminated those issues, as somatic cells from patients can be easily isolated with minimally invasive procedures such as skin biopsy. The obtained skin fibroblasts can be reprogrammed into adult cells from all three germ layers in vitro, which have a great potential in regenerative medicine for cell therapy applications or tissue or organ fabrication. Reprogramming of somatic cells to obtain iPSCs, depending on the method used, may pose substantial risk and thus limit their clinical translation. Human adipocyte stem cells also eliminate the ethical concerns as abundant amount of adipose tissue can be easily isolated from subcutaneous adipose deposits with safe, minimal invasive procedures in an outpatient setting. Additionally, adipocyte stem cells retain pluripotency in vitro (Bunnell et al., 2008). Thus these stem cell types provide a wide range of cell sources for organ printing; however, there are still some impediments with their use. Although autologous stem cells can be differentiated into organ-specific cells for organ printing, there is still a risk of tissue rejection by the recipient (Lanza et al., 2007). Phenotypic behavior of stem cells can even change during the bioprinting process. In addition, organ fabrication requires various types of organ-specific cells, which is not currently feasible with current stem cell isolation and differentiation technologies. Although stem cells offer great promise as an unlimited source of cells for organ printing, a greater understanding of and control over the differentiation process is required to generate expandable organ-specific cells of consistent quality with the desired phenotype and genotype thereby minimizing organ rejection by the recipient posttransplantation. In addition, immunobarrier devices or molecular level interventions in the form of DNA modifications are essential to overcome immunologic rejection of organs. Moreover, imaging modalities such as positron emission tomography and nuclear magnetic resonance (NMR) imaging should be used to monitor the functionality of seeded stem cells noninvasively; NMR offers a unique advantage in monitoring organ integrity and cell function without the need to modify the cells genetically (Stabler et al., 2005).

8.3.2. Cell Expansion

Although current stem cell technologies can expand stem cells into adequate numbers for laboratory experiments, future organ printing technologies will require scale-up manufacturing of stem cells for larger-scale human tissues for transplantation and generation of tissue samples for pharmaceutical use. To attain sufficient numbers of cells for human organ printing, advanced bioprocessing technologies should be implemented to enable practical expansion of stem cells in compliance with good manufacturing practices with appropriate quality assurance measures, bioprocess monitoring control, and automation (Placzek et al., 2009). Each stem cell type has a different expansion potential. Embryonic stem cells have unlimited expansion capacity; however, other stem cells do not. For example, mesenchymal stem cells (MSCs) from young donors have 40 population doubling; whereas MSCs from older donors are limited to 25 population doubling (Stenderup et al., 2003). As previously discussed, the use of ES cells have other issues such as ethical considerations while MSCs have other limitations such as the need for an invasive procedure to obtain a limited sample volume as they are primarily isolated from bone marrow. Therefore other stem cells such as iPSCs and ADSCs stand as promising cell sources that can be obtained in large quantities from abundant tissue volumes isolated using minimally invasive procedures. After isolation, stem cells should be plated and expanded in appropriate culture conditions. In general, expansion culture environments are maintained at 5% carbon dioxide (CO2), 20% oxygen, 37°C, and pH 7.4; however, growth and differentiation of different stem cells may vary under different conditions. For example, it has been demonstrated that a lower pH level of 7.1 increased the expansion of megakaryocyte progenitor cells (Yang et al., 2002). Large volumes of cells need to be expanded in a bioreactor culture with appropriate nutrient and waste exchange, accommodations for high cell density, and a continuous monitoring and feedback mechanism. For organ printing, two different strategies can be used in loading stem cells including (1) bioprinting differentiated stem cells during the organ printing process or (2) bioprinting predifferentiated stem cells followed by differentiation into organ-specific cell lineages within appropriate sectors of the printed organ constructs. The former approach is preferred over the latter one, in general, as differentiation of multiple cell types into different lineages is extremely challenging requiring spatiotemporal delivery of growth factors or plasmid DNA (Ozbolat and Hospodiuk, 2016).

8.3.3. Bioink Preparation

Upon expansion to sufficient numbers, cells are processed for bioink preparation. Depending on the bioink type utilized, such as hydrogels, cell aggregates, microcarriers, or decellularized matrix components as discussed in Chapter 3, different cell quantities are required. Quality assessment of the cells is essential to ensure that their purity, phenotype, genotype, and functionality are acceptable.
As hydrogels are primarily used in tissue construct biofabrication, appropriate hydrogel bioink materials should be selected based on the target organ type as well as the utilized bioprinting modality. Hydrogel-based bioink materials can be processed while the cells are in culture for expansion as some hydrogels require prolonged preparation times. For example, if collagen type I is used as a bioink component, it should be screened for the presence of any contaminants and immunoreactive components; its protein profile and molecular structure should match that of standard collagen type I. Such screening should be performed for each component of the bioink solution. As composite bioink materials may have several components, the sequence of adding each one should be based on well-established protocols as slight variations at the preparation stages may result in substantial differences in the performance of the bioink solution. Then, cells need to be added and homogenously mixed into the bioink solution at a sufficient density to facilitate tissue formation. Although different tissue types require different cell densities, higher cell densities enable better cell–cell interactions and induce successful tissue formation. As the hydrogel-based bioink solutions, in their precursor form, possess a nonporous liquid microenvironment for cells, cells should be added at the latest stage of the bioink preparation. If possible, some media should be added to the bioink solution to minimize any cellular necrosis. In addition, precursor forms of the bioink solution components can also be toxic to cells; therefore it is ideal to bioprint and crosslink the precursor solution in shortest possible period of time.
For other bioink types, such as cell aggregate–based bioink materials, different preparation procedures are required. The cell pellet is relatively easy to process; however, sufficient cohesiveness should be achieved by retaining the pellet for a period of time (15 min–1 h) in glass capillaries (Owens et al., 2013). Extending this time can adversely affect the cell viability due to oxygen insufficiency. In addition, high-precision instruments should be used to economize on bioink material during the process. For tissue spheroid preparation, various methods [i.e., mold culture, microfluidics-based or magnetic assembly techniques (see Chapter 3)] can be used for high-throughput fabrication. Tissue spheroids should be harvested after sufficient cohesiveness and mechanical properties are attained. If neovascularization within spheroids is crucial, such as in the case of pancreatic islets or tumor models, then cocultured spheroids should be harvested after a single day in culture as further culture reduces angiogenic potential. For tissue strands, a minimum culture period should be provided to facilitate sufficient cell aggregation (Yu et al., 2016). As there is no hydrogel or other exogenous materials, cells need to be cultured in specialized reagents, such as serum and growth factors, that are conducive to maintaining cell viability. As these reagents are expensive, future organ bioprinting technologies will need to devise cost-effective scale-up production strategies to increase the availability of humanized reagents with a wide range of commercial availability.
After preparation of the bioink materials, they are loaded into the bioprinter. As native tissues and organs are heterocellular in their organization, careful loading of multiple bioink solutions is essential and adequate calibration should be performed before running the bioprinter. For extended bioprinting processes, additional bioink materials may be needed therefore manual/automatic unloading and loading needs to be performed to complete the entire construct.

8.3.4. Blueprint Modeling

To bioprint an organ construct, a blueprint model is essential to guide the bioprinting process. Organ bioprinting processes are complex compared to the bioprinting of cells within bulk- or lattice-shaped hydrogel scaffolds. These endeavors include scaffold-free bioprinting (Yu et al., 2016; Norotte et al., 2009), integration of scaffold-free approach with vascular network (Yu et al., 2014), a scaffold-based approach with integrated vascular network (Zhang et al., 2013a), and the use of a hard polymeric frame to support cell-laden hydrogel sections (Kang et al., 2016). All these approaches are highly complicated in their construction, where multiple materials need to be bioprinted, hence requiring a complex blueprint model. Such complex blueprint models can be developed using one or more of available techniques such as CAD-based systems, image-based design, freeform design, implicit surfaces, and space filling curves as discussed in Section 2.3. For scaffold-free approaches, particularly, where the bioprinting process guide the tissue self-assembly, tissue remodeling, fusion, and contraction are commonly observed, resulting in significant deviations from the originally bioprinted construct. Therefore the blueprint model and the associated toolpath plan should compensate for the postbioprinting changes.

8.3.5. Process Planning

Upon generation of the blueprint model for the target organ, a process plan should be performed to determine how to bioprint the organ construct including its compartments and components, such as stromal, parenchyma, blood vessels, and support material. An appropriate toolpath plan is thus essential to provide the bioprinter with information about the robot motion and the deposition patterns so that exact deposition of different bioink constituents can be determined. This information is then transferred from toolpath planning software to the machine control software via digital signals that control the motion and dispensing mechanisms, such as actuation of motors and air pressure, respectively (Ozbolat et al., 2014). For toolpath planning, both Cartesian- and parametric-based approaches, as discussed in Section 2.5, can be considered. As organ printing may necessitate bioprinting multiple bioink materials or even the support material, Cartesian-based toolpath planning is preferred due to its simplicity.

8.3.6. Bioprinting of Vascularized Organs

At this step of organ fabrication, organ constructs can be 3D bioprinted using any single or combinations of existing bioprinting modalities including EBB, DBB, and LBB as presented in Chapters 46, respectively. As EBB modality facilitates fabrication of 3D constructs at the clinically relevant volumes, it is currently preferred over other methods, but can also be integrated with others if required. To print organs at clinically relevant volumes, robust technologies and protocols should be developed to enable bioprinting of vascular constructs in multiple-scale ranges from arteries and veins down to capillaries. Since it is difficult to print capillaries at submicron scale using the current bioprinting technology, one alternative strategy can be bioprinting the macrovasculature with the expectation that the interconnecting capillaries will be formed on their own (Yu et al., 2014). In this regard, two alternative approaches have been considered in the literature including indirect bioprinting utilizing a fugitive ink that is removed by thermally induced decrosslinking leaving a vascular network behind (Kolesky et al., 2014; Lee et al., 2014a,b) and direct bioprinting of vasculature network in tandem with the rest of the tissue constructs (Yu et al., 2014).

8.3.6.1. Indirect Bioprinting of Vascular Network

In the last few years, several researchers have attempted bioprinting with a fugitive bioink to create vascular channels including Bertassoni and his coworkers (see Fig. 8.4A1–A2) (Bertassoni et al., 2014), Chen’s group (see Fig. 8.4B1-B2) (Miller et al., 2012), and Lewis and her coworkers (see Fig. 8.4C) (Kolesky et al., 2014). In these studies, cell-laden hydrogels were used as the base material to fabricate the tissue construct where a vascular network was created by 3D printing a sacrificial material, such as Pluronic F-127, agarose, and gelatin, followed by its removal after complete gelation of the hydrogel. Such a construct displayed sufficient structural integrity. The integrated vascular network resulted in increased cell viability inside the construct; regions near the channels exhibited significant differences in cell viability compared to regions away from the channels.
The majority of researchers have attempted to create vascular networks in macroscale by generating an endothelial lining inside the lumen via colonization of endothelial cells through perfusion. Dai et al. took one step forward and have successfully achieved angiogenesis by sprouting endothelial cells within a fibrin network loaded with other support cells (see Fig. 8.4D1–D2) (Lee et al., 2014a,b). Their study demonstrated that creating a vascular channel with the luminal surface covered with endothelial cells improved the diffusion of plasma protein and dextran molecules. Similar angiogenesis models have already been developed in lab-on-a-chip models, where several types of support cells have been used in cancer metastasis studies lead by Kamm’s and George’s groups (Chung et al., 2009; Sheng et al., 2014). Despite the great flexibility in bioprinting channels and the ability to initiate angiogenesis, this technology still faces with several challenges. First of all, loading cells in hydrogels does not support cell–cell interactions, and limited phenotypic stability and activity of cells are observed during in vitro incubation. Fibrin demonstrated a superior environment for angiogenesis while fibrin plays a crucial role in blood clotting (Janmey et al., 2009); however, fibrin is not a hospitable environment for all support cell types (cells that are considered tissue-specific cell types) and does not preserve the construct integrity for long periods of time. With further advancements in these technologies, vascularization that provides an efficient media exchange system for thick tissue fabrication will be a reality in the near future.

8.3.6.2. Direct Bioprinting of Vascular Network

In addition to efforts using temporary sacrificial materials to generate channels, vascular network bioprinting has been demonstrated using various direct bioprinting approaches. For example, scaffold-free bioprinting of vascular networks has been performed using tissue spheroids as building blocks as shown in Fig. 8.5A1–A3 (Norotte et al., 2009). Six days after deposition, tissue spheroids made of human skin fibroblasts (HSFs) completely fused and maturated into a vascular tissue with branches demonstrating the ability of spheroids to self-assemble. In addition to the scaffold-free approach, scaffold-based approaches have been extensively studied by the author’s research group through the use of a coaxial nozzle apparatus (see Fig. 8.5B1–B2), which allows direct bioprinting of the vasculature with immediate crosslinking of sodium alginate bioink generating a smooth and continuous lumen of any desired length (Zhang et al., 2015). The anatomy can be determined by controlling the bioprinting parameters and the shape of the vascular network can be mediated by bioprinting where the vasculature can be loaded with cells such as fibroblast and smooth muscle cells and cultured in perfusion chamber prolonged times (see Fig. 8.5B3–B4). Complex patterns were bioprinted and the vascular network was easily integrated with larger-scale bulk hydrogels with cell viability over 95% retained over a week culture (see Fig. 8.5B5-B7) (Zhang et al., 2013a). The coaxial nozzle bioprinting was further used to demonstrate the embedding of vascular channels in hydrogel constructs increasing the viability of cells compared to cells in bulk hydrogels (Gao et al., 2015).
image
Figure 8.4 Indirect Bioprinting of Vascular Channels Using Fugitive Ink.
(A1) A photograph of bioprinted agarose hydrogel filaments as fugitive ink representing branched vascular network in a gelatin methacryloyl (GelMA) hydrogel block and (A2) a high resolution cross-sectional view of GelMA block stained for live and dead cells (Reproduced/adapted with permission from Bertassoni et al. (2014)); 3D printing carbohydrate glass as a fugitive ink leaving vascular channels in agarose gel loaded with primary rat hepatocytes and stromal fibroblast were stained with a fluorescent live/dead assay (green, Calcein AM; red, Ethidium Homodimer) (B1) showing a considerable percentage of death cells in slab gels without channels compared to (B2) high viability of cells in channeled gels, particularly around the perfused channels (Reproduced/adapted with permission from Miller et al. (2012)); (C) an image acquired during evacuation of the fugitive ink showing channels in GelMA scaffold (upper-left), which were later glued with 10T½ fibroblasts, human umbilical vein endothelial cells (HUVECs), and human dermal fibroblasts (Reproduced/adapted with permission from Kolesky et al. (2014)); and (D1) sprouting of endothelium (stained with red fluorescent protein) into capillary network (stained with green fluorescent protein) within fibrin gel on day 9 and (D2) a high resolution image of the capillary network on day 14. (Reproduced/adapted with permission from Lee et al. (2014a,b)).
image
Figure 8.5 Direct Bioprinting of Vascular Network.
(A1) Scaffold-free bioprinting of a branched vascular network using 300 μm human skin fibroblast spheroids (solid and broken arrows show 1.2 and 0.9 mm in vascular diameter, respectively), where spheroids (A2) fuse and maturate into tissue after 6 days of deposition (A3) (Reproduced/adapted with permission from Norotte et al. (2009)); (B1–B2) Coaxial extrusion setup for bioprinting vascular network, where the crosslinker solution flowing through the core facilitates rapid gelation of alginate vascular tubes, (B3–B4) which are perfusable over prolonged pulsatile flow. (B5) Complex patterns could be easily generated (Reproduced from Zhang et al. (2015)) and (B6) vascular network embedded within bulk hydrogel solutions (Reproduced with permission from Zhang et al. (2013a)). (B7) Cells kept their high viability over time. The similar concept was further enhanced to (C1–C2) bioprinted vascular channels embedded in a large alginate construct (C3) showing L929 mouse fibroblasts in green. (Reproduced/adapted with permission from Gao et al. (2015)).

8.3.6.3. Integration of Vascular Network With Parenchymal Tissue

The ultimate goal of tissue engineering techniques is the generation of a functional tissue or organ with an integrated vascular network. To successfully create multiscale vascularization within organ constructs, the constructs need be placed in a custom-made perfusion bioreactor, where vascular pedicles are connected to facilitate continuous medium flow in vitro. The perfusion bioreactor not only ensures sufficient structural support for the printed organ construct, but also provides an environment similar to in vivo conditions. Essential growth factors such as vascular endothelial growth factor, fibroblast growth factor, and epidermal growth factor (EGF) are supplemented within the circulating culture media. The printed constructs mature over time, creating an organ at the clinically relevant size enclosing the vasculature during in vitro incubation. More importantly, upon the formation of the 3D organ constructs, the previously mentioned angiogenesis growth factors continue to be supplied during in vitro culture to drive the natural process of vascularization between the main vasculature and prevascularized constructs. Ultimately, thick 3D constructs can be fabricated with a biomimetic vasculature system and be readily available for transplantation, disease modeling, or drug screening. This will be a major breakthrough toward fabrication of larger-scale organs.
Studies have demonstrated perfusable blood vessel network embedded in thick tissues using bioprinting and traditional fabrication approaches. For example, a recent study (Yu et al., 2014) demonstrated hybrid bioprinting of vasculature in tandem with tissue strands using a Multi-Arm BioPrinter (Ozbolat et al., 2014), where fibroblast tissue strands quickly fuse to each other, mature and form the tissue around the vasculature. Tissue strands were comprised only of cells and their extracellular matrix (ECM). They were used as building blocks to construct the scale-up organ due to their quick fusion, folding, and maturation capabilities. Using traditional fabrication approaches, scientists integrated vascularization with 3D cell sheet fabrication technology, where endothelial cells within a cardiac cell sheet sprouted and connected to the main blood vessel upon perfusion of growth factor–rich culture media (Sekine et al., 2013). Another study demonstrated that a prevascularized hepatic bud, when transplanted in vivo, can successfully anastomose to the main blood vessel and survive for a long period of time, performing its original function (Takebe et al., 2014). All of these findings offer the potential for organ printing to have a similar nature-driven effect upon perfusion. When the media is perfused through a continuous vascular network within the vascularized tissue constructs, the biological signals as well as the media gradient along the perfusion direction would guide endothelial cell reorganization, migration, and facilitate angiogenesis within the organ and orchestrate capillary network formation toward the media supply.
Newly generated capillaries within the organ constructs are expected to anastomose to the main vasculature, so that media supplied through these newly formed capillaries would guarantee the survival of the organ constructs for longer periods of time. Continuous media circulation within the newly generated constructs would also accelerate the tissue maturation process by supplying sufficient growth factors, which drive tissue-specific cells to secrete ECM and further facilitate tissue or organ maturation, producing a functional vascularized perfusable organ. Later, the matured organ could be used for drug testing by directly delivering different drugs via the perfusion system to evaluate the organ response. Moreover, 3D-printed organ constructs could be implanted in vivo by suturing the main vasculature to the host to replace damaged or diseased tissues or organs.

8.3.7. Advanced Bioreactor Technologies for Organ Culture

After the organ bioprinting process, as discussed previously, the fabricated construct needs to be transferred to a bioreactor for long-term culture to facilitate cell growth and proliferation, vascularization, and organ maturation. A bioreactor can be defined as a system in which the conditions are closely controlled with respect to physiological conditions to induce certain behavior in living cells, tissues, and organs (Korossis et al., 2005). Three major bioreactor types have been used in culturing engineered tissue constructs including static and mixed flasks, rotating wall, and perfusion bioreactors (Gaspar et al., 2012). Mixed flask culture or rotating wall utilizes a convection mass transfer mechanism; however, perfusion bioreactors enable both convection and diffusion-mediated mass transfer. Complex-organ systems such as decellularization-based approaches or bioprinted vascularized organ constructs require perfusion culture that forces culture medium through the vascular inlets and collects it from outlets using a suction mechanism. The ideal bioreactor should provide the appropriate physical stimulation to cells, a continuous supply of nutrients (e.g., glucose, amino acids) and removal of by-products of cellular metabolism. The bioreactor should also facilitate sufficient convection and diffusion of biochemical factors and oxygen, as well as provide mechanical stimuli to induce mechanotransduction and the appropriate deposition of ECM proteins with controlled orientation and structure. In addition, such a bioreactor should maintain aseptic conditions, allow for a high media volume to tissue construct volume ratio, fail to generate any toxic products, and be easy to clean after the culture period. In general, different organ and tissue types, possessing different anatomical, biological, mechanical, and structural characteristics, may need specialized bioreactor designs; however, the three major requirements for bioreactor design include (1) mass transport, (2) mechanical, and (3) electrical stimulation
Mass transport through a bioreactor system is by far the most essential requirement for successful cultivation of printed organs. As organ constructs possess micro- or macrosize pore, mass transport of oxygen, nutrients, and regulatory molecules is highly limited under static conditions. In addition, metabolites, CO2, and lactic acid need to be removed from the organ constructs. As solubility of oxygen within media is low, exposure of cells to oxygen in organ constructs is reduced. Therefore the bioreactor system should support circulation of the fresh media throughout the entire organ construct and facilitate efficient convection.
Mechanical stimulation of organ constructs, via mechanical compression or stretch, hydrodynamic pressure, and fluid flow (Salehi-Nik et al., 2013), is a crucial factor for successful maturation of bioprinted organ constructs (see Fig. 8.6). Mechanical stimulation can improve adhesion and stretching of cells, mediates the deposition direction of ECM proteins, triggers the secretion of biological factors, and determines whether cells aggregate or detach from the organ constructs. For example, adhesion strength and orientation of endothelial cells are highly dependent on shear stress, where cells orient and elongate in the direction of the flow and facilitates the formation of tight intercellular junctions, and the release of more nitric oxide (Li et al., 2015). On the other hand, chondrocytes grown in an organ construct under cyclic compression test demonstrated better mechanical properties and deposition of ECM proteins such as proteoglycans (Kisiday et al., 2004). Cell aggregates are more sensitive to mechanical stimuli compared to cells alone due to their large volume (Henzler, 2000); therefore the likelihood of detachment of cell aggregates from tissue constructs is higher than that of individual cells.
Electrical stimulation is another important factor to enhance the functionality of particular tissue constructs such as cardiac or muscle tissue or the innervation of composite tissues with nerves. For example, bioprinted cardiomyocytes in alginate constructs demonstrated better contractile properties under electrical stimulation (Xu et al., 2009). In addition, electrical stimuli can be integrated with mechanical stimuli to initiate appropriate functionalities. For example, engineered muscle tissues demonstrated higher contractile properties along with better elongation of muscle cells along the tissue construct when the tissue is exposed to optimal electromechanical stimuli (Liao et al., 2008).
image
Figure 8.6 (A) A perfusion bioreactor (B) for cultivation of bioprinted blood vessels for further tissue maturation, and deposition of collagen and elastin proteins (Reproduced/adapted with permission from Norotte et al. (2009)).
In addition to its stimulatory function, the ideal bioreactor should also possess the capability of controlling the tissue quality of each and every printed organ over time. In this regard, noninvasive and nondestructive approaches should be developed to monitor the quality of organs or the metabolic states of cells, such as measurement of glucose uptake or oxygen consumption using embedded sensors. In addition, mechanical quality of load-bearing organs such as bone and cartilage is also important; bioreactors should advance the mechanical stimulation over time to increase the mechanical properties of the developing tissue. Advanced monitoring capabilities should be integrated to read the mechanical parameters from the construct and direct adjustment of the stimulation automatically. All these monitoring capabilities will increase the efficiency of postbioprinting organ culture processes and increase the quality and replicability of organs within predetermined biofabrication specifications (Korossis et al., 2005). This will be crucial for printed organs used in pharmaceutical and drug testing.
In addition to bioreactor considerations, postbioprinting of organs is another important step toward successful transplantation. The transfer of a printed organ to the bioreactor should be performed with extreme care to prevent degradation in quality and function of the organ. Thus future bioprinters can be enclosed in a bioreactor system that will allow direct and rapid connection of printed structures to perfusion channels.

8.3.8. Organ Remodeling and Maturation

During the bioreactor culture, bioprinted organ constructs exhibit different biological and morphological changes over time depending on the bioink and bioprinting strategy utilized in the organ printing process. For example, organ constructs made using a scaffold-free approach undergo a different process than cells suspended and bioprinted in hydrogels.
Different cell aggregate–based bioink materials experience a different series of events during organogenesis (Ozbolat and Hospodiuk, 2016). Cells in pellet form, when bioprinted and confined into a mold, start to adhere to each other to minimize free energy and facilitate cell–cell interactions and connections through cadherin-mediated adhesion (Akkouch et al., 2015). Over time, cells deposit their own ECM, promoting cell adhesion and generating contractile forces resulting in formation of intact neotissues that are smaller than their original size. Cells continue to deposit parenchymal ECM components such as elastin and collagen, where the cohesion and mechanical properties of the tissue increases and the tissue matures and eventually attains a morphology and physiology close to the native tissues. When sufficient structural cohesion is achieved, the mold structure can be removed from the construct to allow better perfusion and diffusion. If other cell aggregate–based bioink materials are used such as tissue spheroids (Mironov et al., 2016) and strands (Yu et al., 2016), tissue fusion starts immediately through cross-migration of cells and deposition of ECM components into the space in aggregates. To minimize the configurational energy during fusion, fused aggregates assume a more rounded geometry followed by deposition of ECM components and maturation of the tissue toward a nativelike morphology.
Cells bioprinted in hydrogel matrix, on the other hand, are exposed to a different environment and exhibit different chain of events during organogenesis. As cells loaded in hydrogels, they first attach to the scaffold matrix, and proliferate and deposit their own ECM. Meanwhile, they express proteinases including matrix metalloproteinases (MMPs), which causes the degradation of the bioink material. For example, MMP-1 and MMP-13 cleave collagen type I in skin and bone tissue, respectively (Nagase et al., 2006). As cells grow and increase in number, and the matrix around them starts to degrade, slow changes are observed in the morphology and physiology of the organ construct.
In addition to these events, neovascularization is another important factor, where endothelial cells form into tubular organization followed by incorporation of pericyte-like supporting cells, such as human normal lung fibroblasts and smooth muscle cells, surrounding the endothelial structures, stabilizing their growth, and improving the mechanical integrity of developing capillaries. Later, these tubes anastomose to each other forming a larger vascular network that can be connected to larger-scale vascular network (Ozbolat, 2015). Depending on the target organ type, its morphology, structural and mechanical properties, function and physiology, different types of strategies, and bioink materials can be utilized.

8.3.9. Transplantation, Immunosurveillance, and In Vivo Safety, Efficacy, and Monitoring of Organs

Bioprinted organs, depending on their type, may pose difficulties associated with transplantation such as cellular ischemia requiring the printed organ to be transported on ice, the need for various flush solutions to prevent cellular edema, delay cell destruction, and maximize functions of organs after perfusion is reestablished. Once a bioprinted organ arrives in the operating room, it needs to be implanted in the appropriate site and attached to an uninjured intact vascular pedicle so that the organ can be perfused. Larger organs require larger vascular pedicles for immediate parenchymal perfusion; however, as the bioprinted organs need to be fully engrafted with the host, angiogenic integration with the recipient microcirculation is also essential. Since it is challenging to bioprint capillaries within bioprinted organ using current bioprinting technologies, one alternative approach is to bioprint endothelial cells within the construct and have the microcirculatory system developed naturally within the host postimplantation (Ozbolat, 2015). Ischemic resilient organs, such as kidney, may be more amenable to this strategy than other visceral organs such as liver and pancreas. In addition, the vascular network within bioprinted organs should be lined with an endothelium layer to prevent blood coagulation. Tight junctions of endothelial cells can be achieved in vitro or such organization can be performed in vivo post-implantation if the size of the printed organs is small (Itoh et al., 2015). The other important requirement for bioprinted organs is their innervation capability depending on their type and function. During transplantation, it has been shown that transplants denervate for a period and recover nerve function over time posttransplantation. For example, histological staining of transplanted kidneys demonstrated evidence of nerve function restoration after weeks posttransplant; recovery continues for years (Mulder et al., 2013). Similar recovery of nerve function can be seen in other organs such as heart (Murphy et al., 2000). Thus nerve tissue should be incorporated into bioprinted organs as one of the future goals to augment currently existing allogeneic transplants.
After the operation, transplant patients are placed in the intensive care unit to manage fluid shifts and electrolyte balance. As 3D-printed organs are envisioned to be fabricated using autologous cells, the need for immunosuppressive agents may be minimal; however, if needed, transplant patients should be treated with immunosuppressive agents as early as possible and their dose should be adjusted based on the blood levels and functional status of the transplanted organ. One of the major postoperative issues is the nosocomial infections that are the fourth leading cause of morbidity and mortality (Kaye, 2011). Such infections arise due to biofilm formation on implants by Staphylococcus epidermidis. During in vitro bioprinting processes and long-term organ culture, bioprinted organs can be contaminated leading to a unique type of nosocomial infection. Therefore postoperative monitoring for clinical signs of infection (i.e., increased/decreased white blood cell count, fever, edema) and bioprinted organ failure is absolutely vital. Infection may require aggressive antimicrobial therapy. Postoperative status of organs can be monitored using noninvasive imaging techniques (i.e., computed tomography or magnetic resonance imaging) for connective tissues. For visceral organs, such as liver and kidney, different analyses can be informative such as serum bilirubin and INR to assess liver function, and creatinine and blood urea nitrogen to assess kidney function. As bioprinted organs are still in their infancy in terms of clinical translation, there might be many other unforeseeable issues and uncertainties. Patients receiving bioprinted implants may require more aggressive monitoring than traditional transplant patients.
Although autologous cells are envisioned to be used in organ printing, the bioink materials used from other species may elicit a host immune response after reimplantation as they acquire a layer of host protein, inducing immune response by interactions with inflammatory cells. Macrophages play an important role in adhesion of foreign body giant cells and release degradation factors, which could decompose the bioprinted organ. This can be mitigated by the use of antioxidants (Christenson et al., 2006). Thus monitoring of cytokine proinflammatory cytokine levels, such as interleukin-1 beta (IL-1β) and IL-6, might be essential in measuring cellular response and bioimplantation success.

8.4. Limitations

Despite the great progress in the last decade, whole-organ bioprinting is still a major roadblock. Current organ models are comprised of very small volumes that are appropriate for drug testing and disease modeling applications providing the appropriate physiological response has been realized; however, bioprinting of human scale organs has yet to be achieved (Ozbolat et al., 2016). There are a few major limitations requiring further investigation. One is to bioprint a multiscale vascular network from arteries and veins down to capillaries. Recent efforts demonstrated bioprinting larger-scale vessels using scaffold-based (Zhang et al., 2015) or scaffold-free (Norotte et al., 2009) approaches successfully. In addition, bioprinting-induced capillary network growth has been demonstrated in scaffold-based systems such as fibrin and collagen hydrogels (Lee et al., 2014a,b). Despite these advances, a vascular network in multiple scales has yet to be produced. Recent attempts in bioplotting in thermally reversible hydrogel bath demonstrated the fabrication of complex vascular network (Hinton et al., 2015); however, the use of limited cell densities hampers further efforts to connect them to capillaries.
Recently demonstrated efforts using fugitive ink for creation of perfusable channels present a suitable approach for fabrication of tissue models for drug testing and pharmaceutical purposes, or cancer or disease models (Peng et al., 2016). Such a system, on the other hand, is not convenient for transplant applications as cells are primarily embedded in hydrogels. Such an approach can support a small gel-based scaffold that can be implanted in connective tissues such as bone or subcutaneous tissue. For internal organs, this strategy can perhaps be used as a patch for solid organ repair, but may not be suitable for whole-organ bioprinting. As vascular networks are used as perfusable channels only and do not actually possess a native blood vessel anatomy, little experimentation has been done. It is expected that these channels will deteriorate in vivo due to biodegradation and may not maintain their morphology.
Recent work demonstrated the use of a thermoplastic-based frame utilizing PCL, which retains its shape upon printing due to its thermoplastic nature (Kang et al., 2016). Hydrogel-based composite bioink was reinforced within porous network along with fugitive Pluronic to generate porosity. Although larger-scale tissue construction was demonstrated, use of PCL, which has a prolonged degradation time, represents a major drawback in the proposed effort. Although PCL supports the structural integrity of hydrogel-based bioink materials, which might be crucial for load-bearing organs, its use for soft tissues may be inappropriate as it can interfere with the soft tissue regeneration. The volume of PCL material can possibly be minimized to reduce the effect of long-term existence of thermoplastic materials.
Use of hydrogel-based bioink currently entails a significant number of challenges as already described in the limitations section of Chapter 3 but the author prefers to detail some crucial points related to organ printing. One of the major drawbacks is the lack of cell–cell contact as cells cannot proliferate to a tissue-level density and this can interfere with the degradation process. Degradation brings significant challenges such as toxicity of by-products, elevated production of lactic acid, and lack of synchronicity between tissue regeneration and degradation of the hydrogel matrix. Therefore scaffold-free bioprinting is a promising approach in advancing tissue regeneration toward morphologically and physiologically relevant tissues; however, it may be dependent on the target organ type as well as the ultimate application such as pharmaceutical or transplantation use. For transplantation applications, there are further issues to be considered if the construct is for direct transplantation or transplantation following a prolonged in vitro culture.
One of the other current limitations is the lack of available organ-specific cells along with their differentiation protocols. For example, pancreas is a highly complex organ with endocrine and exocrine portions, where the endocrine portion is in charge of insulin production. In this regard, a replacement endocrine organ is vitally important for type I diabetes patients. The endocrine portion of an average human pancreas is made of approximately a million cell clusters called islets of Langerhans consisting of four main cell types including alpha (α), β, delta, and gamma cells secreting glucagon, insulin, somatostatin (regulates/stops α and β cells), and pancreatic polypeptide, respectively (Ozbolat and Yu, 2013). As human islets do not survive in vitro for longer periods and β cells die when they are disassociated from islets, successful differentiation of these cells is essential. Although there are a few seminal studies demonstrating the differentiation of functional beta cells (Pagliuca et al., 2015), there is a consensus that alpha cells also regulate the function of beta cells, therefore differentiation of other cell types is also important for proper function of engineered islets. Currently, the differentiation of these cells has not yet been achieved.
The lack of appropriate perfusion bioreactor systems for different organ type is also another obstacle. Although bioreactor technologies are available for geometrically simple organs such as irrigation dripping perfusion bioreactor for tubular tissues [i.e., blood vessels (Mironov et al., 2009)], other complex organs may possess different anatomies and printed organs may have additional special requirements such as a stepwise increase in the mechanical stress applied to the construct as the coherency of the tissue increases. In addition to limitations in bioreactor technologies, there are major limitations with the existing imaging techniques to quantify or establish the quality of bioprinted tissues. As organ printing necessitates the fabrication of larger-scale organ constructs, existing optical imaging technologies, such as confocal images or two-photon microscopy, are not capable of efficiently imaging the cells in solid organs when the cell density is high (Nam et al., 2014). Therefore multimodel imaging, such as using optical imaging along with nuclear medicine and radiographic imaging, can be used to demonstrate functionality, cell incorporation, and anatomy of the regenerating organs at high resolution.

8.5. Future Directions

In vitro fabrication of physiologically relevant tissues is a highly sophisticated phenomenon comprised of a hierarchical arrangement of multiple cell types, including a multiscale network of vasculature in stroma and parenchyma, along with lymphatic vessels and, occasionally, neural and muscle tissue, depending on the tissue type. In vitro–engineered organs that incorporate all of these components are still far out on the horizon. The major roadblock toward this ambitious goal is multiscale vascularization and the plethora of research that is required to further improve the alternative approaches previously presented. As larger vasculatures can be bioprinted using EBB systems, a controllable capillary network can be created naturally as demonstrated in hydrogels (Chung et al., 2009; Sheng et al., 2014). Since the timescale of neovascularization and the postbioprinting maturation of tissue construct is not the same, where neovascularization takes place in 10 days to 2 weeks; printed parenchymal cells require media and oxygen support immediately and therefore macrovascular network should be created with a diffusion distance of 200–300 μm (Zhang et al., 2013b) depending on the biomaterial and its interstitial flow capabilities. In addition, biomaterials with high microporosity can overcome the previously mentioned issues in some extent. Bioprinting technology offers a great benefit in the hierarchical arrangement of cells or construction of tissue blocks in a 3D microenvironment, but the bioink and the postbioprinting maturation phase are as important as the bioprinting process itself. Although hydrogels such as fibrin, collagen, and GelMA support neovascularization, they may not provide the ideal microenvironment and signaling for survival, motility, and differentiation of a wide array of tissue-specific cells; additionally, hydrogel stability over a prolonged in vitro culture period is weak (Aper et al., 2004; Nichol et al., 2010). Thus tissue-specific cell types can be bioprinted in scaffold-free manner. For example, pancreatic islets or lymphatic follicles in prevascularized form can be printed within a very small hydrogel coated on them, which can support growth of contiguous vascular network within spheroids along with capillaries sprouting into the hydrogel coating. These sprouts can further elongate and anastomose with sprouts from other spheroids (Peng et al., 2016). In addition, successful sprouting of these capillaries from spheroids to the macrovascular network is also crucial to make a fully contiguous vascular network. The postbioprinting process is also crucial and necessitates mechanical and chemical stimulation and signaling to regulate tissue remodeling and growth, development of new bioreactor technologies enabling rapid maturation of tissues, multiscale vascularization for survivability of printed organs, and mechanical integrity and innervation for transplantation.
In addition to efforts in printing organs at the clinically relevant dimensions, monitoring of these organs, postprinting as well as establishment of quality and regulatory standards for the entire process from stem cell isolation to posttransplantation monitoring during follow-up care is also essential. Thus involvement from other disciplines, such as quality assurance, economics, and law, will be highly beneficial to enable rapid and cost-effective fabrication of organs that meet stringent quality and safety standards.

8.6. Summary

This chapter presents the roadmap to organ printing technology that requires a sequence of interrelated processes spanning from isolation of stem cells to transplantation into a human; seamlessly automated protocols and systems are essential for customized functional organ fabrication. This pathway includes (1) isolation and differentiation of stem cells, (2) cell expansion, (3) bioink preparation, (4) blueprint modeling, (5) process planning, (6) bioprinting process, (7) organ remodeling and maturation in a bioreactor, (8) transplantation, and (9) posttransplantation monitoring. Although the technology shows a great deal of promise, there is still a long way to go to practically realize this ambitious vision. Overcoming current impediments in cell and biomanufacturing technologies, and innovative technologies for in vivo integration are essential for developing seamlessly automated platforms from stem cell isolation to transplantation.

References

Akkouch A, Yu Y, Ozbolat I.T. Microfabrication of scaffold-free tissue strands for three-dimensional tissue engineering. Biofabrication. 2015;7(3):31002.

Aper T, et al. Use of a fibrin preparation in the engineering of a vascular graft model. European Journal of Vascular and Endovascular Surgery : the Official Journal of the European Society for Vascular Surgery. 2004;28(3):296–302.

Baglioni S, et al. Characterization of human adult stem-cell populations isolated from visceral and subcutaneous adipose tissue. FASEB. 2009;23(10):3494–3505.

Bernard A.B, Lin C.-C, Anseth K.S. A microwell cell culture platform for the aggregation of pancreatic β-cells. Tissue Engineering Part C: Methods. 2012;18(8):583–592.

Bertassoni L.E, et al. Direct-write bioprinting of cell-laden methacrylated gelatin hydrogels. Biofabrication. 2014;6(2):024105.

Bunnell B.A, et al. Adipose-derived stem cells: isolation, expansion and differentiation. Methods. 2008;45(2):115–120.

Christenson E.M, Anderson J.M, Hiltner A. Antioxidant inhibition of poly(carbonate urethane) in vivo biodegradation. Journal of Biomedical Materials Research—Part A. 2006;76(3):480–490.

Chung S, et al. Cell migration into scaffolds under co-culture conditions in a microfluidic platform. Lab on a chip. 2009;9:269–275.

Fennema E, et al. Spheroid culture as a tool for creating 3D complex tissues. Trends in Biotechnology. 2013;31(2):108–115. .

Fisher M.B, Mauck R.L. Tissue engineering and regenerative medicine: recent innovations and the transition to translation. Tissue Engineering Part B: Reviews. 2012;19(1):1–13.

Gao Q, et al. Coaxial nozzle-assisted 3D bioprinting with built-in microchannels for nutrients delivery. Biomaterials. 2015;61:203–215.

Gaspar D.A, Gomide V, Monteiro F.J. The role of perfusion bioreactors in bone tissue engineering. Biomatter. 2012;2(4):167–175.

Gimble J.M, Katz A.J, Bunnell B.A. Adipose-derived stem cells for regenerative medicine. Circulation Research. 2007;100(9):1249–1260.

Gudapati H, Dey M, Ozbolat I. A comprehensive review on droplet-based bioprinting: past, present and future. Biomaterials. 2016;102:20–42.

Henzler H.J. Particle stress in bioreactors. Advances in Biochemical Engineering/Biotechnology. 2000;67:35–82.

Hinton T.J, et al. Three-dimensional printing of complex biological structures by freeform reversible embedding of suspended hydrogels. Science Advances. 2015;1(9).

Itoh M, et al. Scaffold-free tubular tissues created by a Bio-3D printer undergo remodeling and endothelialization when implanted in rat aortae. PLoS One. 2015;10(9):e0136681.

Janmey P.A, Winer J.P, Weisel J.W. Fibrin gels and their clinical and bioengineering applications. Journal of the Royal Society Interface. 2009;6(30):1–10.

Kang H, et al. A 3D bioprinting system to produce human-scale tissue constructs with structural integrity. Nature Biotechnology. 2016;34(3):312–319.

Kaye D. Copper kills 97 percent of hospital bacteria. Clinical Infectious Diseases. 2011;53(7):i–ii.

Kisiday J.D, et al. Effects of dynamic compressive loading on chondrocyte biosynthesis in self-assembling peptide scaffolds. Journal of Biomechanics. 2004;37(5):595–604.

Kolesky D, et al. Bioprinting: 3D bioprinting of vascularized, heterogeneous cell-laden tissue constructs. Advanced Materials. 2014;26(19):3124–3130.

Korossis S, et al. Bioreactors in tissue engineering. In: Ashammakhi N, Reis R.L, eds. Topics in Tissue Engineering. 2005:1–23.

Langer R, Vacanti J.P. Tissue engineering. Science. 1993;260(5110):920–926.

Lanza R, Langer R, Vacanti J, eds. Principles of Tissue Engineering. Elsevier; 2007.

Lee V, et al. Generation of multi-scale vascular network system within 3D hydrogel using 3D bio-printing technology. Cellular and Molecular Bioengineering. 2014;7(3):460–472.

Lee V.K, et al. Creating perfused functional vascular channels using 3D bio-printing technology. Biomaterials. 2014;35(28):8092–8102.

Li X, et al. Vitro recapitulation of functional microvessels for the study of endothelial shear response, nitric oxide and [Ca2+]iPLoS One. 2015;10(5):e0126797.

Liao I.-C, et al. Effect of electromechanical stimulation on the maturation of myotubes on aligned electrospun fibers. Cellular and Molecular Bioengineering. 2008;1(2):133–145.

Miller J.S, et al. Rapid casting of patterned vascular networks for perfusable engineered three-dimensional tissues. Nature Materials. 2012;11(9):768–774.

Mironov V, et al. Organ printing: tissue spheroids as building blocks. Biomaterials. 2009;30(12):2164–2174.

Mironov V, et al. Patterning of Tissue Spheroids Biofabricated From Human Fibroblasts on the Surface of Electrospun Polyurethane Matrix Using 3D Bioprinter. 2016.

Mondy W.L, et al. Computer-aided design of microvasculature systems for use in vascular scaffold production. Biofabrication. 2009;1(3):035002. .

Mulder J, et al. Renal sensory and sympathetic nerves reinnervate the kidney in a similar time-dependent fashion after renal denervation in rats. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2013;304(8):R675–R682.

Murphy D.A, et al. The heart reinnervates after transplantation. Annals of Thoracic Surgery. 2000;69(6):1769–1781.

Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovascular Research. 2006;69(3):562–573.

Nam S.Y, et al. Imaging strategies for tissue engineering applications. Tissue Engineering. Part B, Reviews. 2014;21(1):1–44.

Nichol J.W, et al. Cell-laden microengineered gelatin methacrylate hydrogels. Biomaterials. 2010;31(21):5536–5544.

Norotte C, et al. Scaffold-free vascular tissue engineering using bioprinting. Biomaterials. 2009;30(30):5910–5917.

Owens C.M, et al. Biofabrication and testing of a fully cellular nerve graft. Biofabrication. 2013;5(4):045007.

Ozbolat I.T. Bioprinting scale-up tissue and organ constructs for transplantation. Trends in Biotechnology. 2015;33(7):395–400.

Ozbolat I.T, Chen H, Yu Y. Development of “Multi-arm Bioprinter” for hybrid biofabrication of tissue engineering constructs. Robotics and Computer-Integrated Manufacturing. 2014;30(3):295–304.

Ozbolat I.T, Hospodiuk M. Current advances and future perspectives in extrusion-based bioprinting. Biomaterials. 2016;76:321–343.

Ozbolat I.T, Peng W, Ozbolat V. Application areas of 3D bioprinting. Drug Discovery Today. August 2016;21(8):1257–1271.

Ozbolat I.T, Yu Y. Bioprinting toward organ fabrication: challenges and future trends. IEEE Transactions on Bio-medical Engineering. 2013;60(3):691–699.

Pagliuca F.W, et al. Generation of functional human pancreatic β cells in vitro. Cell. 2015;159(2):428–439.

Pan C, et al. Comparative proteomic phenotyping of cell lines and primary cells to assess preservation of cell type-specific functions. Molecular and Cellular Proteomics : MCP. 2009;8(3):443–450.

Pati F, et al. Printing three-dimensional tissue analogues with decellularized extracellular matrix bioink. Nature Communications. 2014;5:3935.

Peng W, Unutmaz D, Ozbolat I.T. Bioprinting towards physiologically relevant stissue models for pharmaceutics. Trends in Biotechnology. 2016 (in press).

Piqué A. The matrix-assisted pulsed laser evaporation (MAPLE) process: origins and future directions. Applied Physics A. 2011;105(3):517–528.

Placzek M.R, et al. Stem cell bioprocessing: fundamentals and principles. Journal of the Royal Society Interface. 2009;6(32):209–232.

Saidi R.F, Hejazii Kenari S.K. Challenges of organ shortage for transplantation: solutions and opportunities. International Journal of Organ Transplantation Medicine. 2014;5(3):87–96.

Salehi-Nik N, et al. Engineering parameters in bioreactor’s design: a critical aspect in tissue engineering. BioMed Research International. 2013:762132 2013.

Sekine H, et al. In vitro fabrication of functional three-dimensional tissues with perfusable blood vessels. Nature Communications. 2013;4:1399.

Sheng W, et al. Capture, release and culture of circulating tumor cells from pancreatic cancer patients using an enhanced mixing chip. Lab on a Chip. 2014;14:89–98. .

Stabler C.L, et al. In vivo noninvasive monitoring of a tissue engineered construct using 1H NMR spectroscopy. Cell Transplantation. 2005;14(2–3):139–149.

Stenderup K, et al. Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells. Bone. 2003;33(6):919–926.

Takahashi K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–872.

Takebe T, et al. Generation of a vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature Protocols. 2014;9(2):396–409.

Thomson J.A, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145–1147.

Wert G.D. Human embryonic stem cells: research, ethics and policy. Human Reproduction. 2003;18(4):672–682.

Xu T, et al. Fabrication and characterization of bio-engineered cardiac pseudo tissues. Biofabrication. 2009;1(3):035001.

Yang H, Miller W, Papoutsakis E. Higher pH promotes megakaryocytic maturation and apoptosis. Stem Cell. 2002;20:320–328.

Yu Y, et al. Three-dimensional bioprinting using self-assembling scalable scaffold-free “tissue strands” as a new bioink. Scientific Reports. 2016;6:28714.

Yu Y, Zhang Y, Ozbolat I.T. A hybrid bioprinting approach for scale-up tissue fabrication. Journal of Manufacturing Science and Engineering. 2014;136(6):61013.

Zhang Y, Yu Y, Chen H, et al. Characterization of printable cellular micro-fluidic channels for tissue engineering. Biofabrication. 2013;5(2):025004.

Zhang Y, et al. In vitro study of directly bioprinted perfusable vasculature conduits. Biomaterials Science. 2015;3(1):134–143.

Zhang Y, Yu Y, Ozbolat I.T. Direct bioprinting of vessel-like tubular microfluidic channels. Journal of Nanotechnology in Engineering and Medicine. 2013;4(2):020902.

..................Content has been hidden....................

You can't read the all page of ebook, please click here login for view all page.
Reset
3.144.227.9