Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jan 23.
Published in final edited form as: Appl Mater Today. 2022 Jul 6;29:101582. doi: 10.1016/j.apmt.2022.101582

Recent advances in organoid engineering: A comprehensive review

Janitha M Unagolla a, Ambalangodage C Jayasuriya a,b,*
PMCID: PMC10804911  NIHMSID: NIHMS1959765  PMID: 38264423

Abstract

Organoid, a 3D structure derived from various cell sources including progenitor and differentiated cells that self-organize through cell-cell and cell-matrix interactions to recapitulate the tissue/organ-specific architecture and function in vitro. The advancement of stem cell culture and the development of hydrogel-based extracellular matrices (ECM) have made it possible to derive self-assembled 3D tissue constructs like organoids. The ability to mimic the actual physiological conditions is the main advantage of organoids, reducing the excessive use of animal models and variability between animal models and humans. However, the complex microenvironment and complex cellular structure of organoids cannot be easily developed only using traditional cell biology. Therefore, several bioengineering approaches, including microfluidics, bioreactors, 3D bioprinting, and organoids-on-a-chip techniques, are extensively used to generate more physiologically relevant organoids. In this review, apart from organoid formation and self-assembly basics, the available bioengineering technologies are extensively discussed as solutions for traditional cell biology-oriented problems in organoid cultures. Also, the natural and synthetic hydrogel systems used in organoid cultures are discussed when necessary to highlight the significance of the stem cell microenvironment. The selected organoid models and their therapeutic applications in drug discovery and disease modeling are also presented.

Keywords: Organoid, Extracellular matrix, Microfluidics, Bioprinting, Self-assembly

1. Introduction

The three-dimensional (3D) cell culture in the form of cell aggregation was attained its first popularity in 1965–1985, according to the number of published materials [1,2]. Despite the name “organoids” was introduced to the scientific community half a century ago, this name was used to describe classical biological experiments that sought to define organogenesis by cell dissociation and reaggregation experiments [1]. Later, the emergence of the 3D cell cultures has fueled the knowledge on the effectiveness of the 3D cell cultures compared to the conventional two-dimensional (2D) cell cultures. The 2D cell cultures do not provide the conditions required for cellular organization and cell interactions in vivo. Also, the cell signaling networks are altered in 2D cell culture versus 3D cell cultures. In contrast to the 2D cell culture, 3D cell culture models exhibit very close properties to the in vivo conditions. The 3D culture models provide more realistic findings similar to in vivo conditions in translational research [3]. The 3D culture models are generally either two models: scaffold-based models or scaffold-free models. In scaffold-based 3D culture, the cells are grown in the substrate, which mimics the extracellular matrix (ECM) properties in either hydrogel or solid scaffolds; while cells are unable to attach to the substrate and force to make cell aggregates or cell spheroids in scaffold-free method [4,5]. The past decade again witnessed the organoid’s resurgence, yet in a somewhat different form due to the parallel development of 3D culture techniques. The breakthrough of the current organoid resurgence was made by Hans Clevers and his colleagues in 2007 after discovering a new type of stem cells [6]. Organoid technology is now on the verge of emerging as an independent research field.

The term organoid has lost accuracy in recent years and has been defined broadly according to the different cell culture techniques and their respective applications. However, this broad applications from small tissue explants to clonally expanding cells that self organize have made it difficult to define or otherwise, have made its meaning ambiguous. Recently, experts from different part of the world came together and defined the organoid as a “3D structure derived from (pluripotent) stem cells, progenitor, and/or differentiated cells that self-organize through cell-cell and cell-matrix interactions to recapitulate aspects of the native tissue architecture and function in vitro” [7]. However, according to the early definitons and studies of the organoid, it has been established that organoids are exclusively derived from the stem cells [1, 811]. It is now clear that organoid can generate from differentiated cells such as cholangiocytes [12,13]. Due to this recent expert meeting, previously believed [11] organoid definition and characteristics of the organoid was changed to a newer version and in this review we focused on the latest definition of the organoid. So, recently new catergorization for the organoid was proposed. Organoids cen be devided into three distict groups based on their characteristics; (i) epithelial organoids, (ii) multi-tissue organoids, and (iii) multi-organ organoids [7].

The first category, epithelial organoids are derived from a single germ layer (endoderm, mesoderm, or ectoderm). Epithelial organoid can self-renew under appropriate culture conditions and also represents the most widely studied organoid type. Since epithelial organoids do not contain mesodermal component, in some cases, supporting cells are cocultured with epithelial organoids [14,15]. Multi-tissue organoids at least have two germ layer cells or co-differentiation of PSCs, which are established through co-culture. Interestingly, current studies do not support the self-renewal of multi-tissue organoids in normal pathways, but cells interact to achieve a stable level of maturity and function [16, 17]. The most complex and the least studied organoid type is multi-organ organoids, where these organoid can make a significant contribution for study of organogenesis.

Organoids can be derived from different types of cells: (i) pluripotent embryonic stem cells (PESCs) and their synthetic counterparts, induced pluripotent stem cells (iPSCs); (ii) organ restricted adult stem cells (aSCs) [1]; and (iii) differentiated cell types such as cholangiocytes and hepatocytes, as described in the recent literature [18]. PESCs and iPSCs are simply known as pluripotent stem cells (PSCs) in most literature. Both these PSCs and aSCs have shown an unprecedented capacity to self-organize into structures that mimic the fundamental properties of the tissues which they are supposed to form. As mentioned above, human iPSCs (hiPSCs) play a significant role in the recent development of novel organoid models [10,1921]. The discovery of the iPSCs has resulted in the development of multiple differentiation protocols in vitro using various endoderm-derived tissue types, including stomach [22], kidney [23], liver [2426], lung [2729], pancreas [30], and intestine [31]. The invention of directed differentiation of tissue-specific cell types from iPSCs has resulted in the investigation of different organoids, including the brain [32], kidney, liver, lung [33], pancreas, and stomach. A more detailed review of some organoid types is discussed in the latter part of this paper.

As previously mentioned above, unlike the traditional in vitro cell cultures, organoids have complex cellular composition and architecture, making them a physiologically complex model to study tissue development process, tissue homeostasis, and cellular functions and signaling pathways present in tissues in vitro [10,34]. Theoretically, the organoids made from human stem cells should mimic the actual human organ functions, especially conditions that do not replicate well in experimental animals [6,34]. However, most organoid models do not replicate the actual human organ models for various reasons, including the difficulty of generating full tissue components, difficulty controlling cell types, and complexity in cell-cell and cell-matrix interactions in these systems. The dependence on the limited animal-derived hydrogels, including Matrigel and collagen as an ECM, makes organoids unsuitable for expansion and downstream clinical applications due to their uncontrolled modifications and risk of pathogen and immunogen transfer [35]. Due to these reasons, despite the extensive use of organoids in basic and translational research, organoids are currently restricted to drug screening and initial cell replacement strategies [8].

In this review, we discuss the recent advancement and trends in organoid technology, including different organoid models and their significance to the development of current therapeutic studies. We critically evaluate the use of synthetic or natural polymer as an ECM for organoid formation. Spheroids as an intermediate stage for organoid development are also be addressed by emphasizing recent discoveries. We also highlight the key challenges and limitations of the current organoid technologies, which remain to be addressed in the future.

2. The formation of organoids: self-organization

The formation of an organoid from iPSCs or aSCs or differentiated cells is a controlled process, uses biochemical and physical cues for tissue development and homeostasis. The development of the human body from the zygote is a precisely controlled process, which uses stepwise differentiation and self-organization of the cells. Understanding the self-assembly of organoids solely depends on the classic developmental biology theories. The tissue patterning is essential to understand the self-organization of the organoids. There are two distinct approaches to examine tissue patterning in vitro and in vivo. During tissue morphogenesis, cells segregate into discrete domains in vivo [11]. This phenomenon was observed in the Drosophila wing disc, where the anterior-posterior boundary is established through mutually repressive interactions [36], and a similar process was observed in vertebrate embryonic development at the midbrain-hindbrain boundary [37]. The relative morphogenetic movements of the cells in vitro can be examined through the dissociation and reaggregation of tissues as the second approach of tissue patterning. This method was primarily applied in the development of vertebrate organs [38,39].

The organ’s self-assembly mainly occurs due to the segregation of cells with similar adhesive properties into domains that create a thermodynamically most stable pattern, known as Steinberg’s differential adhesion hypothesis. This phenomenon is also known as cell sorting out. The second mechanism which influences the self-assembly is proper spatially restricted fate decisions. These two mechanisms are schematically represented in Fig. 1(B) and Fig. 1(C). The organoid’s self-assembly is possible because of the growing movement away from the 2D culture, which results in the formation of tubules or ducts when the epithelial cells are embedded into ECM hydrogel [11], but not all the epithelial organoids form tubles when embedded in hydrogel. So, ECM hydrogel is a major driving force of self-assembly of the organoids, which is discussed further in a later section of this paper.

Fig. 1.

Fig. 1.

(A) Schematic representation of formation of organoids; (B) Cell sorting out describes the movement of cells into different domains. Different cell types (purple or green) sort themselves because of different adhesive properties conferred by their differential expression of distinct cell adhesion molecules (shown as brown or orange bars); (C) Spatially restricted cell-fate decisions also contribute to self-organization in vivo and in organoids. Progenitors (green) give rise to more differentiated progeny (purple), which, because of spatial constraints of the tissue and/or division orientation, are forced into a more superficial position that promotes their differentiation. These cells can sometimes further divide to give rise to more differentiated progeny (pink), which are further displaced [11].

Apart from the above-mentioned natural self-assembly mechanisms, growth factors and small molecules are used to control the self-renewal and differentiation of stem cells by manipulating cell signaling pathways in a tissue-specific manner [34]. The additional supplementation of signaling cues for cell differentiation and self-assembly also supports the ECM materials. The most commonly used ECM material, Matrigel, provides signaling cues through basement membrane ligands to support cell adhesion [40]. Since most of the organoids extensively rely on stem cells, the stem cell microenvironment or niche plays a significant role in self-assembly and directed cell differentiation. Niche functions as a physical anchor for cells, and also extrinsic and intrinsic biochemical signals are generated with the help of different growth factors. Altogether, the organoid formation can be programmed according to the niche, which provides a wide range of biochemical and biophysical signals, cell-cell interactions, and cell-ECM interactions. However, the dependent on cell-autonomous self-organization during organoid formation is not yet easily controlled, and hence more investigations are required to control the organoid formation [11,34,41].

3. Organoid systems

Organoid systems offer promising platforms for future therapeutic studies and stem cell applications focusing on tissue and organ regeneration. However, similar to most model systems, there is a significant difference between in vitro and in vivo models, which can be addressed through various bioengineering techniques. Bioengineering approaches can be applied to develop bottom-up synthetic organoid constructs, and facilitate controlled organoid formation through different cell cultures. The natural self-assembly of cells with the help of a suitable microenvironment has limitations due to the lack of biochemical and biophysical cues. The organoid’s niche components are established from either cell siganling as autocrine, paracrine, or juxtracrine, which always has limitations; or exogenously added to the system as ECM substrates, small molecules, or growth factors. Spatial and temporal control of the dynamic environment during the differentiation of the cells can not be controlled properly without the help of exogenous substances during complex organoid formation. There are two approaches to organoid engineering: (i) the cell biology approach modulates cell behavior and programs the self-assembly and cell differentiation; (ii) the bioengineering approach manipulates the microenvironment and the cell organization through different approaches, such as scaffold-free printing and 3D bioprinting. However, to accomplish the best organoid system, a combination of both cell biology and bioengineering approaches is required. Recent advances in biomaterials, hydrogel systems, cell-driven tissue assemblies, micro/nanotechnology, and bioprinting techniques have enabled the possibility of designing dynamic and self-assembled organoids.

3.1. Engineering the cell microenvironment (niche)

Engineering the niche, which is similar to humans or animals, would be a breakthrough for humankind. The complexity of the niche is a primary limitation in organoid system development. Niche consists of soluble and surface-bound cytokines, ECM-cell interaction, cell-cell interaction, mechanical forces, and physicochemical cues, such as oxygen and pH [42]. The cytokines can be delivered as growth factors incorporated with an ECM material, using different chemical modifications such as covalent bonding. The controlled delivery of these growth factors using biodegradable scaffolds or microfluidic devices provides spatially and temporally restricted delivery. Recent studies have also demonstrated that the immobilization of these cues to the ECM plays an important role in mediating their biological effects [4244]. For example, dexamethasone, a corticosteroid, and a potent modulator (growth factor) for osteogenic differentiation have incorporated into poly(ethylene glycol) (PEG) based hydrogel system to release slowly with the degradation of PEG. At the same time, mesenchymal stem cells (MSCs) differentiated into bone tissue [45]. The manipulation of the ECM material is vital for the growth and differentiation of stem cells into their intended application.

ECM, the noncellular component presents in all tissues and organs, is one of the essential components of the cell microenvironment and provides not only essential structural support for the cellular constituents but also initiates crucial biochemical and biomechanical cues that are required for tissue morphogenesis, differentiation, and homeostasis [46]. ECM components such as collagen, laminin, and fibronectin provide the physical structure of the tissues and also influence cellular behavior by engaging with integrin receptors [44]. ECM-cell interaction can be stimulated through several approaches, including synthesizing novel biomaterials relevant to specific stem cells [47], producing 3D scaffolds with micro or nanoscale topography [48], 2D micropatterning, and high-throughput microarrays [49]. The decellularized matrices also can be used as ECM components. For example, after the confluency in vitro, the bone marrow stromal cell matrix can be decellularized without disrupting the underlying ECM matrix and subsequently used to mimic endosteal and vascular niches with the help of relevant growth factors [34,50].

The most commonly used ECM component of organoid culture is Matrigel. Matrigel, a heterogeneous, complex mixture of ECM proteins including laminin, collagen IV, and enactin, is secreted by Engelbreth-Holm-swarm mouse sarcoma cells [51,52]. The exclusive dependence on this animal-derived hydrogel is complicated by current organoid methods due to the lot-to-lot compositional and structural variability, pose a risk of immunogen and pathogen transfer, and, most importantly, this tumor-derived matrix has limited clinical translational potential [35,52,53]. Moreover, the heterogeneous nature of the Matrigel does not easily allow for manipulating the morphogenetic processes, which are tightly governed in vivo by spatiotemporal cues [34]. However, Matrigel [22,54] and collagen [5557], are extensively used in organoid systems. As an alternative to the animal-derived ECM, synthetic hydrogels can be tailored to produce the essential signals by incorporating specific biomimetic cues, such as glycosaminoglycans, a major ECM component believed to play an essential role in the cell niche. For example, hyaluronic acid (HA), a glycosaminoglycan highly expressed in bone marrow stromal cells, has played an essential role in regulating hematopoiesis in the hematopoietic stem cell niche modulating neural stem cell niche [44]. The development of synthetic polymer-based ECM, which facilitates cellular infiltration and adequate nutrient transport [58], is still at the primitive stage despite some recent advancements in synthetic hydrogels [34].

The recent advances in chemical methods, such as click chemistry, Michael type addition reactions, and gelation mechanisms, provide more controlled physicochemical properties in synthetic polymers [59]. Also, the bioinert environment of the synthetic polymers can be modified into a biomimetic environment similar to the natural ECM by adding biomimetic cues/signaling proteins through chemical or enzymatic cross-linking. Recently, the PEG-based hydrogel system has been studied for different organoid systems as a biomimetic ECM material. A PEG-based hydrogel, maleimide crosslinked four-arm PEG macromer (PEG-4MAL), has shown superior cytotoxicity and minimal toxicity and inflammation [60,61]. The PEG-4MAL hydrogel system enabled the successful growth of human intestinal organoids (HIO), which normally relies on undefined tumor-derived ECM. PEG-4MAL macromers were functionalized with RGD (Arg-Gly-Asp) adhesive peptides and crosslinked in the presence of hPSCs to generate HIO. The viability and mechanical stability were evaluated using Matrigel-derived HIO as a control model, and different concentrations of PEG-4MAL (3.5–6.0% w/v) were studied to understand polymer density-dependent effects on HIO survival. The viability of 3.5% and 4% PEG-4MAL based HIOs were similar to the Matrigel-based control group, while 5% and 6% PEG-4MAL groups showed lower HIO viability, suggesting the polymer density-dependent effect on survival of HIO as shown in Fig. 2 [62,53]. The ECM hydrogel should provide mechanical stability to the organoid system and the biochemical signals required for cell survival and proliferation, and the stiffness of the polymer is also important to the viability of the organoid structure, as suggested by the above study.

Fig. 2.

Fig. 2.

(A) (i) Illustration of the designed 384 hanging drop spheroid culture array plate; (ii) Photo and key dimensions of the array plate. (iii) Cartoon of the hanging drop formation process in the array plate. Within hours, individual cells start to aggregate and eventually form into a single spheroid around 1 day; (iv) Photo of the 384 hanging drop array plate operated with liquid handling robot capable of simultaneously pipetting 96 cell culture sites; (e) Cartoon of the final humidification chamber used to culture 3D spheroids in the hanging drop array plate [62]; (B) Synthesis of the PEG-4MAL hydrogel and organoid generation; (i) Floating, hPSC-derived human spheroids are collected and mixed with a solution of functionalized PEG-4MAL macromer. (ii) PEG-4MAL hydrogel is cast by pipetting the mixture of functionalized PEG-4MAL and spheroids into the cross-linker solution. Encapsulated spheroids expand and develop into human organoids [53].

3.2. Engineering organoids

Engineering organoids mainly focus on developing a suitable microenvironment for cells and overcoming the challenges aligned with the complexity of the cell niche. These challenges can be overcome by advancing material chemistry, such as easy and less cytotoxic click chemistry and Michael type addition reactions for novel hydrogel synthesis, and developing the bioengineering techniques that generate the stem cell niche by incorporating bioreactors, microarray systems, and microfluidics. The latter part of this, the bioengineering techniques will be further discussed, including the different techniques such as bioreactors, microfluidics, bioprinting, vascularization, and organoid-on-a-chip models. The summary of each bioengineering technique is shown in Table 1. Most organoid system’s main building block is cell spheroids; therefore, engineering 3D cell spheroids is discussed further, mainly focusing on the material perspective.

Table 1.

Summary of the main Microengineered systems used for organoid formation with the type of application, cell types, and applications.

Micro engineered system Type of organoid Cell types Hydrogel type Applications
Bioreactors Brain Human iPSCs Matrigel Studying Zika virus-induced microcephaly [19]
Exploring development of forebrain, midbrain, and hypothalamus organoids [87]
Providing largest molecular map of the diversity of cell types that generated in the brain organoid [148]
Retinal Human iPSCs Generating retinal organoids that contains retina-like architecture with enhanced generation of photoreceptors [85]
Mouse PSCs Developing a RWV bioreactor to promote the growth of retinal organoid that is closely recapitulating the spatiotemporal development of mouse retina in vivo [83]
Kidney Human iPSCs Exploring inexpensive and highly efficient method to grow kidney organoid that contains podocytes, proximal and distal tubles segments in developed nephrons [149]
Hepatic/ liver Human PSCs Exploring the impact of dissolved oxygen concentration on hPSCs hepatic differentiation fate and differentiation efficacy [86]
Adult upcyte® cells Generating liver organoids from adult differentiated cells and studying long-term liver function [150]
Microfluidics Gastric/ Intestinal Gastrointestinal cells Alginate/ Gelatin Exploring feasibility and biocompatibility of nonfluorinated oils for application in gastrointestinal cells to study intercellular chemical interaction [96]
Brain Human NESCs/ hiPSCs Modelling a millifluidic culture system that supports continuous media supply by laminar flow which reduces the “dead core” of the brain organoid [151]
Liver HepG2/ NIH3T3 Collagen Developing early stage drug screening system for hepatotoxicity [152]
Induced hepatic cells Decellularized porcine liver tissue Developing a highly effective, functional 3D hepatic culture system that is suitable as a drug screening platform using a decellularized LEM hydrogel [91]
Human iPSCs Fibrin/ chitosan/ Alginate Synthesizing composite hydrogel capsules that enable the engineering of liver organoids in oil-free droplet microfluidic system [95]
Lung cancer Patient-derived small-cell lung cancer Matrigel Exploring the drug responses under physiologically relevant flow conditions using lung cancer organoid culture platform [93]
Bioprinting Vascularized ossification Human UVECs Alginate Developing a novel airflow-assisted bioprinting method to produce spiral-based spheroids with excellent resolution and complex microenvironment [153]
Colon aSCs GelMA Introducing bovine colon organoid for agri-biotechnological applications [125]
Mammary gland Epithelial/cancer cells Human mammary ECM Exploring a novel mammary ECM hydrogel in 3D bioprinting to develop sustained and prolonged organoid formation and differentiation [126]
Mamary epithelial hMECs Collagen Exploring a 3D bioprinting platform to control the 3D formation of organoids through the “self-assembly” of hMECs [127]
Salivary glands hDPSCs Validating a magnetic 3D bioprinting system to generate innervated secretory epithelial organoids [154]
Brain (cancer derived) Human glioblastoma cells PEGDA BPADMA Developing a 4D printed programmable cell-culture arrays using thermo-responsive shape memory polymer for cancer drug testing [155]
Organoid-on-a-chip Brain hiPSCs Matrigel Investigating the adverse effect of nicotine on cortical development [138]
Stomach/ gastric hPSCs Developing a gastric organoid that reminiscent of gastric motility by introducing a rhythmic stretch and contraction through a luminal flow [145]
Kidney hPSCs/Metanephric mesenchyme cells Exploring the flow generated microenvironment as a pivotal contributor to the structural and functional development of kidney organoids [146]
Intestine Collagen Introducing a new system to develop the crypt and villus architecture that is distinct to intestine epithelium using a diffusion gradient [147]
Cortical hiPSCs Matrigel Investigating the autism-like neurodevelopment disorder due to the prenatal antiepileptic drug valproic acid (VPA) exposure [156]
Liver hPSCs Investigating the improved cell viability and higher expression of endodermal genes and mature hepatic genes under perfused culture conditions to investigate hepatotoxic response after exposure to acetaminophen [157]
hiPSCs Developing a nonalcoholic fatty liver model to explore the underlying conditions of steatosis and study the effective therapies [158]

3.2.1. Engineering 3D cell spheroids

Most currently available organoid development protocols utilize the encapsulation of 3D spheroids, using biologically active materials, including natural and synthetic polymers [53]. Therefore, we introduce the 3D cell spheroids and their fabrication techniques with the recently published studies. 3D spheroid is also a 3D cell culturing method, as explained in the introduction section. In general, cells are cultured as aggregates in 3D culturing methods. The spheroid culture methods are among the most well-investigated areas due to their simplicity, reproducibility, and ability to make tissues similar to physiological tissues [62]. The 3D spheroids also can be considered as an organoid if they contain organ-specific cell types and satisfy the definition of an organoid [7].

Cells cannot attach to the biomaterial-based ECM surface, therefore, in spheroid formation, cells are forced to attach and form aggregates [62,63]. Numerous fabrication techniques have been demonstrated successful spheroid formation, including hanging-drop cultures [6466], microfluidic devices [67], liquid overlay/cell suspension system [68], microwell arrays [6971], and cultures on low-adhesive substrates [72,73]. From the methods mentioned above, hanging drop cultures, microwell arrays, and microfluidic systems are able to generate highly controlled aggregates with uniform size [74].

The development of robot-assisted hanging drop culture systems allows the production of large-scale spheroids in a single array at a low cost. The robot-assisted hanging drop system enabled the production of 384 spheroids in a single array, as shown in Fig. 2(A) [62]. The spheroid size is more critical for cell viability because oxygen and nutrients are transferred to cells using diffusion-driven mechanisms. Therefore, the generation of controlled-sized spheroids, which do not exceed a few hundred micrometers, is vital to avoid necrotic damage to the spheroid’s core. Moreover, these approaches enable incorporating the cells into the biomimetic hydrogel systems while maintaining the nutrient supply and interspheroid metabolic communication [63]. Due to the 3D structure, spheroids offer a wide variety of improved biological properties compared to the 2D single-cell culture systems, including enhanced cell viability, increased proliferation, differentiation, stable metabolic function, and stable morphology. Therefore, spheroids models are extensively used in cancer research as a biomimetic in vitro model, and also, they are extensively used in drug discovery and toxicology screening. 3D spheroid systems are also gained popularity due to the enhanced differentiation capability of multipotent MSCs into osteogenic, chondrogenic, neurogenic, adipogenic, and hepatogenic lineage compared to the 2D culture models. The insufficient oxygen diffusion to the core of spheroids triggers the upregulation of hypoxia-induced survival factors, such as hypoxia-inducible factor (HIF)-1α. These types of reactions result in increased secretion of angiogenic factors. Also, the spheroid diameter >100 μm exhibits more prominent upregulation of HIF-1α and vascular endothelial growth factor (VEGF) secretion and thus, support the vascularization. However, as previously mentioned, too low oxygen levels in larger spheroids may significantly compromise the cell viability, and therefore, it is required to determine the ideal size of spheroids for angiogenesis.

The current organoid formation methods mostly use 3D spheroids as an intermediate building box to form the final organoid. The progress of 3D spheroid technologies enables new paths for organoid engineering as it allows us to generate more complex cell aggregates, including rods, tori, and honeycombs, with a combination of hydrogels. However, these areas must be exploited further. Still, in organoid systems, Matrigel-based cell spheroids are widely used due to their excellent biomimetic capabilities in vitro. Matrigel-based hiPSC spheroids were used in HIO formation [54] and gastric organoid formation [22]. Also, a synthetic hydrogel, PEG-4MAL, was used to make hPSC-derived spheroid to form organoids after crosslink with a functionalized peptide, as shown in Fig. 2(B) [53]

3.2.2. Bioreactors

The bioreactor is a dynamic 3D cell culture platform that can be used in various distinct organoid-related applications, including the expansion of diverse cell types such as PSCs, generation of 3D organoids from PSCs, and tissue-specific differentiation of stem cell-derived organoids [75,76]. The dynamic nature of the medium around the stem cells in the bioreactor system facilitates organoids’ maintenance and differentiation. The flow around the cells further improves medium exchange, facilitating the higher mass transfer and more physiologically relevant gradients of nutrients and waste products between the surface and the core [77,78]. The gradient acts similar to the natural developmental environment during cell differentiation, enabling the correct orientation and lamination of different cell phenotypes [34,79]. The induced mechanotransducive responses from cells have resulted from the shear generated by the flowing medium, enabling better structural and transcriptional outcomes [80]. Different bioreactors have been used extensively in organoid, and regenerative and tissue engineering approaches, including rotating wall vessel (RWV), stirred-tank bioreactors, spin bioreactors, and more described elsewhere [81,82]. The most recent studies of bioreactor-based organoids generating systems are summarized in this review.

DiStefano et al., reported RMV bioreactor to develop retinal organoid using mouse PSCs. The efficacy of retinal differentiation in RMV-cultured organoids was higher than static suspension-cultured organoids, as demonstrated by immunohistochemistry and transcriptome profiling. This system recapitulated the spatiotemporal development of mouse retina in vivo, but further differentiation of retinal organoids in vitro was impaired and required additional functional maturation conditions [83]. Phelan and the group developed a system capable of constantly removing air bubbles from the conventional RWV reactors without interfering with the fluid dynamics enabling optimum cell culture conditions. The air bubble formation of the conventional RWV reactors during operation negates the critical features of the RMV environment, such as low shear, zero headspace, and simulated micro-gravity. This modified bubble capturing bioreactor showed an enhanced organoid formation in alginate beads and spheroids containing A549 human lung adenocarcinoma cells [84].

A stirred-tank bioreactor was used to develop the retinal organoids to address the visual impairment caused by inherited retinal degeneration, retinal diseases, age-related macular degeneration, and glaucoma. The developed organoids using hESCs and hiPSCs showed improved laminar stratification and yield of photoreceptor cells bearing cilia and nascent outer-segment-like structure [85]. In another study, Farzaneh et al., reported the 20–40% of dissolved oxygen (DO) concentration significantly influences the differentiation of hPSC aggregates by enhancing the mesoderm induction. The resulted human fetal-like hepatic organoids contained red blood cells and functional hepatocytes, exhibiting improved liver-specific gene expression, key metabolic functions, and higher cytochrome P450 activity. Further, the implanted hepatic organoids in an acute liver injury mouse model showed albumin production after implanting, suggesting the effect of DO in the liver tissues [86]. Qian et al., developed a cost-effective, simple-to-use miniaturized multiwall spinning bioreactor to generate forebrain-specific organoids from hiPSCs for model ZIKA Virus (ZIKV) exposure. These generated organoids recapitulate human cortical development’s key features, including progenitor zone organization, neurogenesis, gene expression, and notably, a distinct human-specific outer radial glia cell layer. Also, the team has developed protocols for midbrain and hypothalamic organoids [87].

3.2.3. Microfluidics

Nearly two decades ago, the word “microfluidics” was introduced to the biology and cellular engineering research field as a technology that can significantly change the perception of modern biology. Microfluidics is characterized as the science and technology of systems that manipulate a small amount of fluid at the submillimeter length scale [88,89]. The field of microfluidics has evolved from four different areas: (i) molecular analysis; (ii) biodefense; (iii) molecular biology; and (iv) microelectronics. (i) Molecular analysis including high-pressure liquid chromatography (HPLC), gas-pressure chromatography (GPC), uses a minimal amount of samples to achieve high sensitivity and high resolution and, therefore, there was a necessity to develop a more versatile and more compact microscale method in chemistry and biochemistry. (ii) The development of the field-deployable microfluidic systems for detecting biological and chemical threats in 1990 by the United States department of defense paved the stimulus for the rapid growth of microfluidic technology. (iii) After the explosion of genomics in the 1980s, the molecular biology field required analytical methods with higher sensitivity and higher throughput and resolution. (iv) The most influential contribution was coming from microelectronics. Photolithography and related technologies that had been first developed for the semiconductor industry and micro-electromechanical systems (MEMS) are directly applicable for microfluidic systems. Indeed, the earlier microfluidic devices were made using silicon and glass but later replaced with plastics [88]. In recent years, the field of microfluidics has under-gone rapid growth and has become an integral part of many biological and biomedical engineering applications. The simultaneous development of materials and microfluidic platforms is vital to meet the requirement of complex microenvironments [90]. Especially, organoid cultures require a dynamic culture environment to replicate the actual biological conditions in vivo. The microfluidic systems provide dynamic culture conditions with the help of automation by supplying continuous inflow and outflow of medium and nutrition through microchannels and further facilitate the long-term expansion of organoids and uniformity. The microfluidic systems extensively used in drug screening research and integrating microfluidic components into a single chip led to the advent of lab-on-a-chip devices. The recent studies of microfluidic-based organoids generating systems will be summarized in this review, while the more recent organoids-on-a-chip model will be discussed in the later section.

Jin et al., reported a 3D microgel liver organoid as a drug screening platform that showed higher sensitivity and reduced hepatotoxic response variation. Liver extracellular matrix (LEM) hydrogel was synthesized using decellularized porcine liver, and induced hepatic (iHep) cells were differentiated from primary mouse embryonic fibroblasts. The encapsulated iHep cells cocultured with the human umbilical vein endothelial cells (hUVECs) in LEM hydrogel (or 3D cell culture) in a dynamic state (continuous flow) using a simple rocker system showed reduced apoptosis and improved cell-cell adhesion in 3D iHep tissue. These organoids also showed enhanced hepatocyte-specific functions, including urea secretion and serum albumin secretion [91]. Most current organoid protocols involve several steps of cell seeding, differentiation, and maturation before evaluating the drug responses. Therefore, recently several studies were carried on to study the one-stop organoid formation for drug screening. Ao et al., reported a one-stop brain organoid formation method using a microfluidic platform containing perfusable chambers made out of poly (dimethylsiloxane) (PDMS) and poly(tetrafluoroethylene) (PTFE) coated steel mesh, as shown in Fig. 3A. An in situ air-liquid interface was used to reduce the hypoxic core. After day 10, the top chamber medium was removed to facilitate the air-liquid interface culture condition, as shown in Fig. 3A. This microfluidic system was used to analyze the effect of prenatal cannabis exposure on brain development [92]. Jung et al., and the team demonstrated a one-stop microfluidic platform that can reproduce 3D lung cancer organoids in size controllable manner. The lung cancer organoids derived from the patient-specific small-cell lung cancer (SCLC) were demonstrated to preserve SCLC’s morphological and genetic characteristics during the culture period. Also, this microphysiological system can provide insight into the efficacy of the anti-cancer drugs and can further study the mechanisms of possible chemotherapy resistance, which is common in SCLC [93].

Fig. 3.

Fig. 3.

(A) Schematics of brain organoid fabrication and cannabis exposure within the microfluidic device [92]. (B) Printing process of particular tissue constructs with dECM bioink; hHeart tissue construct was printed with only heart dECM (hdECM). Cartilage and adipose tissues were printed with cartilage dECM (cdECM) and adipose dECM (adECM), respectively, and in combination with PCL framework (scale bar, 5 mm) [119]. (C) Bioprinting of aortic valve conduit (i) Aortic valve model reconstructed from micro-CT images. The root and leaflet regions were identified with intensity thresholds and rendered separately into 3D geometries into STL format (green color indicates valve root and red color indicates valve leaflets); (ii) schematic illustration of the bioprinting process with dual cell types and dual syringes; (iii) fluorescent image of first printed two layers of aortic valve conduit; SMC for valve root were labeled by cell tracker green and VIC for valve leaflet were labeled by cell tracker red. (iv) as-printed aortic valve conduit [122].

In another study, Schuster et al., developed an automated, high-throughput 3D gel-based microfluidic platform capable of culturing organoids for continuous monitoring of growth, morphology, and biochemical analysis. This system consists of two integrated devices; a 3D culture chamber and multiplex fluid control device, automated and programmable experimental control with custom-made software, and live-cell time-lapse fluorescence microscopy. The patient-derived tumor organoids developed in this work showed significant differences in individual patient’s drug responses, suggesting the importance of studying drug response at the individual level for cancer patients [94]. Apart from long-term organoid cultures using microfluidic devices, microbeads or composite hydrogel-based microbeads were recently synthesized using microfluidic devices while culturing conventional culture plates. Wang et al., reported a one-step synthesis of composite hydrogel capsules using fibrin hydrogel as the core and alginate-chitosan composite shell. These microbeads were differentiated into liver organoids with encapsulated hiPSCs and human liver cell lines (HepaRG). The formed liver organoids showed urea secretion and albumin secretion, suggesting a proper liver function in vitro [95]. Pajoumshariati et al. used a drop-based microfluidic system to control the size and shape of the droplets using alginate-Ca-EDTA complex with gelatin hydrogel. This microencapsulation system was used to encapsulate crypt and Peyer’s patch immune cells to model the gastrointestinal niche [96].

3.2.4. Bioprinting

Bioprinting is a unique tool, which can be used to pattern the living cell incorporated into biomimetic hydrogel matrices and other biophysical cues. The computer-aided patterning of the complex structures allows the formation of organoids with controlled cell-cell and cell-matrix interactions. The advancement of the 3D bioprinters creates a new era of printing, which can print different cell-encapsulated hydrogels according to their mechanical and biochemical requirements. These complex 3D co-cultures of customized geometries enable bioprinting as a tool for complex organoid formation, which has not yet been appropriately investigated. The imaging data via magnetic resonance imaging (MRI) or computed tomography (CT) can be directly used to develop the complex geometries of the desired tissue or organ using 3D modeling software [97]. Then, those generated models can be precisely positioned in an additive layer-by-layer approach to create 3D tissue or organs using an inkjet or extrusion-based printing. The current printing technology enables to generate a plethora of complex geometries that attempts to resemble native tissues; thus, their biological structure that mimics the structure and function of their native tissue or organ depends on the hydrogel-based bioinks, which contain the nutrients and biologically active sites (ligands, growth factors). Several types of natural and synthetic hydrogel materials have been extensively investigated in the last decade, and many numbers of reviews can be found elsewhere [59,98,99]. In this review, we are not going to discuss the hydrogel properties and their formulation since it is out of the scope of this paper. However, some of the significant studies related to 3D bioprintable hydrogels are discussed here.

3D bioprinting extensively depends on several natural hydrogel systems, including alginate [100102], collagen [103105], chitosan [106,107], gelatin [108110], hyaluronic acid [108,111], as well as synthetic hydrogel systems, such as gelatin methacrylate (GelMA) [112115], and PEG-based materials [116118]. The use of decellularized matrices has attracted increasing attention due to their ability to apply materials from the same tissue of interest, which provides well-matched compositional complexity and architectural fidelity between printed biological structure and the target tissue [119121]. Falguni et al., were able to develop decellularized ECM (dECM) bioinks from three different tissues, namely cartilage, heart, and adipose. The lyophilized ECM was subjected to gelation using acetic acid and pepsin, and the cell encapsulated gel was printed, as shown in Fig. 3(A). Also, polycaprolactone (PCL) was used to support the gel structure where the printing was done with dECM gel and PCL in an alternative layer design only with adipose and cartilage dECM [119]. GelMA and collagen were recently used as a hydrogel while powdered dECM and cells were encapsulated with the gel for liver and heart tissue regeneration study using a digital micromirror device (DMD) [121]. Duan et al., bioprinted an aortic valve conduit with encapsulation of sinus smooth muscle cells (SMCs) in the valve root and human aortic vascular interstitial cells (hAVICs) in the leaflet as shown in Fig. 3(B). The cells were encapsulated in alginate/gelatin hydrogel, and after 7 days, hydrogel showed 83.2% and 81.4% viability for hAVICs and SMCs, respectively [122]. The aortic valve was 3D modeled using the micro CT scan of the freshly harvest porcine aortic valve (Fig. 3B(i)). Also, using a 3D bioprinting approach, it is possible to recapitulate the complex trabecular structure of a whole organ. Hinton et al., were able to print a day five embryonic chick heart using a 3D generated optical image from confocal microscopy. The printed heart was then imaged using a multiphoton microscope to generate a cross-section of the structure, which showed the internal trabeculation similar to the actual chick heart [123]. However, the generation of functional heart from the 3D printed heart was not studied in this study, yet showed the ability to print complex structures using 3D printing. In another study, Schoneberg et al., were able to develop an in vitro blood vessel model using a drop-on-demand bioprinting technique. This vessel model consists of continuous endothelium intimating the tunica intima, an elastic smooth muscle layer, mimicking tunica media, and the surrounding fibrous and collagenous matrix of fibroblasts mimicking the tunica adventitia. The necessary biochemical and biomechanical functionality were provided through the series of natural hydrogel matrixes, including gelatin, collagen, and custom-made bioreactor to mimic the ECM microenvironment [124].

Few recent studies directly focused on organoid printing can be found in the literature. However, most of the studies demonstrated simple bioprinting rather than complex organ printing. Töper et al., demonstrated the 3D bioprinting of colonoid-colon organoids - encapsulated GelMA hydrogel. The crypts were isolated from bovine colon tissue, and the crypts were seeded into wells with Matrigel. GelMA supported the colonoid culture, and the encapsulated colonoid was viable and proliferative, while GelMA maintained structural integrity during the culture period [125]. In another two studies, mammary organoids were generated using simple cell bioprinting on top of the hydrogel layer. A decellularized mammary tissue was used as an ECM hydrogel in one study, and the common cancer cell line and mammary epithelial cells were bioprinted on the hydrogel [126]. In the second study, human mammary epithelial cells were printed on the Collagen type I hydrogel matrix [127]. Alternatively, 3D cell spheroids can also be used in 3D bioprinting [128]. The use of liver spheroids can protect the cells from shear stress exerted on the individual hepatocytes during the printing process.

Despite the current progress in 3D bioprinting, several challenges are still remaining, such as the ability to print in high resolution, the ability to make the vascular structure to support long-term cell viability, and controllable cell densities. The development of new hydrogel systems parallel to the printing technologies will provide the possibility to build organoids with controlled stem cell niches and spatiotemporal ECM with multiple tissues.

3.2.5. Vascularization

In real organs, hierarchically branched vascular networks are available around the organ, and therefore, all the cells are within a few hundred microns away from the capillaries, which facilitate sufficient oxygen and nutrients through diffusion. The formation of vascular networks in the larger organoid system is still a major challenge. The one strategy is a scaffold-based system, where the vascular network is generated through a 3D modeling process, such as bioprinting. The most investigated such system is the coaxial nozzle method, where hollow tubular type structures are printed using biomimetic, cell encapsulated hydrogel systems. Also, the advancement of the microfluidic systems allows the distribution of flow and mass transfer more consistently, supporting the multiple types of vascular cells in vitro to form vascular tissues.

Khademhosseini and the group investigated agarose, a polysaccharide, as a sacrificial material to bioprint hollow microchannels within a hydrogel construct. The cell-laden hydrogel (GelMA) contained SMCs and fibroblasts. This photocurable material later photocured into a stable structure, and agarose fibers were removed using a mild vacuum. These microchannels were further coated with endothelial cells to promote angiogenesis (Fig. 4(A and B)) [129]. However, this direct retraction of the sacrificial template compromised the structural integrity, and therefore, another alternative bioink, Pluronic 127, which can simply remove reducing the temperature to 4°C, was introduced [130]. Alternatively, the coaxial nozzle strategy or core/shell model has been developed by Ozbolt and the team to print vessel-like microchannels in the cell-laden structure [131]. This system is mainly used in a calcium alginate hydrogel system, where Ca2+ acts as a crosslinking agent for alginate. This system consists of a coaxial nozzle, where Ca2+ solution flows through the inner core nozzle and alginate hydrogel flow through the outer core. When the alginate crosslinks with Ca2+, it makes a hollow tubular structure Fig. 4(C and D). The vascular endothelial cells were incorporated into the Ca2+ solution, while SMCs were incorporated into the alginate solution to promote vascular tissue formation. This method successfully made microchannels in vitro in numerous recent studies, but the use of this technique in organoid formation has yet to be discovered further. Another printing technique, scaffold-free bioprinting, is also utilized to make vascular structures in vitro. Norotte et al., investigated the scaffold-free vascular construct using layer-by-layer printing of agarose rods as a non-adhering molding template. Human skin fibroblasts (HSF) and human umbilical vein smooth muscle cells (hUVSMCs) were aggregated into discreet units either as multicellular spheroids or as cylinders of controlled diameter and printed with agarose rods in an alternative pattern as shown in Fig. 4(E) [132]. However, all these strategies will need to be modified to integrate within the 3D macroscale organ or tissue construct. Apart from these techniques, scaffolds can be functionalized with proangiogenic biomolecules, such as platelet-derived growth factors (PDGFs), VEGFs, and basic fibroblast growth factors (bFGFs) for rapid generation of vascular networks or angiogenesis [133,134].

Fig. 4.

Fig. 4.

(A) Schematic representation of bioprinting of agarose template fibers and subsequent formation of microchannels via template micromolding; (i) A bioprinter equipped with a piston fitted inside a glass capillary aspirates the agarose (inset). After gelation in 4°C, agarose fibers are bioprinted at predefined locations; (ii) A hydrogel precursor is casted over the bioprinted mold and photocrosslinked; (iii) The template is removed from the surrounding photocrosslinked gel; (iv) Fully perfusable microchannels are formed; (B) Photographs of the bioprinted templates (green) enclosed in GelMA hydrogels and the respective microchannels perfused with a fluorescent microbead suspension (pink); (i) bioprinted templates in a GelMA hydrogel construct and (i-a) respective network after perfusion; (ii) 3d branching agarose templates embedded in a GelMA hydrogel construct and (ii-a) resulting 3d branching network [129]; (C) The experimental setup: (i) 3D model of the coaxial nozzle, (ii) cross-sectional view of the coaxial nozzle assembly model with fluid flow paths for hydrogel and crosslinker solutions; (D) (i) Perfusion of oxygenized cell type media, (ii) media flow with intentionally generated air bubbles showing flow [131]; (E) Bioprinting tubular structures with cellular cylinders; (i) Design template; (ii) Layer-by-layer deposition of agarose cylinders (stained here in blue for better visualization) and multicellular pig SMC cylinders; (iii) The bioprinter (see Materials and methods) outfitted with two vertically moving print heads; (iv) The printed construct; (v) Engineered pig SMC tubes of distinct diameters resulted after 3 days of post-printed fusion [132].

3.2.6. Organoids-on-a-chip systems

Organoids-on-a-chip is a combination of organoids and organ-on-a-chip models, which are fundamentally different approaches. Organoids are cell-derived miniature tissue construct that replicates the fundamentally same functional characteristics to their in vivo counterparts. On the other hand, the organ-on-a-chip model relies on the human-made constructs in which cells and their microenvironment are precisely controlled. However, the development of the perfect organoid system is still far from achieving. Therefore, researchers are now exploring the possibility of combining the best features of each approach [135]. The complexity of the stem cell microenvironment is the main barrier in organoid development. The control of the biochemical and biophysical microenvironment only from the self-formation of cell aggregates is not sufficient due to the lack of timely activated signaling pathways to cell fate specification, physical aggregation of different cell types, and mechanical forces that guide the process of self-organization. Organ-on-chip models are explored to achieve not only the microenvironmental control but also model the tissue-tissue and multiorgan interaction and reduce the variability. The development of the organ-on-chip system is a combination of all the previously discussed techniques, including microarray systems, bioreactors, 3D bioprinting, and vascularization.

Demers et al., were able to develop a microfluidic system capable of recreating the microenvironment of the diffusion-based patterning of the neural tube in a tightly regulated manner. This system contains a pair of microchannels that serve as a nutrient supplier and waste product remover of the cell chamber. The cell chamber was filled with Matrigel to provide a 3D culturing matrix for embryonic stem cells and also created a stable morphogen gradient via diffusion across the microchannels and the cell matrix [136]. In another study, iPSCs derived perfused human liver model was developed using cell aggregates encapsulated PEG microtissues. These microtissues were cultured in a range of flow rates by adopting the C-trap chip architecture that enabled robust loading with encapsulated organoids (cell aggregates). This microfluidic system enables an opportunity to query patient-specific liver response in vitro while maintaining the microenvironment [137]. The same microfluidic system was used to develop a human brain organoid-on-chip platform. The multicellular cell aggregates (or embryoid bodies (EB)) were formed using hiPSCs and then suspended in Matrigel. Those EB suspended Matrigel were then pipetted on hydrogel channels of the microfluidic chip. Due to the gelation of Matrigel, those cell suspensions were immobilized on the chip, and the microchannels were connected to the neural differentiation media channel, allowing the formation of brain organoids, as shown in Fig. 5(A). The sufficient nutrient exchange within the brain organoid enabled the formation of large and polarized neuroepithelial surrounding a fluid-filled cavity resembling the ventricular zone. This brain organoid-on-chip model successfully investigated the adverse effect of nicotine on cortical development [138].

Fig. 5.

Fig. 5.

(A) The conFiguration of the brain organoid-on-a-chip device and enlarged view of the procedures for brain organoid generation on a chip. The brain organoid-on-a-chip system was established for EBs culture, neural differentiation and formation of brain organoids by integrating 3D Matrigel and fluid flow [138]; (B) Development of vascularized HSEs. (i) Schematic description of the protocol to develop vHSEs; (ii) Two different vasculature patterns were used in our studies and generated using fluorescently tagged alginate. Scale bar: 600 μm [142]; (C) 3D convoluted renal proximal tubule on chip; (a) Schematic of a nephron highlighting the convoluted proximal tubule; (b,c) corresponding schematics and images of different steps in the fabrication of 3D convoluted, perfusable proximal tubules, in which a fugitive ink is first printed on a gelatin-fibrinogen extracellular matrix (ECM) [143].

Another vital aspect of organ-on-chip models is the ability to mimic perfusable blood vessels due to the recent advancement of 3D bioprinting techniques [139141]. Abaci et al., were able to develop human skin construct using a bioengineering approach, which allows printing spatially controlled perfusable 3D vascular network on a precast mold using sacrificial sodium alginate hydrogel, as shown in Fig. 5(B). The vascular network was perfused with endothelial cells (iPSC derived or human umbilical vein derived) while surrounding dermal fibroblasts. This model promoted and guided neovascularization during wound healing in mice [142]. This type of vasculature or microchannel formation is beneficial for the prolonged viability of thick organoids. Similarly, Homan et al., have reported a bioprinting method to create human renal proximal tubules in a perfusable tissue chip embedded in an ECM. As shown in Fig. 5(C), the perfusion chip gasket was first made using silicone ink, and then the ECM base layer was printed using a multi-material 3D printing head using gelatin/fibrinogen gel mixture. The proximal tubule structure was printed on base ECM using fugitive ink pluronic F127. This 3D tubule structure promoted the formation of the tissue-like epithelium with improved phenotypic and functional properties compared to the same cell (proximal tubule epithelial cells) grown in 2D controls [143].

The control of the biophysical microenvironment is also crucial for organoid development. For example, a developing embryo experiences different types of mechanical forces, including fluid shear stress and single-cell generated traction forces. Some mechanical forces are generated due to the coordinated mechanical activity of a large group of cells, such as fetal breathing movement and heart contraction. However, the generation of biomechanical control from cell aggregates or simple hydrogel systems is not possible as these forces act according to ECM signals and soluble morphogens. To address this issue, recent studies have demonstrated microengineered platforms that can actuate mechanical forces in organoids. Lind et al. were able to fabricate cardiac microphysiological devices that can provide continuous contractile stress within the microarchitecture, allowing the self-assembly of physiomimetic laminar cardiac tissue [144]. The multilayer cantilever consists of a base layer, embedded strain sensor, and iPSCs embedded tissue guiding layer supporting self-assembly. Lee et al., developed a stomach-on-chip model in which hPSCs were differentiated to form stomach organoids in the Matrigel matrix. A luminal flow media was introduced to mimic in vivo gastric function through the lumen using a peristaltic pump. The generated fluid flow through the inner compartment produced the rhythmic stretch and contraction to the organoid reminiscent of gastric motility [145]. Homan et al., recently developed a kidney organoid-on-chip platform as a proof-of-concept to Lee’s approach. Under the static condition, hPSCs-derived kidney organoid shows largely avascular and immature glomerular and tubular-like compartments. As a solution, Homan et al. developed a 3D printed millimeter-scale chamber to examine the vascularization and maturation of hPSCs derived kidney organoids, as shown in Fig. 6(A and B). Notably, high fluid flow stress (FFS) resulted in enhanced vasculature and increased maturity in their glomerular and tubular compartments, as shown in Fig. 6(C). Furthermore, concomitant morphogenesis of tubular epithelial cells and podocytes were also enhanced in a shear stress-dependent manner, suggesting the flow generated microenvironment as a pivotal contributor to structural and functional development of kidney organoid [146].

Fig. 6.

Fig. 6.

(A) Developing renal organoids are placed on an engineered ECM, housed within a perfusable millifluidic chip, and subjected to controlled FSS; (B) representative phase contrast images of entire organoids, scale bars: 50 μm; (C) Confocal 3D renderings for vascular markers in whole-mount organoids cultured under, (i) static engineered ECM, (ii) low-FSS, and (iii) high-FSS conditions, scale bars: 100 μm [146]; (D) (i) Schematic of micromolded collagen in the modified insert and below a side view of stamp using electron microscopy; (ii) Schematic showing the application of an additional gradient comprised of DAPT (gamma secretase inhibitor); (iii) fluorescence images of a polarized crypt-villus unit under the 3-growth factor gradient and opposing DAPT gradient. Mature enterocytes (red, ALP+) and proliferative cells (green, EdU+) are also marked; (iv) Immunofluorescence staining (Olfm4/KRT20) of in vitro human small intestinal tissues showing tissue polarity under the combine growth factor and DAPT gradient; scale bar: 100 mm [147].

The human small intestine epithelium consists of a distinct crypt and villus architecture, and migration and differentiation of cells on these two distinct cellular compartments are driven in part by biochemical gradient factors that specify the polarity of each cellular compartments. This unique architecture of the epithelium was fabricated using crosslinked collagen on the PTFE membrane with the help of PDMS stamps. A modified Transwell insert was used to establish the diffusion-based gradient across the collagen membrane when lower and upper parts were loaded with two different media. This system enables the growth of crypt-villus architecture, as shown in Fig. 6(D) [147]. By using bioengineering techniques, including microfluidics and bioreactor systems, it is possible to maintain the complex microenvironment with relevant biochemical and biophysical cues.

4. Model organoid systems

4.1. Brain organoids

The development of human brain embodies involves a high degree of coordination between the neural stem cells (NSCs) and the dynamic microenvironment in which they exist [34]. The expansion of the neuroepithelium to generate radial glial stem cells (RGS) is the beginning of human brain development [32]. The human brain is a complex structure consisting of different region-specific cell types and complex cellular arrangements. In most situations, part of the specific interest region of the brain is developed due to the complexity [159]. Also, hPSCs derived brain organoids are considered as a robust model to recapitulate the significant features of brain development at early stages and study the neurodevelopmental disorders [160,161]. Most of the studied brain organoids were generated in the range of micro to millimeter size, and Matrigel was used as a scaffold that can promote neuroepithelial bud expansion and organoids growth [98].

Bangley et al., developed a cerebral organoid model that primarily produces more ventral regions using ventralizing drug treatment to increase the ventral forebrain region. By fusing organoids of dorsal and ventral forebrain identities, a dorsal-ventral axis was generated. The fused ventral dorsal organoid exhibited the typical migration of human cortical interneurons from ventral into forebrain regions, as shown by inhibition of CXCR4, a known regulator of interneuron migration [159]. In another study, Lancaster et al., were able to demonstrate the arrangement of organ-like configuration by using a bioengineered construct. The synthetic polymer, poly(lactide-co-glycolide) copolymer (PLGA), microfiber filaments were used to construct a 3D model as a floating scaffold to generate elongated embryoid bodies (EB) as shown in Fig. 7(A). During the organoid self-assembly process, PLGA microfibers served as a scaffold that could maintain dense cell-cell contact and increase the surface-area-to-volume ratio. This system has displayed enhanced neuroectoderm formation and improved cortical development [162]. Zhu et al., demonstrated the use of calcium alginate hollow fiber system to develop brain organoid using iPSCs Matrigel cell aggregates, thereby further used for modeling prenatal alcohol exposure to explore the mechanisms underlying neural dysfunction [163]. Another natural hydrogel system, a sodium hyaluronan and chitosan polyelectrolyte complex termed Cell-Mate3D, was used to facilitate the cerebral organoid formation without having any additional neural induction component. The generated organoid showed evidence of early cortico-genesis and exhibited neural rosette and neural tube-like structure. Also, this model can be used as an in vitro disease model for adrenoleukodystrophy (ALD) or any other neurodevelopmental disease using patient-derived iPSCs [164].

Fig. 7.

Fig. 7.

(A) Brain organoid formation (i) Schematic of the method for generating engineered cerebral organoids (enCORs). Timeline and media used are shown at the bottom. (ii) hPSCs attach to and coat the PLGA microfilaments (left panel, arrowheads) as well as fibers derived from sea sponge (middle panel), whereas cellulose fibers with similar dimensions (right panel) fail to form elongated embryoid bodies at day 3 with H9 cells, and instead remain as clumps (arrows) only partially attached to the fibers. (iii) Bioengineered embryoid bodies from H9 cells at two time points, days 8 and 11, during neural induction, showing clearing along the edges and polarized neural ectoderm (arrows) [162]; (B) Alginate Supports HIO Survival In Vitro (i) Representative images of live (Calcein-AM, green) and dead (Ethidium-homodimer-1, red) staining of spheroids in alginate and Matrigel after 7 days in culture. Scale bar, 100 μm; (ii) Rheological characterization of alginate hydrogels. Data shown are the mean ± SD from n ≥ 3 gels per condition [171].

4.2. Intestinal organoids

The intestines, derived from definitive endoderm, are a vital organ that performs the function of digestion, nutrient absorption, and waste elimination, which are further divided into small and large intestines. The complex structure of the intestine consists of several functionally distinct areas, including the duodenum, jejunum, and ilium, and also its surface comprises a highly folded epithelium of villi, microvilli, and intestinal crypts [165,166]. The first-ever modern organoid system reported in the literature was developed by Hans Clevers’s lab based on the self-renewal epithelium of the small intestine. The Lgr5+ stem cell, which can undergo self-renewal of the epithelium, was first reported by this group and thereby presented the single sorted Lgr5+ stem cells that can also initiate the crypt-villus units in the intestine [167]. Recently, Serra et al., reported that the generation of intestinal organoids is not only limited to Lgr5+ cells but also depends on transient YAP1 activation. YAP1 requires to display transient cell-to-cell viability in localization that further initiates a Notch and DLL1 lateral inhibition, which guides the dedifferentiation of Paneth cell and subsequent crypt formation [168].

In another study, Watson et al., generated human intestinal organoid (HIO) in vitro from iPSCs that can engraft in vivo. To establish the in vivo model, the cells cultured in Matrigel were differentiated to form organoids for 35 days and then embedded in collagen type I and transplanted them under the kidney capsule of immunocompromised mice. These transplanted organoids had shown digestive functions similar to the HIO, as shown by permeability and peptide uptake studies [169]. Since ECM mechanical properties influence epithelial cell behavior, several studies are demonstrated that the use of hydrogel matrices can provide the desired mechanical microenvironment for HIO formation [170]. In that aspect, Capeling et al., were able to develop an alginate-based hydrogel ECM to support the growth and differentiation of HIOs. Five different concentrations of alginate (ranged from 0.5% to 4% (w/v)) were used, as it was reported that 1% alginate concentration provided the optimal hydrogel matrix, showing the same viability of HIOs in Matrigel as shown in Fig. 7(B) [171]. The storage modulus of Matrigel was very close to the 1% alginate, suggesting the significance of the mechanical microenvironment for HIO formation. Similarly, the PEG-4MAL synthetic hydrogel system was developed to match the storage modulus of Matrigel, which is further discussed in section 3.1 [52]. The extrinsic mechanical cues provided by chemically modified hydrogels have a significant role in hPSCs expansion, differentiation, and HIO formation.

4.3. Kidney organoids

The human kidney is a highly complex mesoderm organ containing up to 2 million epithelial neurons that regulate blood filtration. The functional kidney requires more than 20 distinct cell types, arranging in a spatial architecture, required for excretion and regulation of pH and electrolyte and fluid balance [23,172]. The human kidney can be generated from four types of progenitor population, including nephron progenitors, renal intestinal progenitors, ureteric epithelial progenitors, and endothelial progenitors [173]. Several studies have been demonstrated the ability to form kidney organoids from the differentiation of hPSCs [174,175]. The formation of kidney organoid through primitive posterior streak and intermediate mesoderm from differentiated human embryonic stem cells was reported using Matrigel-coated monolayer culture [176]. The same group studied the different methods to generate all four types of progenitors from hPSCs. Although both collecting duct and nephrons are originated from the intermediate mesoderm, they have a distinct temporospatial origin. Therefore, in another study, Takasato et al., developed kidney organoids that contained nephrons associated with a collecting duct network surrounded by endothelial cells and renal interstitium. Also, by carefully balancing the anterior-posterior patterning of intermediate mesoderm using small molecules such as growth factors, this multicellular kidney organoid was generated (Fig. 8(A)) [23]. This same group developed a method to form all types of progenitors from hPSCs [173]. Apart from the Matrigel, the fibrinogen/gelatin hydrogel system was used to precisely control the ECM microenvironment [143]. In another study, silk has demonstrated the capability of forming kidney organoids using hPSCs, while vascular endothelial growth factors promoted blood vessel formation [177].

Fig. 8.

Fig. 8.

(A) (i) Global bright field observations of self-organizing kidney organoids across a time series. The success rate of organoid differentiation was 94.2%; scale bars: 1 mm. (ii) Tile scan immunofluorescence of a whole kidney organoid displaying structural complexity; scale bar: 1 mm. (iii) High-power immunofluorescence microscopy showing a nephron segmented into 4 compartments, including the collecting duct (CD, GATA3+ECAD+), distal tubule (DT, GATA3ECAD+LTL), proximal tubule (PT, ECADLTL+) and the glomerulus (G, WT1+). Scale bar, 100 μm [15]. (B) (i) Schematic representation of in vitro liver bud formation; (ii) Self-organization of three-dimensional human iPSC-LBs in co-cultures of human iPSC-HEs with HUVECs and human MSCs. The time-lapse fluorescence imaging of human iPSC-HEs is also shown here; scale bar: 5 mm [184].

4.4. Liver organoids

The liver is an essential organ that regulates many metabolic functions, including the production of hormones, regulation of cholesterol and glucose storage, and the synthesis of some plasma proteins; and also, the liver regulates the detoxication of the body. So, liver injuries can generate dramatic consequences, leading to hepatic pathologies such as liver fibrosis, hepatitis, cirrhosis, or hepatocarcinoma. Fortunately, the liver has some unique potential to maintain its homeostasis by generating healthy tissue because of the proliferation potential of mature hepatocytes. However, due to the severity of the injuries, sometimes the proliferative capacity of the hepatocytes is not sufficient enough to regenerate the lost tissue [178,179]. Since the liver regulates the detoxication of the body, it is easily susceptible to drug-induced liver failures. Two types of endothelial cells: the hepatocytes and biliary epithelial cells (also known as cholangiocytes) are mainly responsible for liver formation. Apart from these cells, several other non-epithelial cell types, including stellate cells, stromal cells, Kupffer cells, and endothelial cells together generate the functional liver structure called the hepatic lobule [180,181]. Adult hepatic progenitor cells (HPCs) are considered a good source for hepatocytes and hepatocytes like cells compared to the popular ESCs or iPSCs. The HPCs possess several advantages over iPSCs, such as more efficient and less time-consuming differentiation, lower risk of spontaneous mutations, chromosomal rearrangements, and teratoma formation [182].

Broutier et al., developed a protocol to develop long-term expandable liver culture combined with Matrigel, acts as an ECM, and hepatocyte growth factors (HGF), epidermal growth factor (EGF), FGF, and Rspo1 using mouse liver tissues and induced progenitor cells for months [180]. Several studies focused on the development of vascularized liver organoids. For example, Baptista et al., were able to develop a scaffold that human liver cells could easily enter and proliferate to repopulate the decellularized liver scaffold. This decellularization process was preserved the macrovascular skeleton of the entire liver. The human fetal liver cells and endothelial cells were perfused through the vascular structure, and those cells were able to repopulate areas throughout the liver in vitro [183]. In another study, the vascularized and functional liver was generated from iPSCs by in vivo transplantation of in vitro derived liver buds, as shown in Fig. 8(B). Specified hepatic cells were self-organized into 3D liver buds by recapitulating organogenetic interactions between human endothelial and human MSCs. The iPSCs derived liver buds were transplanted into mice models, and human vasculature in liver buds became functional within 48 h by connecting to the host vessels [184]. Manipulated iPSCs showed great potential in future regenerative medicine.

Recently, Sorrentino et al., developed a chemically defined mechano-modulatory 3D culture system for basic research and regenerative medicine applications where PEG based synthetic hydrogel system provided niche for liver organoid culture [185]. In another study, cellulose nanofibril hydrogel system was used as an alternative to Matrigel in generation of liver organoids. The niche provided by the cellulose nanofibrils hydrogel system generated functional hepatocyte-like cells similar or superior to the Matrigel. The mechanical properties of cellulose nanofibrils further supported the differentiation of liver organoids [186].

5. Therapeutic potential of organoids

5.1. Drug development and toxicity screening

The current drug toxicity screening methods are heavily dependent on the traditional 2D cell cultures (monolayer) on plastic, which results in unacceptable failure rates [152,187,188]. The new investigational drugs that have gone through phase II and phase III clinical trials were failed to proceed into the next stage of development 66% and 30%, respectively [152]. The current toxicity assays are the main reason for this high failure rate and hence highlight the need for reliable toxicity assessment. Also, the current concept for in vitro-in vivo correlations does not address the drug toxicity mechanisms but emphasized drug dissolution and bioavailability equivalence [187]. Organoid systems demonstrate improved toxicity screening due to their ability to harness the organ-specific cellular/tissue organization, such as embryogenesis, organogenesis, and angiogenesis, and thereby overcome the challenges. Also, inflammatory and toxicity pathways of the organoids are more similar to the actual physiological conditions, whereas none of those activities are possible in cell mono-or-co-culture in 2D monolayers on hard plastic supports. In the current toxicity screening paradigm, after the initial 2D monolayer studies, animal models are the immediate next step for promising new drugs. However, there is a significant gap between organoid models or simplified 3D models (cell spheroids) and the effects seen in animal models due to the complete lack of correlation in the genomic response to acute inflammatory stress between human and murine models. Furthermore, this discrepancy is proved by the years of data indicating that 57% of the rodent studies failed to predict the actual human toxicity [189]. Despite the abundant knowledge on the implementation of organoid systems, organoid models in toxicity screening are still limited. This might be mainly due to the complexity of the functional organoid formation with the correct stem cell niche, as previously addressed in this review.

Patient-derived organoid models also play a significant role in developing personalized treatment regimes. The disease organoid model can be implemented through disease tissues/cells from disease-site biopsies, which deliver sufficient material to identify the mutations or phenotypic profiling to facilitate more customized treatments. Likewise, both healthy and diseased organoids generated from the patient enable the screening of new drug combinations that selectively targets the diseased tissues only, which further facilitates the more effective treatments with minimal side effects. Since most of the side effects of drugs can be attributed to acute liver toxicity, the liver/hepatic organoids can be successfully used for predicting in vivo liver toxicity of experimental drugs before moving to more expensive clinical trials [10,21]. The majority of drug toxicity screening studies were focused on tumor models because the organoid technology provided a unique platform to ongoing extensive cancer research works due to various reasons, including the ability to expand long term, modify genetically, and remain stable genetically and phenotypically.

Hou et al., developed a primary pancreatic organoid tumor model for high-throughput screening (HTS). The 3D pancreatic cell culture was developed using standard flat-bottom well plates with cell repellent surface, where 3D bioprinting technology that relies on the magnetic force was used to make 3D organoids on surface well plates using cell spheroids in the absence of exogenous ECM components. Nearly 3000 approved drugs were tested on these patient-derived organoids and their 2D monolayer cell culture counterparts [190]. According to the results, some of the approved drugs had shown cytotoxicity against 3D models over 2D culture, showing the efficacy of 3D models, as explained before. Recently, Saito et al., were able to find a couple of drugs, which suppressed the organoid derived from biliary tract carcinomas (BTCs). BTCs are one of the most aggressive malignancies and have shown a poor prognosis. Therefore, the team was able to develop in vitro organoids using cancer tissues obtained from a patient who has diagnosed with BTCs. The organoid derived from non-cancer tissues was also developed to find a drug with minimal side effects. Finally, they found that the antifungal drugs amorolfine and fenticonazole significantly suppressed organoid growth derived from BTCs with minimal toxicity to healthy organoid tissues [191]. Also, Yan et al., developed a human gastric cancer organoid biobank that comprises normal, dysplastic, cancer, and lymph node metastases, including detailed whole-exome and transcription analysis. The developed biobank organoid system from patient-derived tissues was tested for different drugs similar to the respective patients, and the results were promising as the tumor response of the patient and the cancer organoid have shown the same response [192]. It is evident that organoid cultures have the potential to predict in vivo drug response.

Liver organoid models also provide insight into drug toxicity and food toxicity studies. The identification of hepatotoxic drugs in the initial drug development phase would save millions of dollars. We look into more recent liver organoid models used for toxicity studies. Leite et al., were able to develop a human hepatic organoid model, which enables the identification of drug-induced liver fibrosis in vitro. The current models for liver fibrosis in vitro do not consider the role of hepatocyte injury, which triggers a plethora of events leading to the accumulation of ECM proteins like collagen known as fibrosis. The 3D spheroids cultured from hepatic stellate cells (HSCs) and HepaRG were grown into functional liver organoids. Interestingly, allyl alcohol, which was not identified as a hepatotoxic compound using HSC monocultures, showed indirect HSC activation that leads to fibrosis. Moreover, this liver organoid model identified the compound that is not hepatotoxic, but the presence of functional hepatocytes indirectly activates the HSC [193]. Apart from drug screening, some studies were focused on environmental liver toxicity due to heavy metals and pesticides. For example, Forsythe et al., were able to develop a liver organoid from HSCs. Using this model, hepatotoxicity values were obtained for different heavy metal compounds, including lead, mercury, thallium, and pesticide glyphosate. The 3D hepatic model showed more sensitive and accurate readings for toxicity values compared to the 2D HepG models [194]. Recently, Mun et al., were able to develop hPSC-derived hepatocyte-like liver organoids, which can be successfully used for drug screening and disease modeling similar to HSC derived liver organoids [195].

5.2. Organoid as a model to study diseases

The commitment of the organoids as a disease model for various therapeutic potentials is steadily being established. Compared to the traditional cell cultures of a single cell type, the organoid cultures have a clear advantage for disease modeling due to their ability to mimic pathologies at the organ level [9,196]. One of the caveats in the use of animal models when used for disease modeling is the interspecies differences, which can be addressed by using human-derived organoids. A wide variety of organoid models have already been developed that reproduce genetic diseases, cancer, or host-pathogen interactions. For example, one of the most cited studies in recent years was focused on the reproduction of pathological features in the human gastric organoid. The gastric organoid developed from human gastric corpus tissue was microinjected with Helicobacter pylori bacteria, and the resultant organoid successfully reproduced the signs of bacterial infection. This model plays an essential role in studying gastric pathologies in humans because the species-specific features of the stomach make current animal models unsuitable. Also, H. pylori infection in mice is usually mild gastritis and does not progress to ulceration or cancers as it does in humans [197]. In another study, the same type of human gastric organoid model was developed using the direct differentiation of hPSCs instead of gastric cells. After microinjection of H. pylori to the organoid, the progression of the infection mainly depends on the virulence factor CagA with the association of the c-Met receptor [22]. Together, these types of studies help to elucidate the mechanisms underlying human stomach development and diseases.

Another best example of the host-pathogen interaction is the ZIKV incorporated brain or cerebral organoid systems [9,196]. ZIKV has a robust teratogenic effect: infection in pregnant women is associated with microcephaly in newborns. The development of the human brain organoids by mimicking fetal brain formation has allowed us to discover the deficiencies caused by the ZIKV in the brain. Garcez et al., were able to use hiPSCs cultured as neural stem cells, neurospheres, and brain organoids to explore the consequences of ZIKV infection during neurogenesis [19,160,198]. The ZIKV targets the brain cells as it reduces the viability of neurospheres and brain organoids that further abrogates neurogenesis. Furthermore, the main phenotypic effect of ZIKV is premature differentiation of neural progenitors associated with centrosome perturbation, leading to progenitor depletion, disruption of the ventricular zone, and cortical thinning [199]. The effect of different ZIKV strains was also studied to understand the pathogenic activities of different forms [200], and these models were successfully used for anti-ZIKV drug development [201].

Recently, Boretto et al., were able to develop a disease model to capture endometrial diseases. Apart from these disease models, different cancer organoid models were extensively studied to understand the progression of the tumor, and their potential therapeutic procedures, including pancreatic [202], lung [203], liver [204], intestinal [205], and prostate [206] cancer models. The cancer organoids as a disease model for pre-clinical trials and drug screening were extensively reviewed elsewhere [207210], and therefore, we do not review further on this topic.

6. Future perspectives and limitations

Organoids are one of the most versatile and physiologically relevant models to study human diseases and drug toxicity screening. So far, the traditional cell biology principles, together with the novel bioengineering approaches, have evolved organoid into a new era of translational research platform instead of being a basic research tool. Most importantly, the development of miniature organ-like structures using available microengineering techniques eventually curtail the time to develop autologous tissue/organ for transplantation. Although the development of physiologically similar human organoid for transplantation is far from reality, Taguchi et al., succeeded in inducing metanephric nephron progenitors that capable of reconstructing 3D nephric tubules and glomeruli, the two components of critical kidney function using both human and mouse PSCs in adult mice, thereby prove the capability of kidney transplantation [211]. In this respect, kidney organoid shows enormous therapeutic potential since kidneys have the highest rate of end-stage organ failure, leading to the highest demand for transplant [11].

Most of the current organoid systems were primarily developed via exogenous signals, in which using numerous cell types and bioengineering approaches were able to create the sophisticated stem cell niche. Nonetheless, the targeted genome editing, primarily via CRISPR (clustered regularly interspaced short palindromic repeats) or TALEN (transcription activator-like effector nucleases) technology, enables the manipulation of endogenous genes in clinically relevant cells and organisms. This method can be used to enhance the microenvironment by reprogramming the internal decision-making component of cells. The reprogramming of the cells in organoids would widen the understanding of the organoid formation and function via self-regulation and self-assembly. Most importantly, this technique can be used to correct genetic defects in humans. The first proof-of-concept demonstration of genome editing in the organoid was done by Schwank et al., in which the genome editing of human stem cells in intestinal organoid was demonstrated via CRISPR/Cas9. The intestinal organoids isolated from two cystic fibrosis (CF) patients were used to correct the CF transmembrane conductor receptor (CFTR) mutation by deleting the phenylalanine at position 508. The corrected organoid showed the restoration of CFTR function in the swelling assay [212]. CRISPR/Cas9 genome editing has been recently used in different cancer organoid models [213,214] and kidney organoids [215]. The genome editing of the organoid systems would be an exciting area to explore further to develop treatments for genetic disorders, including CF, Alzheimer’s, and autism.

The preparation of organoid cultures is generally considered a tedious laboratory procedure because of the small size, fragility, and dynamic culture requirements. Therefore, organoids generated through routine procedures have shown substantial variability in size, functional capacity, structural organization, and gene expression. As a solution for this issue, microengineered systems are uniquely suited because of their ability to handle fluids and dynamic cell culture more precisely. For example, Au et al., developed a digital microfluidic platform capable of generating arrays of individually addressable, free-floating, 3D hydrogel-based hepatic organoids. The hepatic organoids were formed in a programmed manner by activating the electrodes in a defined sequence. This programmed actuation system was also used to analyze the acetaminophen-induced hepatotoxicity without manual intervention [152]. Another way of reducing organoid culture variability is by using biosensing elements in culture platforms to allow continuous uninterrupted in situ monitoring. The microfluidic system developed by Zhang et al., had shown the capability to monitor drug-induced organ toxicity in a dual-organ human liver-and-heart-on-a-chip platform. The microfluidics controlling breadboard consists of physical sensors for monitoring microenvironmental parameters (e.g., O2, pH, temperature), electrochemical immunobiosensors for measuring soluble biomarkers, and a miniature microscope for observation of the organoid morphologies [216].

Finally, the lack of vascularization is an apparent reason for the limited lifespan of organoids because of limitations in nutrient supply. This also affects the size of the organoid in which this limited growth affects their maturation. We have discussed various solutions from other emerging bioengineering approaches such as microfluidics, 3D bioprinting, and organ-on-chip models to address this issue. Fabrication of channels that can host endothelial cells and form perfusable vascular units is a promising solution for this issue [217]. However, the current vascularization approaches are mainly restricted to simplified cellular systems.

7. Conclusion

Organoids have the capacity to mimic the specific cellular functions, 3D architecture, and cell-type compositions similar to the actual organ and thereby hold great promise for a range of biological and biomedical applications. In the study of drug discovery, organoid cultures may reduce millions of dollars spent on unnecessary drug developments due to the variability in animal models and humans. Since the organoids can resemble actual human organs, the number of animal models and clinical trials can be reduced, thereby addressing some ethical issues. However, the path towards translating organoid technology into real-life preclinical and clinical applications is still considerably more challenging. The demonstrated results in organoids in drug discovery, personalized medicine, and gene therapy suggested that more widespread acceptance of organoid technology in these fields will become a reality in the coming decades.

The complexity of the cell microenvironment (niche) is still a significant challenge to be addressed in organoid technology. The development of the novel hydrogel system except Matrigel is crucial for regulating the self-organization organoids in a more controlled manner. Increasing the organoid’s life span, which is crucial to create mature and functional tissues that reach homeostasis, is a vital challenge to be addressed in the future. The current 3D bioprinting techniques, microfluidics, bioreactors, and organ-on-a-chip platforms will help facilitate biochemical and biomechanical cues necessary for self-assembly and the prolonged life span of the organoids. However, these techniques must also be improved to meet the complexity of organoid formation and culture. Overcoming these challenges will require a multidisciplinary approach, combining both cell biology and bioengineering.

Acknowledgments

The authors acknowledge funding support from the National Institutes of Health (R01DE023356) and the University of Toledo.

Abbreviations:

aSCs

Adult stem cells

ALD

Adrenoleukodystrophy

BTCs

Biliary tract carcinomas

CFTR

CF transmembrane conductor receptor

CRISPR

Clustered regularly interspaces short palindromic repeats

CT

Computed tomography

CF

Cystic fibrosis

dECM

Decellularized ECM

DMD

Digital micromirror device

EB

Embryoid bodie

EGF

Epidermal growth factors

ECM

Extracellular matrix

FGFs

Fibroblast growth factors

FFS

Fluid flow stress

GPC

Gas pressure chromatography

GelMA

Gelatin methacrylate

HPCs

Hepatic progenitor cells

HGF

Hepatocyte growth factors

HPLC

High-pressure liquid chromatography

HTS

High-throughput screening

hAVICs

Human aortic vascular interstitial cells

hDPSCs

Human dental pulp stem cells

HIO

Human intestinal organoids

hMECs

Human mammary epithelial cells

hNESCs

Human neuroepithelial stem cells

hPSCs

Human pluripotent stem cells

HSF

Human skin fibroblasts

hUVSMCs

Human umbilical vein smooth muscle cells

hUVECs

Human umbilical vein endothelial cells

HA

Hyaluronic acid

HIF

Hypoxia-inducible factor

iHep Cells

Induced hepatic cells

iPSCs

Induced pluripotent stem cells

LEM

Liver extracellular matrix

MRI

Magnetic resonance imaging

PEG-4MAL

Maleimide crosslinked four-arm PEG macromere

MSCs

Mesenchymal stem cells

MEMS

Micro-electromechanical systems

NSCs

Neural stem cells

PDGFs

Platelet-derived growth factors

PLGA

Poly (lactide-co-glycolide) copolymer

PESCs

Pluripotent embryonic stem cells

PSCs

Pluripotent stem cells

PCL

Polycaprolactone

Footnotes

Declaration of Competing Interest

The authors declare no conflict of interest

Data availability

Processed data can be obtained from the cited references.

References

  • [1].Clevers H, Review modeling development and disease with organoids, Cell 165 (2016) 1586–1597, 10.1016/j.cell.2016.05.082. [DOI] [PubMed] [Google Scholar]
  • [2].Simian M, Bissell MJ, Organoids : a historical perspective of thinking in three dimensions, J. Cell Biol 216 (2022) 31–40, 10.1083/jcb.201610056JCB, n.d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Ravi M, Paramesh V, Kaviya SR, Anuradha E, Paul Solomon FD, 3D cell culture systems: advantages and applications, J. Cell. Physiol 230 (2015) 16–26, 10.1002/jcp.24683. [DOI] [PubMed] [Google Scholar]
  • [4].Jensen C, Teng Y, Is it time to start transitioning from 2D to 3D cell culture? Front. Mol. Biosci 7 (2020) 1–15, 10.3389/fmolb.2020.00033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Fontoura JC, Viezzer C, dos Santos FG, Ligabue RA, Weinlich R, Puga RD, Antonow D, Severino P, Bonorino C, Comparison of 2D and 3D cell culture models for cell growth, gene expression and drug resistance, Mater. Sci. Eng. C 107 (2020), 110264, 10.1016/j.msec.2019.110264. [DOI] [PubMed] [Google Scholar]
  • [6].Shen H, Writer S, Organoids have opened avenues into investigating numerous diseases. But how well do they mimic the real thing ? Proc. Natl. Acad. Sci 115 (2018) 3507–3509, 10.1073/pnas.1803647115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Marsee A, Roos FJM, Verstegen MMA, Roos F, Verstegen M, Clevers H, Vallier L, Takebe T, Huch M, Peng WC, Forbes S, Lemaigre F, de Koning E, Gehart H, van der Laan L, Spee B, Boj S, Baptista P, Schneeberger K, Soroka C, Heim M, Nuciforo S, Zaret K, Saito Y, Lutolf M, Cardinale V, Simons B, van IJzendoorn S, Kamiya A, Chikada H, Wang S, Mun SJ, Son MJ, Onder TT, Boyer J, Sato T, Georgakopoulos N, Meneses A, Broutier L, Boulter L, Grün D, IJzermans J, Artegiani B, van Boxtel R, Kuijk E, Carpino G, Peltz G, Banales J, Man N, Aloia L, LaRusso N, George G, Rimland C, Yeoh G, Grappin-Botton A, Stange D, Prior N, Tirnitz-Parker JEE, Andersson E, Braconi C, Hannan N, Lu WY, Strom S, Sancho-Bru P, Ogawa S, Corbo V, Lancaster M, Hu H, Fuchs S, Hendriks D, Forbes SJ, van der Laan LJW, Building consensus on definition and nomenclature of hepatic, pancreatic, and biliary organoids, Cell Stem Cell 28 (2021) 816–832, 10.1016/j.stem.2021.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Vives J, Batlle-Morera L, The challenge of developing human 3D organoids into medicines, Stem Cell Res. Ther 11 (2020) 1–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Artegiani B, Clevers H, Use and application of 3D-organoid technology, Hum. Mol. Genet 27 (2018) 99–107, 10.1093/hmg/ddy187. [DOI] [PubMed] [Google Scholar]
  • [10].Fatehullah A, Tan SH, Barker N, Organoids as an in vitro model of human development and disease, Nat. Cell Biol 18 (2016) 246–254, 10.1038/ncb3312. [DOI] [PubMed] [Google Scholar]
  • [11].Lancaster MA, Knoblich JA, Organogenesis in a dish: modeling development and disease using organoid technologies, Science 345 (2014), 1247125, 10.1126/science.1247125. [DOI] [PubMed] [Google Scholar]
  • [12].Aloia L, McKie MA, Vernaz G, Cordero-Espinoza L, Aleksieva N, van den Ameele J, Antonica F, Font-Cunill B, Raven A, Aiese Cigliano R, Belenguer G, Mort RL, Brand AH, Zernicka-Goetz M, Forbes SJ, Miska EA, Huch M, Epigenetic remodelling licences adult cholangiocytes for organoid formation and liver regeneration, Nat. Cell Biol 21 (2019) 1321–1333, 10.1038/s41556-019-0402-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Sampaziotis F, Justin AW, Tysoe OC, Sawiak S, Godfrey EM, Upponi SS, Gieseck RL, De Brito MC, Berntsen NL, Gómez-Vázquez MJ, Ortmann D, Yiangou L, Ross A, Bargehr J, Bertero A, Zonneveld MCF, Pedersen MT, Pawlowski M, Valestrand L, Madrigal P, Georgakopoulos N, Pirmadjid N, Skeldon GM, Casey J, Shu W, Materek PM, Snijders KE, Brown SE, Rimland CA, Simonic I, Davies SE, Jensen KB, Zilbauer M, Gelson WTH, Alexander GJ, Sinha S, Hannan NRF, Wynn TA, Karlsen TH, Melum E, Markaki AE, Saeb-Parsy K, Vallier L, Reconstruction of the mouse extrahepatic biliary tree using primary human extrahepatic cholangiocyte organoids, Nat. Med 23 (2017) 954–963, 10.1038/nm.4360. [DOI] [PubMed] [Google Scholar]
  • [14].Wang D, Wang J, Bai L, Pan H, Feng H, Clevers H, Zeng YA, Long-term expansion of pancreatic islet organoids from resident procr+ progenitors, Cell 180 (2020) 1198–1211, 10.1016/j.cell.2020.02.048, e19. [DOI] [PubMed] [Google Scholar]
  • [15].Palikuqi B, Nguyen DHT, Li G, Schreiner R, Pellegata AF, Liu Y, Redmond D, Geng F, Lin Y, Gómez-Salinero JM, Yokoyama M, Zumbo P, Zhang T, Kunar B, Witherspoon M, Han T, Tedeschi AM, Scottoni F, Lipkin SM, Dow L, Elemento O, Xiang JZ, Shido K, Spence JR, Zhou QJ, Schwartz RE, De Coppi P, Rabbany SY, Rafii S, Adaptable haemodynamic endothelial cells for organogenesis and tumorigenesis, Nature 585 (2020) 426–432, 10.1038/s41586-020-2712-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Wu F, Wu D, Ren Y, Huang Y, Feng B, Zhao N, Zhang T, Chen X, Chen S, Xu A, Generation of hepatobiliary organoids from human induced pluripotent stem cells, J. Hepatol 70 (2019) 1145–1158, 10.1016/j.jhep.2018.12.028. [DOI] [PubMed] [Google Scholar]
  • [17].Ouchi R, Togo S, Kimura M, Shinozawa T, Koido M, Koike H, Thompson W, Karns RA, Mayhew CN, McGrath PS, McCauley HA, Zhang RR, Lewis K, Hakozaki S, Ferguson A, Saiki N, Yoneyama Y, Takeuchi I, Mabuchi Y, Akazawa C, Yoshikawa HY, Wells JM, Takebe T, Modeling Steatohepatitis in Humans with Pluripotent Stem Cell-Derived Organoids, Cell Metab 30 (2019) 374–384, 10.1016/j.cmet.2019.05.007, e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Tanimizu N, Ichinohe N, Sasaki Y, Itoh T, Sudo R, Yamaguchi T, Katsuda T, Ninomiya T, Tokino T, Ochiya T, Miyajima A, Mitaka T, Generation of functional liver organoids on combining hepatocytes and cholangiocytes with hepatobiliary connections ex vivo, Nat. Commun (2021) 12, 10.1038/s41467-021-23575-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Qian X, Nguyen HN, Song MM, Hadiono C, Ogden SC, Hammack C, Yao B, Hamersky GR, Jacob F, Zhong C, Yoon KJ, Jeang W, Lin L, Li Y, Thakor J, Berg DA, Zhang C, Kang E, Chickering M, Nauen D, Ho CY, Wen Z, Christian KM, Shi PY, Maher BJ, Wu H, Jin P, Tang H, Song H, Ming GL, Brain-Region-Specific Organoids Using Mini-bioreactors for Modeling ZIKV Exposure, Cell 165 (2016) 1238–1254, 10.1016/j.cell.2016.04.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Wörsd örfer P, Dalda N, Kern A, Krüger S, Wagner N, Kwok CK, Henke E, Ergün S, Generation of complex human organoid models including vascular networks by incorporation of mesodermal progenitor cells, (2019) 1–13. doi: 10.1038/s41598-019-52204-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Drost J, Clevers H, Organoids in cancer research, Nat. Rev. Cancer 18 (2018) 407–418, 10.1038/s41568-018-0007-6. [DOI] [PubMed] [Google Scholar]
  • [22].McCracken KW, Catá EM, Crawford CM, Sinagoga KL, Schumacher M, Rockich BE, Tsai YH, Mayhew CN, Spence JR, Zavros Y, Wells JM, Modelling human development and disease in pluripotent stem-cell-derived gastric organoids, Nature 516 (2014) 400–404, 10.1038/nature13863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Takasato M, Er PX, Chiu HS, Maier B, Baillie GJ, Ferguson C, Parton RG, Wolvetang EJ, Roost MS, De Sousa Lopes SMC, Little MH, Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis, Nature 526 (2015) 564–568, 10.1038/nature15695. [DOI] [PubMed] [Google Scholar]
  • [24].Si-Tayeb K, Noto FK, Nagaoka M, Li J, Battle MA, Duris C, North PE, Dalton S, Duncan SA, Highly efficient generation of human hepatocyte-like cells from induced pluripotent stem cells, Hepatology 51 (2010) 297–305, 10.1002/hep.23354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Nguyen R, Da Won Bae S, Qiao L, George J, Developing liver organoids from induced pluripotent stem cells (iPSCs): An alternative source of organoid generation for liver cancer research, Cancer Lett 508 (2021) 13–17, 10.1016/j.canlet.2021.03.017. [DOI] [PubMed] [Google Scholar]
  • [26].Rao S, Hossain T, Mahmoudi T, 3D human liver organoids: An in vitro platform to investigate HBV infection, replication and liver tumorigenesis, Cancer Lett 506 (2021) 35–44, 10.1016/j.canlet.2021.02.024. [DOI] [PubMed] [Google Scholar]
  • [27].Longmire TA, Ikonomou L, Hawkins F, Christodoulou C, Cao Y, Jean JC, Kwok LW, Mou H, Rajagopal J, Shen SS, Dowton AA, Serra M, Weiss DJ, Green MD, Snoeck HW, Ramirez MI, Kotton DN, Efficient derivation of purified lung and thyroid progenitors from embryonic stem cells, Cell Stem Cell 10 (2012) 398–411, 10.1016/j.stem.2012.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].McCauley KB, Hawkins F, Serra M, Thomas DC, Jacob A, Kotton DN, Efficient derivation of functional human airway epithelium from pluripotent stem cells via temporal regulation of Wnt signaling, Cell Stem Cell 20 (2017) 844–857, 10.1016/j.stem.2017.03.001, e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Jacob A, Morley M, Hawkins F, McCauley KB, Jean JC, Heins H, Na CL, Weaver TE, Vedaie M, Hurley K, Hinds A, Russo SJ, Kook S, Zacharias W, Ochs M, Traber K, Quinton LJ, Crane A, Davis BR, White FV, Wambach J, Whitsett JA, Cole FS, Morrisey EE, Guttentag SH, Beers MF, Kotton DN, Differentiation of human pluripotent stem cells into functional lung alveolar epithelial cells, Cell Stem Cell 21 (2017) 472–488, 10.1016/j.stem.2017.08.014, e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].D’Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart NG, Moorman MA, Kroon E, Carpenter MK, Baetge EE, Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells, Nat. Biotechnol 24 (2006) 1392–1401, 10.1038/nbt1259. [DOI] [PubMed] [Google Scholar]
  • [31].Spence JR, Mayhew CN, Rankin SA, Kuhar MF, Vallance JE, Tolle K, Hoskins EE, Kalinichenko VV, Wells SI, Zorn AM, Shroyer NF, Wells JM, Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro, Nature 470 (2011) 105–110, 10.1038/nature09691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME, Homfray T, Penninger JM, Jackson AP, Knoblich JA, Cerebral organoids model human brain development and microcephaly, Nature 501 (2013) 373–379, 10.1038/nature12517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Dye BR, Hill DR, Ferguson MA, Tsai YH, Nagy MS, Dyal R, Wells JM, Mayhew CN, Nattiv R, Klein OD, White ES, Deutsch GH, Spence JR, In vitro generation of human pluripotent stem cell derived lung organoids, Elife 2015 (2015) 1–25, 10.7554/eLife.05098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Yin X, Mead BE, Safaee H, Langer R, Karp JM, Engineering stem cell organoids, Stem Cell 18 (2016) 25–38, 10.1016/j.stem.2015.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Gjorevski N, Sachs N, Manfrin A, Giger S, Bragina ME, Ordonez-Moran P, Clevers H, Lutolf MP, Designer matrices for intestinal stem cell and organoid culture, Letter (2016), 10.1038/nature20168, 000. [DOI] [PubMed] [Google Scholar]
  • [36].Dahmann C, Oates AC, Brand M, Boundary formation and maintenance in tissue development, Nat. Rev. Genet 12 (2011) 43–55, 10.1038/nrg2902. [DOI] [PubMed] [Google Scholar]
  • [37].Kiecker C, Lumsden A, Compartments and their boundaries in vertebrate brain development, Nat. Rev. Neurosci 6 (2005) 553–564, 10.1038/nrn1702. [DOI] [PubMed] [Google Scholar]
  • [38].Weiss P, Taylor AC, Reconstitution of complete organs from single-cell suspension of chick embryos in advanced stages of differentiation, Proc. Natl. Acad. Sci 46 (1960) 1177–1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Moscona A, Rotation-mediated histogenetic aggregation of dissociated cells. A quantifiable approach to cell interactions in vitro, Exp. Cell Res 22 (1961) 455–475, 10.1016/0014-4827(61)90122-7. [DOI] [PubMed] [Google Scholar]
  • [40].Xu C, Inokuma MS, Denham J, Golds K, Kundu P, Gold JD, Carpenter MK, Feeder-free growth of undifferentiated human embryonic stem cells, Nat. Biotechnol 19 (2001) 971–974, 10.1038/nbt1001-971. [DOI] [PubMed] [Google Scholar]
  • [41].Li L, Xie T, Stem cell niche: structure and function, Annu. Rev. Cell Dev. Biol 21 (2005) 605–631, 10.1146/annurev.cellbio.21.012704.131525. [DOI] [PubMed] [Google Scholar]
  • [42].Peerani R, Zandstra PW, Enabling stem cell therapies through synthetic stem cell-niche engineering, J. Clin. Invest 120 (2010) 60–70, 10.1172/JCI41158.60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Tan S, Barker N, Engineering the niche for stem cells, Growth Factors 31 (2013) 175–184, 10.3109/08977194.2013.859683. [DOI] [PubMed] [Google Scholar]
  • [44].Vazin T, Schaffer DV, Engineering strategies to emulate the stem cell niche, Trends Biotechnol 28 (2010) 117–124, 10.1016/j.tibtech.2009.11.008. [DOI] [PubMed] [Google Scholar]
  • [45].Nuttelman CR, Tripodi MC, Anseth KS, Dexamethasone-functionalized gels induce osteogenic differentiation of encapsulated hMSCs, J. Biomed. Mater. Res. -Part A 76 (2006) 183–195, 10.1002/jbm.a.30537. [DOI] [PubMed] [Google Scholar]
  • [46].Frantz C, Stewart KM, Weaver VM, The extracellular matrix at a glance, J. Cell Sci 123 (2010) 4195–4200, 10.1242/jcs.023820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Li YJ, Chung HC, Rodriguez RT, Firpo MT, Healy KE, Hydrogels as artificial matrices for human embryonic stem cell self-renewal, J. Biomed. Mater. Res. Part A 79 (2006) 963–973, 10.1002/jbm.a.30732. [DOI] [PubMed] [Google Scholar]
  • [48].Biggs MJP, Richards RG, Gadegaard N, Wilkinson CDW, Oreffo ROC, Dalby MJ, The use of nanoscale topography to modulate the dynamics of adhesion formation in primary osteoblasts and ERK/MAPK signalling in STRO-1+ enriched skeletal stem cells, Biomaterials 30 (2009) 5094–5103, 10.1016/j.biomaterials.2009.05.049. [DOI] [PubMed] [Google Scholar]
  • [49].Nelson CM, Jean RP, Tan JL, Liu WF, Sniadecki NJ, Spector AA, Chen CS, Emergent patterns of growth controlled by multicellular form and mechanics, Proc. Natl. Acad. Sci. U. S. A 102 (2005) 11594–11599, 10.1073/pnas.0502575102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Tiwari A, Tursky ML, Mushahary D, Wasnik S, Collier FM, Suma K, Kirkland MA, Pande G, Ex vivo expansion of haematopoietic stem/progenitor cells from human umbilical cord blood on acellular scaffolds prepared from MS-5 stromal cell line, J. Tissue Eng. Regen. Med 7 (2012) 871–883, 10.1002/term.1479. [DOI] [PubMed] [Google Scholar]
  • [51].Hughes CS, Postovit LM, Lajoie GA, Matrigel: a complex protein mixture required for optimal growth of cell culture, Proteomics 10 (2010) 1886–1890, 10.1002/pmic.200900758. [DOI] [PubMed] [Google Scholar]
  • [52].Cruz-acunña R, Quiroós M, Farkas AE, Dedhia PH, Huang S, Siuda D, Garcíahernández V, Miller AJ, Spence JR, Nusrat A, García AJ, Synthetic hydrogels for human intestinal organoid generation and colonic wound repair, Nat. Cell Biol (2017), 10.1038/ncb3632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Cruz-acuña R, Quirós M, Huang S, Siuda D, Spence JR, Nusrat A, García AJ, PEG-4MAL hydrogels for human organoid generation, culture, and in vivo delivery, Nat. Protoc 13 (2018) 1–18, 10.1038/s41596-018-0036-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Mithal A, Capilla A, Heinze D, Berical A, Villacorta-martin C, Vedaie M, Jacob A, Abo K, Szymaniak A, Peasley M, Stuffer A, Mahoney J, Kotton DN, Hawkins F, Mostoslavsky G, organoids from human induced pluripotent, Nat. Commun (2022), 10.1038/s41467-019-13916-6 n.d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Jabaji Z, Brinkley GJ, Khalil HA, Sears CM, Lei NY, Lewis M, Dunn JCY, Stelzner M, Type I Collagen as an Extracellular Matrix for the In Vitro Growth of Human Small Intestinal Epithelium, PLoS One 9 (2014), e107814, 10.1371/journal.pone.0107814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Lu Y, Zhang G, Shen C, Uygun K, Yarmush ML, Meng Q, A novel 3D liver organoid system for elucidation of hepatic glucose metabolism, Biotechnol. Bioeng 109 (2012) 595–604, 10.1002/bit.23349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Li X, Ootani A, Kuo C, An air–liquid interface culture system for 3D organoid culture of diverse primary gastrointestinal tissues, in: Ivanov AI (Ed.), Gastrointest. Physiol. Dis. Methods Protoc. Methods Mol. Biol, Humana Press, New York, NY, 2016, pp. 33–40, 10.1007/978-1-4939-3603-8. [DOI] [PubMed] [Google Scholar]
  • [58].Fisher SA, Tam RY, Shoichet MS, Tissue mimetics : engineered hydrogel matrices provide, Tissue Eng. Part A 20 (2014) 895–898, 10.1089/ten.tea.2013.0765. [DOI] [PubMed] [Google Scholar]
  • [59].Unagolla JM, Jayasuriya AC, Hydrogel-based 3D bioprinting: a comprehensive review on cell-laden hydrogels, bioink formulations, and future perspectives, Appl. Mater. Today 18 (2020), 100479, 10.1016/j.apmt.2019.100479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Phelps EA, Enemchukwu NO, Fiore VF, Sy JC, Murthy N, Sulchek TA, Barker TH, García AJ, Maleimide cross-linked bioactive PEG hydrogel exhibits improved reaction kinetics and cross-linking for cell encapsulation and in situ delivery, Adv. Mater 24 (2012) 64–70, 10.1002/adma.201103574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Phelps EA, Headen DM, Taylor WR, Thulé PM, García AJ, Biomaterials Vasculogenic bio-synthetic hydrogel for enhancement of pancreatic islet engraftment and function in type 1 diabetes, Biomaterials 34 (2013) 4602–4611, 10.1016/j.biomaterials.2013.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Tung Y-C, Hsiao AY, Allen SG, Torisawa Y, Ho M, Takayama S, High-throughput 3D spheroid culture and drug testing using a 384 hanging drop array, Analyst 136 (2011) 473–478, 10.1039/c0an00609b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Laschke MW, Menger MD, Life is 3D : boosting spheroid function for tissue engineering, Trends Biotechnol 35 (2017) 133–144, 10.1016/j.tibtech.2016.08.004. [DOI] [PubMed] [Google Scholar]
  • [64].Yoon HH, Bhang SH, Shin J, Shin J, Kim B-S, Enhanced cartilage formation via three-dimensional cell engineering of human adipose-derived stem cells, Tissue Eng. Part A 18 (2012) 1949–1956, 10.1089/ten.tea.2011.0647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Montani C, Steimberg N, Boniotti J, Biasiotto G, Zanella I, Diafera G, Biunno I, Caimi L, Mazzoleni G, Di D, Fibroblasts maintained in 3 dimensions show a better differentiation state and higher sensitivity to estrogens, Toxicol. Appl. Pharmacol 280 (2014) 421–433, 10.1016/j.taap.2014.08.021. [DOI] [PubMed] [Google Scholar]
  • [66].Hsiao AY, Tung Y, Kuo C, Mosadegh B, Bedenis R, Pienta KJ, Takayama S, Micro-ring structures stabilize microdroplets to enable long term spheroid culture in 384 hanging drop array plates, Biomed. Microdevices 14 (2012) 313–323, 10.1007/s10544-011-9608-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Ota H, Kodama T, Miki N, Rapid formation of size-controlled three dimensional hetero-cell aggregates using micro-rotation flow for spheroid study, Biomicrofluidics 5 (2011), 034105, 10.1063/1.3609969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Want AJ, Nienow AW, Hewitt CJ, Coopman K, Large-scale expansion and exploitation of pluripotent stem cells for regenerative medicine purposes : beyond the T flask, Regen. Med 7 (2012) 71–84, 10.2217/RME.11.101. [DOI] [PubMed] [Google Scholar]
  • [69].Truckenmüller R, Giselbrecht S, Rivron N, Gottwald E, Saile V, van den Berg A, Wessling M, van Blitterswijk CA, Thermoforming of film-based biomedical microdevices, Adv. Mater 23 (2011) 1311–1329, 10.1002/adma.201003538. [DOI] [PubMed] [Google Scholar]
  • [70].Napolitano AP, Chai P, Dean DM, Morgan JR, On micromolded nonadhesive hydrogels, Tissue Eng 13 (2007) 2087–2094, 10.1089/ten.2006.0190. [DOI] [PubMed] [Google Scholar]
  • [71].Rivron NC, Vrij EJ, Rouwkema J, Le S, Van Den Berg A, Tissue deformation spatially modulates VEGF signaling and angiogenesis, Proc. Natl. Acad. Sci 109 (2012) 6886–6891, 10.1073/pnas.1201626109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Lopa S, Piraino F, Kemp RJ, Di Caro C, Lovati AB, Di Giancamillo A, Moroni L, Peretti GM, Rasponi M, Fabrication of multi-well chips for spheroid cultures and implantable constructs through rapid prototyping techniques, Biotechnol. Bioeng 112 (2015) 1457–1471, 10.1002/bit.25557. [DOI] [PubMed] [Google Scholar]
  • [73].Kim J, Lim I, Joo HJ, Choi S, Choi J, Cui L, Im L, Hong SJ, Lim D, Sphere formation of adipose stem cell engineered by poly-2- hydroxyethyl methacrylate induces in vitro angiogenesis through fi broblast growth factor 2, Biochem. Biophys. Res. Commun 468 (2015) 372–379, 10.1016/j.bbrc.2015.10.083. [DOI] [PubMed] [Google Scholar]
  • [74].Fennema E, Rivron N, Rouwkema J, Van Blitterswijk C, De Boer J, Spheroid culture as a tool for creating 3D complex tissues, Trends Biotechnol 31 (2013) 108–115, 10.1016/j.tibtech.2012.12.003. [DOI] [PubMed] [Google Scholar]
  • [75].Phelan MA, Lelkes PI, Swaroop A, Mini and customized low-cost bioreactors for optimized high-throughput generation of tissue organoids, Stem Cell Investig. 5 (2018), 10.21037/sci.2018.09.06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Selden C, Fuller B, Role of bioreactor technology in tissue engineering for clinical use and therapeutic target design, Bioengineering 5 (2018) 1–10, 10.3390/bioengineering5020032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Sant S, Johnston PA, The production of 3D tumor spheroids for cancer drug discovery, Drug Discov. Today Technol 23 (2017) 27–36, 10.1016/j.ddtec.2017.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Van Winkle AP, Gates ID, Kallos MS, Mass transfer limitations in embryoid bodies during human embryonic stem cell differentiation, Cells Tissues Organs 196 (2012) 34–47, 10.1159/000330691. [DOI] [PubMed] [Google Scholar]
  • [79].McMurtrey RJ, Analytic models of oxygen and nutrient diffusion, metabolism dynamics, and architecture optimization in three-dimensional tissue constructs with applications and insights in cerebral organoids, Tissue Eng. - Part C Methods. 22 (2016) 221–249, 10.1089/ten.tec.2015.0375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Wieck MM, Schlieve CR, Thornton ME, Fowler KL, Isani M, Grant CN, Hilton AE, Hou X, Grubbs BH, Frey MR, Grikscheit TC, Prolonged absence of mechanoluminal stimulation in human intestine alters the transcriptome and intestinal stem cell niche, Cell. Mol. Gastroenterol. Hepatol 3 (2017) 367–388, 10.1016/j.jcmgh.2016.12.008, e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Rodrigues CAV, Fernandes TG, Diogo MM, da Silva CL, Cabral JMS, Stem cell cultivation in bioreactors, Biotechnol. Adv 29 (2011) 815–829, 10.1016/j.biotechadv.2011.06.009. [DOI] [PubMed] [Google Scholar]
  • [82].Martin I, Wendt D, Heberer M, The role of bioreactors in tissue engineering, Trends Biotechnol 22 (2004) 80–86, 10.1016/j.tibtech.2003.12.001. [DOI] [PubMed] [Google Scholar]
  • [83].DiStefano T, Chen HY, Panebianco C, Kaya KD, Brooks MJ, Gieser L, Morgan NY, Pohida T, Swaroop A, Accelerated and improved differentiation of retinal organoids from pluripotent stem cells in rotating-wall vessel bioreactors, Stem Cell Reports 10 (2018) 300–313, 10.1016/j.stemcr.2017.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Phelan MA, Gianforcaro AL, Gerstenhaber JA, Lelkes PI, An air bubble-isolating rotating wall vessel bioreactor for improved spheroid/organoid formation, Tissue Eng. - Part C Methods. 25 (2019) 479–488, 10.1089/ten.tec.2019.0088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Ovando-Roche P, West EL, Branch MJ, Sampson RD, Fernando M, Munro P, Georgiadis A, Rizzi M, Kloc M, Naeem A, Ribeiro J, Smith AJ, Gonzalez-Cordero A, Ali RR, Use of bioreactors for culturing human retinal organoids improves photoreceptor yields, Stem Cell Res. Ther 9 (2018) 1–14, 10.1186/s13287-018-0907-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Farzaneh Z, Abbasalizadeh S, Asghari-Vostikolaee MH, Alikhani M, Cabral JMS, Baharvand H, Dissolved oxygen concentration regulates human hepatic organoid formation from pluripotent stem cells in a fully controlled bioreactor, Biotechnol. Bioeng (2020) 1–18, 10.1002/bit.27521. [DOI] [PubMed] [Google Scholar]
  • [87].Qian X, Jacob F, Song MM, Nguyen HN, Song H, Ming GL, Generation of human brain region–specific organoids using a miniaturized spinning bioreactor, Nat. Protoc 13 (2018) 565–580, 10.1038/nprot.2017.152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Whitesides GM, The origins and the future of microfluidics, Nature 442 (2006) 368–373, 10.1038/nature05058. [DOI] [PubMed] [Google Scholar]
  • [89].Sackmann EK, Fulton AL, Beebe DJ, The present and future role of microfluidics in biomedical research, Nature 507 (2014) 181–189, 10.1038/nature13118. [DOI] [PubMed] [Google Scholar]
  • [90].Hou X, Zhang YS, De Santiago GT, Alvarez MM, Ribas J, Jonas SJ, Weiss PS, Andrews AM, Aizenberg J, Khademhosseini A, Interplay between materials and microfluidics, Nat. Rev. Mater 2 (2017), 10.1038/natrevmats.2017.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Jin Y, Kim J, Lee JS, Min S, Kim S, Ahn DH, Kim YG, Cho SW, Vascularized liver organoids generated using induced hepatic tissue and dynamic liver-specific microenvironment as a drug testing platform, Adv. Funct. Mater 28 (2018) 1–15, 10.1002/adfm.201801954. [DOI] [Google Scholar]
  • [92].Ao Z, Cai H, Havert DJ, Wu Z, Gong Z, Beggs JM, Mackie K, Guo F, One-stop microfluidic assembly of human brain organoids to model prenatal cannabis exposure, Anal. Chem 92 (2020) 4630–4638, 10.1021/acs.analchem.0c00205. [DOI] [PubMed] [Google Scholar]
  • [93].Jung DJ, Shin TH, Kim M, Sung CO, Jang SJ, Jeong GS, A one-stop microfluidic-based lung cancer organoid culture platform for testing drug sensitivity, Lab Chip 19 (2019) 2854–2865, 10.1039/c9lc00496c. [DOI] [PubMed] [Google Scholar]
  • [94].Schuster B, Junkin M, Kashaf SS, Romero-Calvo I, Kirby K, Matthews J, Weber CR, Rzhetsky A, White KP, Tay S, Automated microfluidic platform for dynamic and combinatorial drug screening of tumor organoids, Nat. Commun 11 (2020) 1–12, 10.1038/s41467-020-19058-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Wang Y, Liu H, Zhang M, Wang H, Chen W, Qin J, One-step synthesis of composite hydrogel capsules to support liver organoid generation from hiPSCs, Biomater. Sci 8 (2020) 5476–5488, 10.1039/d0bm01085e. [DOI] [PubMed] [Google Scholar]
  • [96].Pajoumshariati SR, Azizi M, Wesner D, Miller PG, Shuler ML, Abbaspourrad A, Microfluidic-based cell-embedded microgels using nonfluorinated oil as a model for the gastrointestinal niche, ACS Appl. Mater. Interfaces 10 (2018) 9235–9246, 10.1021/acsami.7b16916. [DOI] [PubMed] [Google Scholar]
  • [97].Ashammakhi N, Hasan A, Kaarela O, Byambaa B, Sheikhi A, Gaharwar AK, Khademhosseini A, Advancing frontiers in bone bioprinting, Adv. Healthc. Mater 8 (2019) 1–24, 10.1002/adhm.201801048. [DOI] [PubMed] [Google Scholar]
  • [98].Liu H, Wang Y, Cui K, Guo Y, Zhang X, Qin J, Advances in hydrogels in organoids and organs-on-a-chip, Adv. Mater 31 (2019), 1902042, 10.1002/adma.201902042. [DOI] [PubMed] [Google Scholar]
  • [99].Zhang YS, Yue K, Aleman J, Mollazadeh-Moghaddam K, Bakht SM, Yang J, Jia W, Dell’Erba V, Assawes P, Shin SR, Dokmeci MR, Oklu R, Khademhosseini A, 3D bioprinting for tissue and organ fabrication, Ann. Biomed. Eng 45 (2017) 148–163, 10.1007/s10439-016-1612-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Colosi C, Shin SR, Manoharan V, Massa S, Costantini M, Barbetta A, Dokmeci MR, Dentini M, Khademhosseini A, Microfluidic bioprinting of heterogeneous 3D tissue constructs using low-viscosity bioink, Adv. Mater 28 (2016) 677–684a, 10.1002/adma.201503310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Gao Q, He Y, zhong Fu J, Liu A, Ma L, Coaxial nozzle-assisted 3D bioprinting with built-in microchannels for nutrients delivery, Biomaterials 61 (2015) 203–215, 10.1016/j.biomaterials.2015.05.031. [DOI] [PubMed] [Google Scholar]
  • [102].Tabriz AG, Hermida MA, Leslie NR, Shu W, Three-dimensional bioprinting of complex cell laden alginate hydrogel structures Three-dimensional bioprinting of complex cell laden alginate hydrogel structures, Biofabrication 7 (2015) 45012. http://stacks.iop.org/1758-5090/7/i=4/a=045012. [DOI] [PubMed] [Google Scholar]
  • [103].Xing R, Liu K, Jiao T, Zhang N, Ma K, Zhang R, Zou Q, Ma G, Yan X, An injectable self-assembling collagen-gold hybrid hydrogel for combinatorial antitumor photothermal/photodynamic therapy, Adv. Mater 28 (2016) 3669–3676, 10.1002/adma.201600284. [DOI] [PubMed] [Google Scholar]
  • [104].Nicole D, Louis W, Tylar P, Joseph S, Caroline D, Sonya S, Stephen K, Lawrence JB, Correlating rheological properties and printability of collagen bioinks: the effects of riboflavin photocrosslinking and pH, Biofabrication 9 (2017) 34102. http://stacks.iop.org/1758-5090/9/i=3/a=034102. [DOI] [PubMed] [Google Scholar]
  • [105].Yanez M, Rincon J, Dones A, De Maria C, Gonzales R, Boland T, In vivo assessment of printed microvasculature in a bilayer skin graft to treat full-thickness wounds, Tissue Eng. Part A. 21 (2014) 224–233, 10.1089/ten.tea.2013.0561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Dong L, Wang SJ, Zhao XR, Zhu YF, Yu JK, 3D-printed poly (ϵ-caprolactone) scaffold integrated with cell-laden chitosan hydrogels for bone tissue engineering, Sci. Rep 7 (2017) 4–12, 10.1038/s41598-017-13838-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Huang J, Fu H, Wang Z, Meng Q, Liu S, Wang H, Zheng X, Dai J, Zhang Z, BMSCs-laden gelatin/sodium alginate/carboxymethyl chitosan hydrogel for 3D bioprinting, RSC Adv 6 (2016) 108423–108430, 10.1039/c6ra24231f. [DOI] [Google Scholar]
  • [108].Gaetani R, Feyen DAM, Verhage V, Slaats R, Messina E, Christman KL, Giacomello A, Doevendans PAFM, Sluijter JPG, Epicardial application of cardiac progenitor cells in a 3D-printed gelatin/hyaluronic acid patch preserves cardiac function after myocardial infarction, Biomaterials 61 (2015) 339–348, 10.1016/j.biomaterials.2015.05.005. [DOI] [PubMed] [Google Scholar]
  • [109].Rutz AL, Hyland KE, Jakus AE, Burghardt WR, Shah RN, A multimaterial bioink method for 3D printing tunable, cell-compatible hydrogels, Adv. Mater 27 (2015) 1607–1614, 10.1002/adma.201405076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Kang HW, Lee SJ, Ko IK, Kengla C, Yoo JJ, Atala A, A 3D bioprinting system to produce human-scale tissue constructs with structural integrity, Nat. Biotechnol 34 (2016) 312–319, 10.1038/nbt.3413. [DOI] [PubMed] [Google Scholar]
  • [111].Ouyang L, Highley CB, Rodell CB, Sun W, Burdick JA, 3D printing of shear-thinning hyaluronic acid hydrogels with secondary cross-linking, ACS Biomater. Sci. Eng 2 (2016) 1743–1751, 10.1021/acsbiomaterials.6b00158. [DOI] [PubMed] [Google Scholar]
  • [112].Bertassoni LE, Cardoso JC, Manoharan V, Cristino AL, Bhise NS, Araujo WA, Zorlutuna P, Vrana NE, Ghaemmaghami AM, Dokmeci MR, Khademhosseini A, Direct-write bioprinting of cell-laden methacrylated gelatin hydrogels, Biofabrication 6 (2014), 10.1088/1758-5082/6/2/024105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Nichol JW, Koshy ST, Bae H, Hwang CM, Yamanlar S, Khademhosseini A, Cell-laden microengineered gelatin methacrylate hydrogels, Biomaterials 31 (2010) 5536–5544, 10.1016/j.biomaterials.2010.03.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Visser J, Melchels FPW, Jeon JE, van Bussel EM, Kimpton LS, Byrne HM, Dhert WJA, Dalton PD, Hutmacher DW, Malda J, Reinforcement of hydrogels using three-dimensionally printed microfibres, Nat. Commun 6 (2015) 6933, 10.1038/ncomms7933. [DOI] [PubMed] [Google Scholar]
  • [115].Zhang YS, Arneri A, Bersini S, Shin SR, Zhu K, Goli-Malekabadi Z, Aleman J, Colosi C, Busignani F, Dell’Erba V, Bishop C, Shupe T, Demarchi D, Moretti M, Rasponi M, Dokmeci MR, Atala A, Khademhosseini A, Bioprinting 3D microfibrous scaffolds for engineering endothelialized myocardium and heart-on-a-chip, Biomaterials 110 (2016) 45–59, 10.1016/j.biomaterials.2016.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Jia W, Gungor-Ozkerim PS, Zhang YS, Yue K, Zhu K, Liu W, Pi Q, Byambaa B, Dokmeci MR, Shin SR, Khademhosseini A, Direct 3D bioprinting of perfusable vascular constructs using a blend bioink, Biomaterials 106 (2016) 58–68, 10.1016/j.biomaterials.2016.07.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Gao G, Schilling AF, Hubbell K, Yonezawa T, Truong D, Hong Y, Dai G, Cui X, Improved properties of bone and cartilage tissue from 3D inkjet-bioprinted human mesenchymal stem cells by simultaneous deposition and photocrosslinking in PEG-GelMA, Biotechnol. Lett 37 (2015) 2349–2355, 10.1007/s10529-015-1921-2. [DOI] [PubMed] [Google Scholar]
  • [118].Daniele MA, Adams AA, Naciri J, North SH, Ligler FS, Interpenetrating networks based on gelatin methacrylamide and PEG formed using concurrent thiol click chemistries for hydrogel tissue engineering scaffolds, Biomaterials 35 (2014) 1845–1856, 10.1016/j.biomaterials.2013.11.009. [DOI] [PubMed] [Google Scholar]
  • [119].Pati F, Jang J, Ha DH, Won Kim S, Rhie JW, Shim JH, Kim DH, Cho DW, Printing three-dimensional tissue analogues with decellularized extracellular matrix bioink, Nat. Commun 5 (2014) 3935, 10.1038/ncomms4935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Ma X, Yu C, Wang P, Xu W, Wan X, Lai CSE, Liu J, Koroleva-Maharajh A, Chen S, Rapid 3D bioprinting of decellularized extracellular matrix with regionally varied mechanical properties and biomimetic microarchitecture, Biomaterials 185 (2018) 310–321, 10.1016/j.biomaterials.2018.09.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Yu C, Ma X, Zhu W, Wang P, Miller KL, Stupin J, Koroleva-Maharajh A, Hairabedian A, Chen S, Scanningless and continuous 3D bioprinting of human tissues with decellularized extracellular matrix, Biomaterials 194 (2019) 1–13, 10.1016/j.biomaterials.2018.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Duan B, Hockaday LA, Kang KH, Butcher JT, 3D Bioprinting of heterogeneous aortic valve conduits with alginate/gelatin hydrogels, J. Biomed. Mater. Res. -Part A 101 A (2013) 1255–1264, 10.1002/jbm.a.34420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Hinton TJ, Jallerat Q, Palchesko RN, Park JH, Grodzicki MS, Shue HJ, Ramadan MH, Hudson AR, Feinberg AW, Three-dimensional printing of complex biological structures by freeform reversible embedding of suspended hydrogels, Sci. Adv (2015) 1, 10.1126/sciadv.1500758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Schöneberg J, De Lorenzi F, Theek B, Blaeser A, Rommel D, Kuehne AJC, Kießling F, Fischer H, Engineering biofunctional in vitro vessel models using a multilayer bioprinting technique, Sci. Rep 8 (2018) 1–13, 10.1038/s41598-018-28715-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Töpfer E, Pasotti A, Telopoulou A, Italiani P, Boraschi D, Ewart MA, Wilde C, Bovine colon organoids: From 3D bioprinting to cryopreserved multi-well screening platforms, Toxicol. Vitr (2019) 61, 10.1016/j.tiv.2019.104606. [DOI] [PubMed] [Google Scholar]
  • [126].Mollica PA, Booth-Creech EN, Reid JA, Zamponi M, Sullivan SM, Palmer XL, Sachs PC, Bruno RD, 3D bioprinted mammary organoids and tumoroids in human mammary derived ECM hydrogels, Acta Biomater 95 (2019) 201–213, 10.1016/j.actbio.2019.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Reid JA, Mollica PA, Bruno RD, Sachs PC, Consistent and reproducible cultures of large-scale 3D mammary epithelial structures using an accessible bioprinting platform, Breast Cancer Res 20 (2018) 1–13, 10.1186/s13058-018-1069-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Skardal A, Devarasetty M, Kang HW, Mead I, Bishop C, Shupe T, Lee SJ, Jackson J, Yoo J, Soker S, Atala A, A hydrogel bioink toolkit for mimicking native tissue biochemical and mechanical properties in bioprinted tissue constructs, Acta Biomater 25 (2015) 24–34, 10.1016/j.actbio.2015.07.030. [DOI] [PubMed] [Google Scholar]
  • [129].Bertassoni LE, Cecconi M, Manoharan V, Nikkhah M, Hjortnaes J, Cristino AL, Barabaschi G, Demarchi D, Dokmeci MR, Yang Y, Khademhosseini A, Hydrogel bioprinted microchannel networks for vascularization of tissue engineering constructs, Lab. Chip 14 (2014) 2202–2211, 10.1039/c4lc00030g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Kolesky DB, Truby RL, Gladman AS, Busbee TA, Homan KA, Lewis JA, 3D bioprinting of vascularized, heterogeneous cell-laden tissue constructs, Adv. Mater 26 (2014) 3124–3130, 10.1002/adma.201305506. [DOI] [PubMed] [Google Scholar]
  • [131].Zhang Y, Yu Y, Chen H, Ozbolat IT, Characterization of printable cellular micro-fluidic channels for tissue engineering, Biofabrication 5 (2013), 10.1088/1758-5082/5/2/025004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Norotte C, Marga FS, Niklason LE, Forgacs G, Scaffold-free vascular tissue engineering using bioprinting, Biomaterials 30 (2009) 5910–5917, 10.1016/j.biomaterials.2009.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Richardson TP, Peters MC, Ennett AB, Mooney DJ, Polymeric system for dual growth factor delivery nature biotechnology, Nat. Biotechnol 19 (2001) 1029–1034. [DOI] [PubMed] [Google Scholar]
  • [134].Zisch AH, Lutolf MP, Hubbell JA, Biopolymeric delivery matrices for angiogenic growth factors, Cardiovasc. Pathol 12 (2003) 295–310, 10.1016/S1054-8807(03)00089-9. [DOI] [PubMed] [Google Scholar]
  • [135].Park SE, Georgescu A, Huh D, Organoids-on-a-chip, Science (80-.) 364 (2019) 960–965, 10.1126/science.aaw7894ARTICLE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Demers CJ, Soundararajan P, Chennampally P, Cox GA, Briscoe J, Collins SD, Smith RL, Development-on-chip: In vitro neural tube patterning with a microfluidic device, Dev 143 (2016) 1884–1892, 10.1242/dev.126847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Schepers A, Li C, Chhabra A, Seney BT, Bhatia S, Engineering a perfusable 3D human liver platform from iPS cells, Lab Chip 16 (2016) 2644–2653, 10.1039/c6lc00598e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Wang Y, Wang L, Zhu Y, Qin J, Human brain organoid-on-a-chip to model prenatal nicotine exposure, Lab Chip 18 (2018) 851–860, 10.1039/c7lc01084b. [DOI] [PubMed] [Google Scholar]
  • [139].Sasmal P, Datta P, Wu Y, Ozbolat IT, 3D bioprinting for modelling vasculature, Microphysiol. Syst 2 (2018) 9, 10.21037/mps.2018.10.02. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Sarker MD, Naghieh S, Sharma NK, Chen X, 3D biofabrication of vascular networks for tissue regeneration: a report on recent advances, J. Pharm. Anal 8 (2018) 277–296, 10.1016/j.jpha.2018.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Fetah K, Tebon P, Goudie MJ, Eichenbaum J, Ren L, Barros N, Nasiri R, Ahadian S, Ashammakhi N, Dokmeci MR, Khademhosseini A, The emergence of 3D bioprinting in organ-on-chip systems, Prog. Biomed. Eng 1 (2019), 012001, 10.1088/2516-1091/ab23df. [DOI] [Google Scholar]
  • [142].Abaci HE, Guo Z, Coffman A, Gillette B, Lee WH, Sia SK, Christiano AM, Human skin constructs with spatially controlled vasculature using primary and iPSC-derived endothelial cells, Adv. Healthc. Mater 5 (2016) 1800–1807, 10.1002/adhm.201500936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Homan KA, Kolesky DB, Skylar-Scott MA, Herrmann J, Obuobi H, Moisan A, Lewis JA, Bioprinting of 3D convoluted renal proximal tubules on perfusable chips, Sci. Rep 6 (2016) 1–13, 10.1038/srep34845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Lind JU, Busbee TA, Valentine AD, Pasqualini FS, Yuan H, Yadid M, Park SJ, Kotikian A, Nesmith AP, Campbell PH, Vlassak JJ, Lewis JA, Parker KK, Instrumented cardiac microphysiological devices via multimaterial three-dimensional printing, Nat. Mater 16 (2017) 303–308, 10.1038/nmat4782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Lee KK, McCauley HA, Broda TR, Kofron MJ, Wells JM, Hong CI, Human stomach-on-a-chip with luminal flow and peristaltic-like motility, Lab Chip 18 (2018) 3079–3085, 10.1039/c8lc00910d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Homan KA, Gupta N, Kroll KT, Kolesky DB, Skylar-Scott M, Miyoshi T, Mau D, Valerius MT, Ferrante T, Bonventre JV, Lewis JA, Morizane R, Flow-enhanced vascularization and maturation of kidney organoids in vitro, Nat. Methods 16 (2019) 255–262, 10.1038/s41592-019-0325-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [147].Wang Y, Gunasekara DB, Reed MI, DiSalvo M, Bultman SJ, Sims CE, Magness ST, Allbritton NL, A microengineered collagen scaffold for generating a polarized crypt-villus architecture of human small intestinal epithelium, Biomaterials 128 (2017) 44–55, 10.1016/j.biomaterials.2017.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Quadrato G, Nguyen T, Macosko EZ, Sherwood JL, Yang SM, Berger DR, Maria N, Scholvin J, Goldman M, Kinney JP, Boyden ES, Lichtman JW, Williams ZM, McCarroll SA, Arlotta P, Cell diversity and network dynamics in photosensitive human brain organoids, Nature 545 (2017) 48–53, 10.1038/nature22047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Przepiorski A, Sander V, Tran T, Hollywood JA, Sorrenson B, Shih JH, Wolvetang EJ, McMahon AP, Holm TM, Davidson AJ, A simple bioreactor-based method to generate kidney organoids from pluripotent stem cells, Stem Cell Rep. 11 (2018) 470–484, 10.1016/j.stemcr.2018.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Ramachandran SD, Schirmer K, Münst B, Heinz S, Ghafoory S, Wölfl S, Simon-Keller K, Marx A, Oie CI, Ebert MP, Walles H, Braspenning J, Breitkopf-Heinlein K, In vitro generation of functional liver organoid-like structures using adult human cells, PLoS One 10 (2015) 1–14, 10.1371/journal.pone.0139345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Berger E, Magliaro C, Paczia N, Monzel AS, Antony P, Linster CL, Bolognin S, Ahluwalia A, Schwamborn JC, Millifluidic culture improves human midbrain organoid vitality and differentiation, Lab Chip 18 (2018) 3172–3183, 10.1039/c8lc00206a. [DOI] [PubMed] [Google Scholar]
  • [152].Au SH, Chamberlain MD, Mahesh S, Sefton V, Wheeler AR, Hepatic orgonoids for microfluidic drug screening, Lab Chip 14 (2014) 3290–3299, 10.1039/c4lc00531g. [DOI] [PubMed] [Google Scholar]
  • [153].Zhao H, Chen Y, Shao L, Xie M, Nie J, Qiu J, Zhao P, Ramezani H, Fu J, Ouyang H, He Y, Airflow-assisted 3D bioprinting of human heterogeneous microspheroidal organoids with microfluidic nozzle, Small 14 (2018) 1–7, 10.1002/smll.201802630. [DOI] [PubMed] [Google Scholar]
  • [154].Adine C, Ng KK, Rungarunlert S, Souza GR, Ferreira JN, Engineering innervated secretory epithelial organoids by magnetic three-dimensional bioprinting for stimulating epithelial growth in salivary glands, Biomaterials 180 (2018) 52–66, 10.1016/j.biomaterials.2018.06.011. [DOI] [PubMed] [Google Scholar]
  • [155].Chadwick M, Yang C, Liu L, Gamboa CM, Jara K, Lee H, Sabaawy HE, Rapid processing and drug evaluation in glioblastoma patient-derived organoid models with 4D bioprinted arrays, IScience 23 (2020), 101365, 10.1016/j.isci.2020.101365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Cui K, Wang Y, Zhu Y, Tao T, Yin F, Guo Y, Liu H, Li F, Wang P, Chen Y, Qin J, Neurodevelopmental impairment induced by prenatal valproic acid exposure shown with the human cortical organoid-on-a-chip model, Microsystems Nanoeng 6 (2020), 10.1038/s41378-020-0165-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Wang Y, Wang H, Deng P, Chen W, Guo Y, Tao T, Qin J, In situ differentiation and generation of functional liver organoids from human iPSCs in a 3D perfusable chip system, Lab Chip 18 (2018) 3606–3616, 10.1039/c8lc00869h. [DOI] [PubMed] [Google Scholar]
  • [158].Wang Y, Wang H, Deng P, Tao T, Liu H, Wu S, Chen W, Qin J, Modeling human nonalcoholic fatty liver disease (NAFLD) with an organoids-on-a-chip system, ACS Biomater. Sci. Eng 6 (2020) 5734–5743, 10.1021/acsbiomaterials.0c00682. [DOI] [PubMed] [Google Scholar]
  • [159].Bagley JA, Reumann D, Bian S, Lévi-Strauss J, Knoblich JA, Fused cerebral organoids model interactions between brain regions, Nat. Methods 14 (2017) 743–751, 10.1038/nmeth.4304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Garcez PP, Loiola EC, Da Costa RM, Higa LM, Trindade P, Delvecchio R, Nascimento JM, Brindeiro R, Tanuri A, Rehen SK, Zika virus: Zika virus impairs growth in human neurospheres and brain organoids, Science (80-.) 352 (2016) 816–818, 10.1126/science.aaf6116. [DOI] [PubMed] [Google Scholar]
  • [161].Tang H, Hammack C, Ogden SC, Wen Z, Qian X, Li Y, Yao B, Shin J, Zhang F, Lee EM, Christian KM, Didier RA, Jin P, Song H, Ming GL, Zika virus infects human cortical neural progenitors and attenuates their growth, Cell Stem Cell 18 (2016) 587–590, 10.1016/j.stem.2016.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Lancaster MA, Corsini NS, Wolfinger S, Gustafson EH, Phillips AW, Burkard TR, Otani T, Livesey FJ, Knoblich JA, Guided self-organization and cortical plate formation in human brain organoids, Nat. Biotechnol 35 (2017) 659–666, 10.1038/nbt.3906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Zhu Y, Wang L, Yin F, Yu Y, Wang Y, Shepard MJ, Zhuang Z, Qin J, Probing impaired neurogenesis in human brain organoids exposed to alcohol, Integr. Biol. (United Kingdom) 9 (2017) 968–978, 10.1039/c7ib00105c. [DOI] [PubMed] [Google Scholar]
  • [164].Lindborg BA, Brekke JH, Vegoe AL, Ulrich CB, Haider KT, Subramaniam S, Venhuizen SL, Eide CR, Orchard PJ, Chen W, Wang Q, Pelaez F, Scott CM, Kokkoli E, Keirstead SA, Dutton JR, Tolar J, O’Brien T, Rapid induction of cerebral organoids from human induced pluripotent stem cells using a chemically defined hydrogel and defined cell culture medium, Stem Cells Transl. Med 5 (2016) 970–979, 10.5966/sctm.2015-0305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Fair KL, Colquhoun J, Hannan NRF, Intestinal organoids for modelling intestinal development and disease, Philos. Trans. R. Soc. B Biol. Sci 373 (2018), 20170217, 10.1098/rstb.2017.0217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Shaw D, Gohil K, Basson MD, Intestinal mucosal atrophy and adaptation, World J. Gastroenterol 18 (2012) 6357–6375, 10.3748/wjg.v18.i44.6357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Sato T, Vries RG, Snippert HJ, Van De Wetering M, Barker N, Stange DE, Van Es JH, Abo A, Kujala P, Peters PJ, Clevers H, Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche, Nature 459 (2009) 262–265, 10.1038/nature07935. [DOI] [PubMed] [Google Scholar]
  • [168].Serra D, Mayr U, Boni A, Lukonin I, Rempfler M, Challet Meylan L, Stadler MB, Strnad P, Papasaikas P, Vischi D, Waldt A, Roma G, Liberali P, Self-organization and symmetry breaking in intestinal organoid development, Nature 569 (2019) 66–72, 10.1038/s41586-019-1146-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Watson CL, Mahe MM, Múnera J, Howell JC, Sundaram N, Poling HM, Schweitzer JI, Vallance JE, Mayhew CN, Sun Y, Grabowski G, Finkbeiner SR, Spence JR, Shroyer NF, Wells JM, Helmrath MA, An in vivo model of human small intestine using pluripotent stem cells, Nat. Med 20 (2014) 1310–1314, 10.1038/nm.3737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Enemchukwu NO, Cruz-Acuña R, Bongiorno T, Johnson CT, García JR, Sulchek T, García AJ, Synthetic matrices reveal contributions of ECM biophysical and biochemical properties to epithelial morphogenesis, J. Cell Biol 212 (2016) 113–124, 10.1083/jcb.201506055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Capeling MM, Czerwinski M, Huang S, Tsai YH, Wu A, Nagy MS, Juliar B, Sundaram N, Song Y, Han WM, Takayama S, Alsberg E, Garcia AJ, Helmrath M, Putnam AJ, Spence JR, Nonadhesive alginate hydrogels support growth of pluripotent stem cell-derived intestinal organoids, Stem Cell Rep. 12 (2019) 381–394, 10.1016/j.stemcr.2018.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].van den Berg CW, Ritsma L, Avramut MC, Wiersma LE, van den Berg BM, Leuning DG, Lievers E, Koning M, Vanslambrouck JM, Koster AJ, Howden SE, Takasato M, Little MH, Rabelink TJ, Renal subcapsular transplantation of PSC-derived kidney organoids induces neo-vasculogenesis and significant glomerular and tubular maturation in vivo, Stem Cell Rep. 10 (2018) 751–765, 10.1016/j.stemcr.2018.01.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Takasato M, Er PX, Chiu HS, Little MH, Generation of kidney organoids from human pluripotent stem cells, Nat. Protoc 11 (2016) 1681–1692, 10.1038/nprot.2016.098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Phipson B, Er PX, Combes AN, Forbes TA, Howden SE, Zappia L, Yen HJ, Lawlor KT, Hale LJ, Sun J, Wolvetang E, Takasato M, Oshlack A, Little MH, Evaluation of variability in human kidney organoids, Nat. Methods 16 (2019) 79–87, 10.1038/s41592-018-0253-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Morizane R, Bonventre JV, Kidney organoids: a translational journey, Trends Mol. Med 23 (2017) 246–263, 10.1016/j.molmed.2017.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].Takasato M, Er PX, Becroft M, Vanslambrouck JM, Stanley EG, Elefanty AG, Little MH, Directing human embryonic stem cell differentiation towards a renal lineage generates a self-organizing kidney, Nat. Cell Biol 16 (2014) 118–126, 10.1038/ncb2894. [DOI] [PubMed] [Google Scholar]
  • [177].Gupta AK, Coburn JM, Davis-Knowlton J, Kimmerling E, Kaplan DL, Oxburgh L, Scaffolding kidney organoids on silk, J. Tissue Eng. Regen. Med 13 (2019) 812–822, 10.1002/term.2830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Michalopoulos GK, Khan Z, Liver stem cells: experimental findings and implications for human liver disease, Gastroenterology 149 (2015) 876–882, 10.1053/j.gastro.2015.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Garnier D, Li R, Delbos F, Fourrier A, Collet C, Guguen-Guillouzo C, Chesné C, Nguyen TH, Expansion of human primary hepatocytes in vitro through their amplification as liver progenitors in a 3D organoid system, Sci. Rep 8 (2018) 1–10, 10.1038/s41598-018-26584-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Broutier L, Andersson-Rolf A, Hindley CJ, Boj SF, Clevers H, Koo BK, Huch M, Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation, Nat. Protoc 11 (2016) 1724–1743, 10.1038/nprot.2016.097. [DOI] [PubMed] [Google Scholar]
  • [181].Duncan AW, Dorrell C, Grompe M, Stem cells and liver regeneration, Gastroenterology 137 (2009) 466–481, 10.1053/j.gastro.2009.05.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Nantasanti S, De Bruin A, Rothuizen J, Penning LC, Schotanus BA, Concise review: organoids are a powerful tool for the study of liver disease and personalized treatment design in humans and animals, Stem Cells Transl. Med 5 (2016) 325–330, 10.5966/sctm.2013-0207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Baptista PM, Siddiqui MM, Lozier G, Rodriguez SR, Atala A, Soker S, The use of whole organ decellularization for the generation of a vascularized liver organoid, Hepatology 53 (2011) 604–617, 10.1002/hep.24067. [DOI] [PubMed] [Google Scholar]
  • [184].Takebe T, Sekine K, Enomura M, Koike H, Kimura M, Ogaeri T, Zhang RR, Ueno Y, Zheng YW, Koike N, Aoyama S, Adachi Y, Taniguchi H, Vascularized and functional human liver from an iPSC-derived organ bud transplant, Nature 499 (2013) 481–484, 10.1038/nature12271. [DOI] [PubMed] [Google Scholar]
  • [185].Sorrentino G, Rezakhani S, Yildiz E, Nuciforo S, Heim MH, Lutolf MP, Schoonjans K, Mechano-modulatory synthetic niches for liver organoid derivation, Nat. Commun 11 (2020) 1–10, 10.1038/s41467-020-17161-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Krüger M, Oosterhoff LA, van Wolferen ME, Schiele SA, Walther A, Geijsen N, De Laporte L, van der Laan LJW, Kock LM, Spee B, Cellulose nanofibril hydrogel promotes hepatic differentiation of human liver organoids, Adv. Healthc. Mater 9 (2020), 10.1002/adhm.201901658. [DOI] [PubMed] [Google Scholar]
  • [187].Astashkina A, Grainger DW, Critical analysis of 3-D organoid in vitro cell culture models for high-throughput drug candidate toxicity assessments, Adv. Drug Deliv. Rev 69–70 (2014) 1–18, 10.1016/j.addr.2014.02.008. [DOI] [PubMed] [Google Scholar]
  • [188].Kondo J, Inoue M, Application of cancer organoid model for drug screening and personalized therapy, Cells 8 (2019) 470, 10.3390/cells8050470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Ranga A, Gjorevski N, Lutolf MP, Drug discovery through stem cell-based organoid models, Adv. Drug Deliv. Rev 69–70 (2014) 19–28, 10.1016/j.addr.2014.02.006. [DOI] [PubMed] [Google Scholar]
  • [190].Hou S, Tiriac H, Sridharan BP, Scampavia L, Madoux F, Seldin J, Souza GR, Watson D, Tuveson D, Spicer TP, Advanced development of primary pancreatic organoid tumor models for high-throughput phenotypic drug screening, SLAS Discov. 23 (2018) 574–584, 10.1177/2472555218766842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Saito Y, Muramatsu T, Kanai Y, Ojima H, Sukeda A, Hiraoka N, Arai E, Sugiyama Y, Matsuzaki J, Uchida R, Yoshikawa N, Furukawa R, Saito H, Establishment of patient-derived organoids and drug screening for biliary tract carcinoma, Cell Rep 27 (2019) 1265–1276, 10.1016/j.celrep.2019.03.088. [DOI] [PubMed] [Google Scholar]
  • [192].Yan HHN, Siu HC, Law S, Ho SL, Yue SSK, Tsui WY, Chan D, Chan AS, Ma S, Lam KO, Bartfeld S, Man AHY, Lee BCH, Chan ASY, Wong JWH, Cheng PSW, Chan AKW, Zhang J, Shi J, Fan X, Kwong DLW, Mak TW, Yuen ST, Clevers H, Leung SY, A comprehensive human gastric cancer organoid biobank captures tumor subtype heterogeneity and enables therapeutic screening, Cell Stem Cell 23 (2018) 882–897, 10.1016/j.stem.2018.09.016. [DOI] [PubMed] [Google Scholar]
  • [193].Leite SB, Roosens T, El Taghdouini A, Mannaerts I, Smout AJ, Najimi M, Sokal E, Noor F, Chesne C, van Grunsven LA, Novel human hepatic organoid model enables testing of drug-induced liver fibrosis in vitro, Biomaterials 78 (2016) 1–10, 10.1016/j.biomaterials.2015.11.026. [DOI] [PubMed] [Google Scholar]
  • [194].Forsythe SD, Devarasetty M, Shupe T, Bishop C, Atala A, Soker S, Skardal A, Environmental toxin screening using human-derived 3D bioengineered liver and cardiac organoids, Front. Public Heal 6 (2018) 103, 10.3389/fpubh.2018.00103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Mun SJ, Ryu JS, Lee MO, Son YS, Oh SJ, Cho HS, Son MY, Kim DS, Kim SJ, Yoo HJ, Lee HJ, Kim J, Jung CR, Chung KS, Son MJ, Generation of expandable human pluripotent stem cell-derived hepatocyte-like liver organoids, J. Hepatol 71 (2019) 970–985, 10.1016/j.jhep.2019.06.030. [DOI] [PubMed] [Google Scholar]
  • [196].Rossi G, Manfrin A, Lutolf MP, Progress and potential in organoid research, Nat. Rev. Genet 19 (2018) 671–687, 10.1038/s41576-018-0051-9. [DOI] [PubMed] [Google Scholar]
  • [197].Bartfeld S, Bayram T, Van De Wetering M, Huch M, Begthel H, Kujala P, Vries R, Peters PJ, Clevers H, In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection, Gastroenterology 148 (2015) 126–136, 10.1053/j.gastro.2014.09.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Cugola FR, Fernandes IR, Russo FB, Freitas BC, Dias JLM, Guimarães KP, Benazzato C, Almeida N, Pignatari GC, Romero S, Polonio CM, Cunha I, Freitas CL, Brandaõ WN, Rossato C, Andrade DG, Faria DDP, Garcez AT, Buchpigel CA, Braconi CT, Mendes E, Sall AA, Zanotto PMDA, Peron JPS, Muotri AR, Beltrao-Braga PCBB, The Brazilian Zika virus strain causes birth defects in experimental models, Nature 534 (2016) 267–271, 10.1038/nature18296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Gabriel E, Ramani A, Karow U, Gottardo M, Natarajan K, Gooi LM, Goranci-Buzhala G, Krut O, Peters F, Nikolic M, Kuivanen S, Korhonen E, Smura T, Vapalahti O, Papantonis A, Schmidt-Chanasit J, Riparbelli M, Callaini G, Krönke M, Utermöhlen O, Gopalakrishnan J, Recent Zika virus isolates induce premature differentiation of neural progenitors in human brain organoids, Cell Stem Cell 20 (2017) 397–406, 10.1016/j.stem.2016.12.005, e5. [DOI] [PubMed] [Google Scholar]
  • [200].Rosa-Fernandes L, Cugola FR, Russo FB, Kawahara R, de Melo Freire CC, Leite PEC, Bassi Stern AC, Angeli CB, de Oliveira DBL, Melo SR, de A. Zanotto PM, Durigon EL, Larsen MR, Beltrão-Braga PCB, Palmisano G, Zika virus impairs neurogenesis and synaptogenesis pathways in human neural stem cells and neurons, Front. Cell. Neurosci 13 (2019) 1–16, 10.3389/fncel.2019.00064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [201].Xu M, Lee EM, Wen Z, Cheng Y, Huang WK, Qian X, Tcw J, Kouznetsova J, Ogden SC, Hammack C, Jacob F, Nguyen HN, Itkin M, Hanna C, Shinn P, Allen C, Michael SG, Simeonov A, Huang W, Christian KM, Goate A, Brennand KJ, Huang R, Xia M, Ming GL, Zheng W, Song H, Tang H, Identification of small-molecule inhibitors of Zika virus infection and induced neural cell death via a drug repurposing screen, Nat. Med 22 (2016) 1101–1107, 10.1038/nm.4184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [202].Huang L, Holtzinger A, Jagan I, Begora M, Lohse I, Ngai N, Nostro C, Wang R, Muthuswamy LB, Crawford HC, Arrowsmith C, Kalloger SE, Renouf DJ, Connor AA, Cleary S, Schaeffer DF, Roehrl M, Tsao MS, Gallinger S, Keller G, Muthuswamy SK, Ductal pancreatic cancer modeling and drug screening using human pluripotent stem cell- and patient-derived tumor organoids, Nat. Med 21 (2015) 1364–1371, 10.1038/nm.3973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [203].Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, Balgobind AV, Wind K, Gracanin A, Begthel H, Korving J, van Boxtel R, Duarte AA, Lelieveld D, van Hoeck A, Ernst RF, Blokzijl F, Nijman IJ, Hoogstraat M, van de Ven M, Egan DA, Zinzalla V, Moll J, Boj SF, Voest EE, Wessels L, van Diest PJ, Rottenberg S, Vries RGJ, Cuppen E, Clevers H, A living biobank of breast cancer organoids captures disease heterogeneity, Cell 172 (2018) 373–386, 10.1016/j.cell.2017.11.010. [DOI] [PubMed] [Google Scholar]
  • [204].Broutier L, Mastrogiovanni G, Verstegen MMA, Francies HE, Gavarró LM, Bradshaw CR, Allen GE, Arnes-Benito R, Sidorova O, Gaspersz MP, Georgakopoulos N, Koo BK, Dietmann S, Davies SE, Praseedom RK, Lieshout R, IJzermans JNM, Wigmore SJ, Saeb-Parsy K, Garnett MJ, Van Der Laan LJW, Huch M, Human primary liver cancer-derived organoid cultures for disease modeling and drug screening, Nat. Med 23 (2017) 1424–1435, 10.1038/nm.4438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernández-mateos J, Khan K, Lampis A, Eason K, Huntingford I, Burke R, Rata M, Koh D, Tunariu N, Collins D, Hulkki-wilson S, Ragulan C, Spiteri I, Moorcraft SY, Chau I, Rao S, Watkins D, Fotiadis N, Bali M, Darvish-damavandi M, Lote H, Eltahir Z, Smyth EC, Begum R, Clarke PA, Hahne JC, Dowsett M, De Bono J, Workman P, Sadanandam A, Fassan M, Sansom OJ, Eccles S, Starling N, Braconi C, Sottoriva A, Robinson SP, Cunningham D, Valeri N, Patient-derived organoids model treatment response of metastatic gastrointestinal cancers, Science (80-.) 359 (2018) 920–926, 10.1126/science.aao2774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [206].Puca L, Bareja R, Prandi D, Shaw R, Benelli M, Karthaus WR, Hess J, Sigouros M, Donoghue A, Kossai M, Gao D, Cyrta J, Sailer V, Vosoughi A, Pauli C, Churakova Y, Cheung C, Deonarine LD, McNary TJ, Rosati R, Tagawa ST, Nanus DM, Mosquera JM, Sawyers CL, Chen Y, Inghirami G, Rao RA, Grandori C, Elemento O, Sboner A, Demichelis F, Rubin MA, Beltran H, Patient derived organoids to model rare prostate cancer phenotypes, Nat. Commun 9 (2018), 10.1038/s41467-018-04495-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [207].Weeber F, Ooft SN, Dijkstra KK, Voest EE, Tumor organoids as a pre-clinical cancer model for drug discovery, Cell Chem. Biol 24 (2017) 1092–1100, 10.1016/j.chembiol.2017.06.012. [DOI] [PubMed] [Google Scholar]
  • [208].Nagle PW, Plukker JTM, Muijs CT, van Luijk P, Coppes RP, Patient-derived tumor organoids for prediction of cancer treatment response, Semin. Cancer Biol 53 (2018) 258–264, 10.1016/j.semcancer.2018.06.005. [DOI] [PubMed] [Google Scholar]
  • [209].Fan H, Demirci U, Chen P, Emerging organoid models: Leaping forward in cancer research, J. Hematol. Oncol 12 (2019) 142, 10.1186/s13045-019-0832-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [210].Tuveson D, Clevers H, Cancer modeling meets human organoid technology, Science (80-.) 364 (2019) 952–955, 10.1126/science.aaw6985. [DOI] [PubMed] [Google Scholar]
  • [211].Taguchi A, Kaku Y, Ohmori T, Sharmin S, Ogawa M, Sasaki H, Nishinakamura R, Redefining the in vivo origin of metanephric nephron progenitors enables generation of complex kidney structures from pluripotent stem cells, Cell Stem Cell 14 (2014) 53–67, 10.1016/j.stem.2013.11.010. [DOI] [PubMed] [Google Scholar]
  • [212].Schwank G, Koo BK, Sasselli V, Dekkers JF, Heo I, Demircan T, Sasaki N, Boymans S, Cuppen E, Van Der Ent CK, Nieuwenhuis EES, Beekman JM, Clevers H, Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients, Cell Stem Cell 13 (2013) 653–658, 10.1016/j.stem.2013.11.002. [DOI] [PubMed] [Google Scholar]
  • [213].Roper J, Tammela T, Cetinbas NM, Akkad A, Roghanian A, Rickelt S, Almeqdadi M, Wu K, Oberli MA, Sánchez-Rivera F, Park YK, Liang X, Eng G, Taylor MS, Azimi R, Kedrin D, Neupane R, Beyaz S, Sicinska ET, Suarez Y, Yoo J, Chen L, Zukerberg L, Katajisto P, Deshpande V, Bass AJ, Tsichlis PN, Lees J, Langer R, Hynes RO, Chen J, Bhutkar A, Jacks T, Yilmaz ÖH, In vivo genome editing and organoid transplantation models of colorectal cancer and metastasis, Nat. Biotechnol 35 (2017) 569–576, 10.1038/nbt.3836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Matano M, Date S, Shimokawa M, Takano A, Fujii M, Ohta Y, Watanabe T, Kanai T, Sato T, Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids, Nat. Med 21 (2015) 256–262, 10.1038/nm.3802. [DOI] [PubMed] [Google Scholar]
  • [215].Freedman BS, Brooks CR, Lam AQ, Fu H, Morizane R, Agrawal V, Saad AF, Li MK, Hughes MR, Vander Werff R, Peters DT, Lu J, Baccei A, Siedlecki AM, Valerius MT, Musunuru K, McNagny KM, Steinman TI, Zhou J, Lerou PH, Bonventre JV, Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids, Nat. Commun 6 (2015), 10.1038/ncomms9715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Zhang YS, Aleman J, Shin SR, Kilic T, Kim D, Shaegh SAM, Massa S, Riahi R, Chae S, Hu N, Avci H, Zhang W, Silvestri A, Nezhad AS, Manbohi A, De Ferrari F, Polini A, Calzone G, Shaikh N, Alerasool P, Budina E, Kang J, Bhise N, Ribas J, Pourmand A, Skardal A, Shupe T, Bishop CE, Dokmeci MR, Atala A, Khademhosseini A, Multisensor-integrated organs-on-chips platform for automated and continual in situ monitoring of organoid behaviors, Proc. Natl. Acad. Sci. U. S. A 114 (2017) E2293–E2302, 10.1073/pnas.1612906114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Miri AK, Khalilpour A, Cecen B, Maharjan S, Shin SR, Khademhosseini A, Multiscale bioprinting of vascularized models, Biomaterials 198 (2019) 204–216, 10.1016/j.biomaterials.2018.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Processed data can be obtained from the cited references.

RESOURCES