17
Chapter 40 Hepatic tissue engineering Amanda X. Chen 1,2, *, Arnav Chhabra 2,3,4, *, Heather E. Fleming 2,3 and Sangeeta N. Bhatia 2,3,4,5,6,7 1 Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States, 2 David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States, 3 Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, United States, 4 Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States, 5 Wyss Institute for Biologically Inspired Engineering, Boston, MA, United States, 6 Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, United States, 7 Howard Hughes Medical Institute, Chevy Chase, MD, United States Liver disease burden Fatal liver disease accounts for B2 million deaths annu- ally worldwide and has steadily increasing rates over the years [1]. Liver failure can be divided into three major categories: (1) acute liver failure (ALF) that presents as a rapid loss of liver function in patients without preexisting liver disease, (2) chronic liver disease due to metabolic dysfunction, and (3) chronic liver failure accompanied by tissue remodeling and scarring. ALF is a rare syndrome with an annual incidence of less than 10 cases per million people in the developed world. In the United States, B2000 cases of ALF are diagnosed each year [2]. It commonly develops in healthy adults in their 30s. Patients with ALF usually present with abnormal liver biochemistry, coagulopathy, and encepha- lopathy. The causes vary geographically. Damage due to drug exposure (e.g., acetaminophen) is the most common cause in the West, while in large parts of the East, viruses (e.g., Hepatitis A and E) are the most prominent cause of ALF [3]. Clinically, ALF can be subdivided based on the period of time between the appearance of jaundice and onset of hepatic encephalopathy. Data from O’Grady et al. proposed the following classification: hyperacute for periods between 0 and 7 days, acute for periods between 7 and 28 days, and subacute for periods between 4 and 12 weeks [4]. In hyperacute cases the cause is usually acet- aminophen toxicity or viral infection. Subacute cases that evolve slowly often result from idiosyncratic drug- induced liver injury (DILI). Even though patients with a subacute presentation have less coagulopathy and enceph- alopathy, paradoxically they have a consistently worse medical outcome than those with a more rapid onset of the disease [5]. Chronic liver disease develops on the background of a constant injurious insult, either resulting from a metabolic disorder or a number of etiologies that lead to widespread tissue remodeling and pathologic deposition of extracellu- lar matrix (ECM). Inborn liver-based errors of metabolism are life-threatening conditions caused by genetic defects in single enzymes or transporters and lead to blockade of a specific metabolic pathway. While they can be accom- panied by progressive fibrosis and cirrhosis, such as in the case of α1-antitrypsin ZZ deficiency, hemochromato- sis, Wilson’s disease, and hereditary tyrosinemia [6], the liver parenchyma often remains intact. Some examples of metabolic disorders with an intact parenchyma include hypercholesterolemia, CriglerNajjar syndrome, ornithine transcarbamylase deficiency, organic acidurias, and hyperoxaluria [7]. In a biopsy the lack of parenchymal destruction often leads to a delayed diagnosis, exposing the patient to sequelae. In all cases of liver disease the lack of FDA-approved noninvasive biomarkers makes it challenging to diagnose and treat liver diseases. Chronic liver disease occurs in the setting of nonalco- holic fatty liver disease (NAFLD). NAFLD is marked by hepatic steatosis and is related to the presence of meta- bolic syndrome in association with obesity, diabetes, and/ or arterial hypertension [8]. A subset of NAFLD patients * These authors contributed equally. 737 Principles of Tissue Engineering. DOI: https://doi.org/10.1016/B978-0-12-818422-6.00041-1 Copyright © 2020 Elsevier Inc. All rights reserved.

Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

Chapter 40

Hepatic tissue engineeringAmanda X. Chen1,2,*, Arnav Chhabra2,3,4,*, Heather E. Fleming2,3 and Sangeeta N. Bhatia2,3,4,5,6,7

1Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States, 2David H. Koch Institute for

Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States, 3Harvard-MIT Health Sciences and Technology,

Massachusetts Institute of Technology, Cambridge, MA, United States, 4Institute for Medical Engineering and Science, Massachusetts Institute of

Technology, Cambridge, MA, United States, 5Wyss Institute for Biologically Inspired Engineering, Boston, MA, United States, 6Department of

Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, United States, 7Howard Hughes Medical

Institute, Chevy Chase, MD, United States

Liver disease burden

Fatal liver disease accounts for B2 million deaths annu-

ally worldwide and has steadily increasing rates over the

years [1]. Liver failure can be divided into three major

categories: (1) acute liver failure (ALF) that presents as a

rapid loss of liver function in patients without preexisting

liver disease, (2) chronic liver disease due to metabolic

dysfunction, and (3) chronic liver failure accompanied by

tissue remodeling and scarring.

ALF is a rare syndrome with an annual incidence of

less than 10 cases per million people in the developed

world. In the United States, B2000 cases of ALF are

diagnosed each year [2]. It commonly develops in healthy

adults in their 30s. Patients with ALF usually present with

abnormal liver biochemistry, coagulopathy, and encepha-

lopathy. The causes vary geographically. Damage due to

drug exposure (e.g., acetaminophen) is the most common

cause in the West, while in large parts of the East, viruses

(e.g., Hepatitis A and E) are the most prominent cause of

ALF [3]. Clinically, ALF can be subdivided based on the

period of time between the appearance of jaundice and

onset of hepatic encephalopathy. Data from O’Grady

et al. proposed the following classification: hyperacute for

periods between 0 and 7 days, acute for periods between

7 and 28 days, and subacute for periods between 4 and 12

weeks [4]. In hyperacute cases the cause is usually acet-

aminophen toxicity or viral infection. Subacute cases that

evolve slowly often result from idiosyncratic drug-

induced liver injury (DILI). Even though patients with a

subacute presentation have less coagulopathy and enceph-

alopathy, paradoxically they have a consistently worse

medical outcome than those with a more rapid onset of

the disease [5].

Chronic liver disease develops on the background of a

constant injurious insult, either resulting from a metabolic

disorder or a number of etiologies that lead to widespread

tissue remodeling and pathologic deposition of extracellu-

lar matrix (ECM). Inborn liver-based errors of metabolism

are life-threatening conditions caused by genetic defects

in single enzymes or transporters and lead to blockade of

a specific metabolic pathway. While they can be accom-

panied by progressive fibrosis and cirrhosis, such as in

the case of α1-antitrypsin ZZ deficiency, hemochromato-

sis, Wilson’s disease, and hereditary tyrosinemia [6], the

liver parenchyma often remains intact. Some examples of

metabolic disorders with an intact parenchyma include

hypercholesterolemia, Crigler�Najjar syndrome, ornithine

transcarbamylase deficiency, organic acidurias, and

hyperoxaluria [7]. In a biopsy the lack of parenchymal

destruction often leads to a delayed diagnosis, exposing

the patient to sequelae. In all cases of liver disease the

lack of FDA-approved noninvasive biomarkers makes it

challenging to diagnose and treat liver diseases.

Chronic liver disease occurs in the setting of nonalco-

holic fatty liver disease (NAFLD). NAFLD is marked by

hepatic steatosis and is related to the presence of meta-

bolic syndrome in association with obesity, diabetes, and/

or arterial hypertension [8]. A subset of NAFLD patients

* These authors contributed equally.

737Principles of Tissue Engineering. DOI: https://doi.org/10.1016/B978-0-12-818422-6.00041-1

Copyright © 2020 Elsevier Inc. All rights reserved.

Page 2: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

will develop signs of nonalcoholic steatohepatitis

(NASH), a more severe condition associated with lobular

inflammation and hepatocellular ballooning and can lead

to fibrosis and cirrhosis [9]. In NAFLD the liver is unable

to utilize carbohydrates and fatty acids properly, leading

to toxic overaccumulation of lipid species. These metabo-

lites induce cellular stress, injury, and death, which pre-

dispose the liver to sequelae such as cirrhosis and

hepatocellular carcinoma [10].

In the United States the number of NAFLD cases is

projected to expand from 83.1 million in 2015 (B26% of

the population) to 100.9 million by 2030 (B28% of the

population) [11]. An increasing percentage of these cases

are projected to be classified as NASH, rising from 20%

to 27% of adults with NAFLD during this interval [12].

While diagnosing NASH at an early stage remains a chal-

lenge, multiplexed protease-activated nanosensors have

demonstrated utility in monitoring NASH progression and

treatment response in a 3,5-diethylcarbonyl-1,4-dihydro-

collidine model of fibrosis in mice. With further develop-

ment, these noninvasive readouts can be used to diagnose

disease.

Current state of liver therapies

In order to mitigate the clinical burden of liver disease,

several therapeutic strategies have been undertaken

(Fig. 40.1).

Extracorporeal liver support devices

Liver failure is associated with abnormal accumulation of

numerous endogenous substances such as bilirubin,

ammonia, free fatty acids, and proinflammatory cytokines

[13]. Extracorporeal liver support devices have been

developed to detoxify the blood and plasma in order to

bridge patients to liver transplantation (LT) or allow the

native liver to recover from injury. Artificial liver (AL)

devices use nonliving components for detoxification, such

as membrane separation or sorbents, to selectively remove

toxins but have limited clinical use because they do not

replace the synthetic and metabolic roles of the liver [13].

Bioartificial liver (BAL) devices, on the other hand, con-

tain a cell-housing bioreactor that aims to provide the

detoxification and synthetic functions of the liver and are

an ongoing topic of clinical investigation. Current ver-

sions are either based on hollow fiber cartridges [14�16]

or on perfused three-dimensional (3D) matrices [17].

BALs, just like other hepatocyte-based therapies, face

many challenges, such as the lack of readily available

functional cell sources and the loss of cell viability and

phenotype during the treatment process.

Biopharmaceuticals

In the setting of ALF, N-acetyl cysteine (NAC) is FDA-

approved to reduce the extent of liver injury after acet-

aminophen overdose [18]. In the setting of chronic liver

disease, however, most of the FDA-approved therapies

are for hepatitis A, B, and C. A detailed listing can be

found in Table 40.1. While a few treatments have shown

moderate efficacy, there are currently no biopharmaceuti-

cals that are approved for NAFLD, NASH, or cirrhosis.

Glitazones, for example, upregulate adiponectin, an adi-

pokine with antisteatogenic and insulin-sensitizing proper-

ties [19]. Vitamin E, an antioxidant, can prevent liver

injury by blocking apoptotic pathways and protecting

against oxidative stress [19]. Despite clinical studies of a

large number of therapeutic candidates, no single agent or

combination has shown improvement to liver-related mor-

bidity and mortality in patients with NASH. Until a drug

is FDA-approved for NASH indications, lifestyle modifi-

cations and optimizing metabolic risk factors are the best

medical-treatment options for these patients.

Liver transplantation

The first attempt at human LT took place at the University

of Colorado on March 1, 1963 but turned out to be unsuc-

cessful [20]. Based on the pioneering work of Thomas

Starzl, the first extended survival of a human recipient after

LT was achieved on July 23, 1967 with a 19-month-old

female patient with hepatocellular carcinoma who survived

FIGURE 40.1 Cell-based therapies for liver disease. Extracorporeal

devices perfuse patient’s blood or plasma through bioreactors.

Hepatocytes are transplanted directly or implanted onto scaffolds.

Genetically modified large animals can be used for xenotranplantation.

738 PART | ELEVEN Gastrointestinal system

Page 3: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

13 months before succumbing to metastatic disease [21].

After the initial success of the surgery, advancements were

made to improve donor organ quality, recipient selection,

operative and perioperative management, immunosuppres-

sion and infectious complications. These advancements have

made orthotopic LT the primary treatment for end-stage

liver disease and certain cancers. These transplants have

1-year patient survival rates over 80% [22]. However, many

challenges remain, including donor organ shortages,

recipients with more advanced disease at transplant, a grow-

ing need for retransplantation, and adverse effects associated

with long-term immunosuppression. To overcome a growing

imbalance between the supply and demand of donor livers,

transplant centers have developed strategies to expand the

donor pool. These strategies include live donor LT [23],

split-LT [24], and extended criteria for donor livers [25].

Despite all these efforts, the number of liver transplants has

not increased in the last decade.

TABLE 40.1 List of FDA-approved therapies for chronic liver diseases.

Drug

name

Year

approved

Indication(s) Mechanism

Heplisav-B 2017 Hepatitis B Combines hepatitis B surface antigen with a proprietary Toll-likereceptor 9 agonist to enhance the immune response

Mavyret 2017 HCV genotype 1�6 Fixed-dose combination of glecaprevir, an HCV NS3/4A proteaseinhibitor, and pibrentasvir, an HCV NS5A inhibitor

Vosevi 2017 Hepatitis C Fixed-dose combination of sofosbuvir, an HCV nucleotide analogNS5B polymerase inhibitor, velpatasvir, an HCV NS5A inhibitor, andvoxilaprevir, an HCV NS3/4A protease inhibitor

Ocaliva 2016 Primary biliary cholangitis FXR agonist

Zepatier 2016 HCV genotype 1 or 4 Fixed-dose combination product containing elbasvir, an HCV NS5Ainhibitor, and grazoprevir, an HCV NS3/4A protease inhibitor

Cholbam 2015 Bile acid synthesis andperoxisomal disorders

Primary bile acid synthesized from cholesterol in the liver

Daklinza 2015 HCV genotype 3 Inhibitor of NS5A, a nonstructural protein encoded by HCV

Technivie 2015 HCV genotype 4 Fixed-dose combination of ombitasvir, an HCV NS5A inhibitor,paritaprevir, an HCV NS3/4A protease inhibitor, and ritonavir, aCYP3A inhibitor

Olysio 2013 Hepatitis C Small molecule orally active inhibitor of the NS3/4A protease of HCV

Sovaldi 2013 Hepatitis C Inhibitor of the HCV NS5B RNA-dependent RNA polymerase

Incivek 2011 HCV genotype 1 Inhibitor of the HCV NS3/4A serine protease

Victrelis 2011 HCV genotype 1 Inhibitor of the HCV NS3 serine protease

Viread 2008 Hepatitis B Oral nucleotide analogue DNA polymerase inhibitor

Tyzeka 2006 Hepatitis B Inhibitor of HBV DNA polymerase

Baraclude 2005 Chronic hepatitis B withevidence of active viralreplication

Small-molecule guanosine nucleoside analog with selective activityagainst HBV polymerase

Hepsera 2002 Chronic hepatitis B withevidence of active viralreplication

Inhibitor of HBV DNA polymerase

Pegasys 2002 Chronic hepatitis C withcompensated liver disease

Binds to and activates human type 1 interferon receptors

Peg-intron 2001 Chronic hepatitis C Binds to and activates human type 1 interferon receptors

Ribavirin 2001 Chronic hepatitis C Synthetic nucleoside analog with antiviral activity

Twinrix 2001 Hepatitis A and B Recombinant vaccine

FXR, Farnesoid X receptor; HBV, hepatitis B virus; HCV, hepatitis C virus; NS3, nonstructural protein 3.

Hepatic tissue engineering Chapter | 40 739

Page 4: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

An alternative to human LT is xenotransplantation,

though it has been clinically intractable due to concerns

about immunological rejection and zoonotic pathogen

transfer. With the advent of accessible genetic engineer-

ing technologies to circumvent the aforementioned chal-

lenges, the breeding efficiency of animals can be

leveraged to mass-produce tissue for human organ trans-

plants. Niu et al. applied CRISPR-Cas9 to inactivate all

62 copies of porcine endogenous retroviruses, thus paving

the way for pig-to-human transplants [26]. Relatedly,

Langin et al. genetically engineered porcine heart xeno-

grafts and demonstrated long-term pig-to-baboon orthoto-

pic transplantation [27].

Hepatocyte transplantation

Given the several drawbacks of LTs, alternative strategies

have been pursued. A potential alternative to LT is alloge-

neic hepatocyte transplantation (HT). Transplanted cells

can provide the missing or impaired hepatic function once

engrafted. Given their synthetic and metabolic capabili-

ties, mature hepatocytes are the primary candidates for

liver cell transplantations. HT offers several advantages

over LT. It is less invasive and can be performed repeat-

edly to meet metabolic requirements. Furthermore, multi-

ple patients can be treated with a single dissociated donor

tissue, and harvested cells can be cryopreserved for later

use on an as-needed basis.

The first experimental attempt of HT was done in

1976 to treat an animal model for Crigler�Najjar syn-

drome type I [28]. Along with other observations, it led to

the first transplant of autologous hepatocytes in 10

patients with liver cirrhosis in 1992 in Japan [29]. Since

then, reports have been published on more than 100

patients with liver disease treated by HT worldwide [30].

Human HT has resulted in partial correction of a number

of liver diseases, including urea cycle disorders [31], fac-

tor VII deficiency [32], glycogen storage disease type 1

[33], infantile Refsum disease [34], phenylketonuria [35],

severe infantile oxalosis [36], and ALF [37]. HT faces

several limitations: limited supply of high-quality mature

hepatocytes, freeze�thaw damage due to cryopreserva-

tion, poor cellular engraftment (estimated to be from

0.1% to 0.3% of host liver mass in mice after infusion of

3%2 5% of the total recipient liver cells) [30], and allo-

geneic rejection.

Clinically, the most widely used administration route

for HT is through the portal vein or one of its branches.

Hepatocytes traverse the sinusoidal vasculature and create

transient occlusions. The occlusions lead to vascular per-

meabilization which allows transplanted cells to reach the

liver parenchyma [35]. The number of cells that are

injected intraportally and subsequently engraft is a func-

tion of portal pressure and liver architecture. Thus other

administration routes have been explored for patients with

cirrhosis who have high portal pressures due to fibrosis.

In animal studies, hepatocytes transplanted into the

spleen proliferate for extended periods of time and display

normal hepatic function. The spleen has been shown to be

well-suited for hepatocyte engraftment because it func-

tions as a vascular filter and provides an immediate blood

supply [30]. The peritoneal cavity represents an attractive

administration route as it is easily accessible and can

house a large number of cells. Due to cell number

requirements associated with metabolic compensation, it

has been used in patients with ALF [38]. As an alternative

to the portal vein, spleen and peritoneum, the lymph node

(LN) has also been shown to demonstrate engraftment of

donor hepatocytes [39,40]. While this strategy has not

been utilized in the clinic yet, the preclinical data is

promising.

Current clinical trials

Several pathways have been implicated in the biology and

pathogenesis of NAFLD development: insulin resistance,

lipotoxicity, oxidative stress, altered immune/cytokine/

mitochondrial functioning, and apoptosis. New therapeu-

tic modalities are being developed to target many of these

pathways. For a detailed overview of NAFLD-targeted

drugs that are currently in the clinical trial pipeline, please

refer to Younossi et al. [41].

In vitro models

To build high-fidelity cellular models and therapies, com-

ponents of the native liver microenvironment must be

incorporated (Fig. 40.2). The liver’s highly organized

structure is key to its role as a complex tissue supporting

myriad synthetic and metabolic functions. In addition to

hepatocytes the main parenchyma of the liver, there are

several nonparenchymal cell types such as liver sinusoidal

endothelial cells, Kupffer cells, cholangiocytes, and stel-

late cells. In each lobule of the liver an array of parallel

hepatocyte cords are sandwiched between the sinusoid,

carrying circulating blood, and the bile duct, carrying

hepatocyte-secreted bile acids. Notably, this arrangement

dictates a unique set of architecturally driven cell�cell

and cell�matrix cues, which gives rise to liver-specific

phenotypes. Gradients of physicochemical stimuli along

the sinusoid drive zonal phenotypes with disparate meta-

bolic and synthetic functional profiles [42]. Interrupting

the natural order of cell arrangement in the liver is

directly connected with diseases discussed in the “Liver

disease burden” section. In this chapter, we will primarily

focus on human platforms, which are biologically distinct

from animal-derived cell models of the liver that are

reviewed more in depth elsewhere [43].

740 PART | ELEVEN Gastrointestinal system

Page 5: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

Two-dimensional liver culture

Hepatocytes are responsible for more than 500 metabolic

and synthetic functions of the human body, often catego-

rized broadly as protein synthesis and secretion, detoxifi-

cation, bile synthesis, and nitrogen metabolism. Primary

hepatocytes quickly lose their phenotype and function

after a few days in traditional monolayer culture and

require a collagen-coated surface for adherence and sur-

vival [6]. In contrast, when primary human hepatocytes

(PHHs) are cultured between two layers of collagen gel

(i.e., sandwich culture configuration), they retain viability,

polarity and many axes of relevant metabolic and syn-

thetic function [44,45]. Guguen-Guillouzo et al. found

that a random coculture with a liver epithelial cell line

was sufficient to support hepatic albumin secretion, sug-

gesting the importance of heterotypic cell interactions for

long-term ex vivo culture [46]. Bale et al. demonstrated

that other nonparenchymal liver cells can better recapitu-

late hepatic response to inflammatory stimuli, through

higher order intercellular interactions captured in a multi-

cellular platform [47]. It was later discovered that a

micropatterned architecture consisting of hepatocyte-filled

islands surrounded by a nonbiomimetic cell type and

mouse fibroblasts can also stabilize hepatocytes, suggest-

ing the existence of conserved coculture signals across

species. These micropatterned cocultures (MPCCs) enable

the study of DILI and hepatotropic pathogen infection for

several weeks in vitro [48�50]. Furthermore, Davidson

et al. added hepatic stellate cells to the traditional MPCC

to create an in vitro model of NASH [51].

Despite the utility of two-dimensional (2D) hepatic

cultures in screening assays, a wealth of literature sug-

gests that they are dissimilar to hepatocytes in vivo.

Specifically, 2D formats, even with overlaid collagen

matrix, are more flattened than their native cuboidal

architecture. On a subcellular level, this translates to

major differences in cytoarchitecture, which is linked to

aberrant polarization and nonphysiological behavior

[52,53]. Griffith and Swartz have described the improved

presentation of relevant biochemical and mechanical cues

in 3D cultures, typically cell-laden hydrogels, compared

to traditional 2D cultures [54].

Three-dimensional liver constructs

Commonly, 3D hepatic cultures consist of primary cell or

induced pluripotent stem cell (iPSC)-derived spheroid and

organoid cultures, which are typically embedded in ECM-

based hydrogels [55]. Spheroids and organoids can be

manufactured using a variety of techniques, such as

microwell mold-based technologies, which offer a high

degree of composition and size control but are difficult to

scale. Spinning flasks and bioreactors can produce large

populations of spheroids, though they are typically non-

uniform in size and function. Bell et al. showed that pri-

mary human hepatic spheroids fabricated and cultured in

microwell plates serve as useful models of hepatotoxicity

and multiple liver pathologies [56].

Toward transplantable cell therapies, Stevens et al.

used microwell molds to create 3D hepatic spheroid

FIGURE 40.2 Advances in hepatic tissue engi-

neering. Traditional tissue culture approaches such as

the addition of extracellular matrix, soluble factors,

cocultivation with supporting cell types, hanging

drop, microwell molding, and nonadhesive surfaces

have enabled the early study of hepatocyte phenotype

in vitro in both 2D and 3D cultures. The advent of

technologies from disciplines such as chemical engi-

neering and electrical engineering has led to a new

level of control for hepatic tissue cultures, such as

micropatterning to template cell interactions, micro-

physiological systems to study the impact of bioac-

tive perfusate, polymeric biomaterials for

constructing 3D cell-laden grafts, perfusion technolo-

gies for decellularization/recellularization strategies,

and 3D printing for scalable engineering of cellular

grafts. 2D, Two-dimensional; 3D, three-dimensional.

Hepatic tissue engineering Chapter | 40 741

Page 6: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

cocultures of PHHs and fibroblasts, which can be embed-

ded in agarose, fibrin, or polyethylene glycol hydrogel

scaffolds. The resulting tissue constructs support hepatic

function in vitro and in vivo after ectopic transplantation

into the peritoneal cavity [57]. Furthermore, Stevens et al.

demonstrated that implanting a tissue seed, consisting

of hepatic aggregates and vascular cords, into an

FRNG [fumarylacetoacetate hydrolase-deficient (Fah2/2),

recombinase activating gene-deficient (Rag12/2), nonob-

ese diabetes (NOD), and interleukin-2 receptor γchain-deficient (Il2rγ-null)] mouse model of hereditary

tyrosinemia leads to a 50-fold expansion in serum human

albumin and formation of perfusable vessels after 80 days

of cycled exposure to regenerative stimuli [58]. Relatedly,

Takebe et al. constructed liver buds consisting of iPSC-

derived hepatocyte-like cells (HLCs), mesenchymal stem

cells, and human umbilical vein endothelial cells; mesen-

teric transplantation of these human liver buds resulted in

vascularization and rescued a lethal TK-NOG mouse

model of liver injury [59].

To create clinically viable engineered liver constructs,

cell sourcing (discussed further in the “Cell sourcing” sec-

tion), and clinical-scale manufacturing of organoids and

spheroids must be addressed. Toward this end, Takebe

et al. constructed a large-scale liver bud microwell culture

platform, enabling the formation of 108 liver buds [60].

Physiological microfluidic models of liver

Despite improvements to in vitro liver platforms as model

systems and implants, static cultures lack physiologically

relevant dynamic components. The native liver’s dynamic

physiology arises from blood circulation and multiorgan

cross talk. Thus to improve biological fidelity, many have

attempted to create microfluidic models of the liver [61]. By

leveraging techniques from the semiconductor manufactur-

ing industry such as soft lithography, groups have fabricated

microphysiological systems with preformed channels to

allow for perfusion of nutrients and to aid in waste removal

[62�67]. These so-called liver-on-chip platforms allow for

the study of biochemical and mechanical cues such as

growth factor gradients and shear stress. Lee et al. demon-

strated that the presence of human stellate cells and applica-

tion of shear via flow enabled the formation of stable hepatic

spheroids on chip [68]. Furthermore, by linking microfluidic

channels between multiple tissue models, multiorgan phe-

nomena such as drug metabolite toxicity and disease pro-

gression can be captured in vitro, which is not possible in

traditional cultures [69�71].

Controlling three-dimensional architecture and

cellular organization

Another approach to improving the functionality of

tissue-engineered constructs is to more closely mimic

in vivo microarchitecture by generating scaffolds with a

highly defined material and cellular architecture, which

would provide better control over the 3D environment at

the microscale.

A range of rapid prototyping and patterning strategies

have been developed for polymers using multiple modes

of assembly, including fabrication using heat, light, adhe-

sives, or molding, and these techniques have been exten-

sively reviewed elsewhere [72]. For example, 3D printing

with adhesives combined with particulate leaching has

been utilized to generate porous poly(lactic-co-glycolic

acid (PLGA) scaffolds for hepatocyte attachment [73],

and microstructured ceramic [74] and silicon scaffolds

[75,76] have been proposed as platforms for hepatocyte

culture. Furthermore, molding and microsyringe deposi-

tion have been demonstrated to be robust methods for fab-

ricating specified 3D PLGA structures toward the

integration into implantable systems [77].

Microfabrication techniques have similarly been

employed for the generation of patterned cellular hydrogel

constructs. For instance, microfluidic molding has been

used to form biological gels containing cells into various

patterns [78]. In addition, syringe deposition in conjunc-

tion with micropositioning was recently illustrated as a

means to generate patterned gelatin hydrogels containing

hepatocytes [79]. Patterning of synthetic hydrogel systems

has also recently been explored. Specifically, the photopo-

lymerization property of poly(ethylene glycol) (PEG)

hydrogels enables the adaptation of photolithographic

techniques to generate patterned hydrogel networks. In

this process, patterned masks printed on transparencies

act to localize the ultraviolet exposure of the prepolymer

solution and, thus, dictate the structure of the resultant

hydrogel. The major advantages of photolithography-

based techniques for patterning of hydrogel structures are

its simplicity and flexibility. Photopatterning has been

employed to surface pattern biological factors [80], pro-

duce hydrogel structures with a range of sizes and shapes

[81,82], as well as build multilayer cellular networks

[83,84]. Consequently, hydrogel photopatterning technol-

ogy is ideally suited for the regulation of scaffold archi-

tecture at the multiple length scales required for

implantable hepatocellular constructs. As a demonstration

of these capabilities, photopatterning of PEG hydrogels

was utilized to generate hepatocyte and fibroblast cocul-

ture hydrogels with a defined 3D branched network,

resulting in improved hepatocyte viability and functions

under perfusion [85]. More recently, a “bottom-up”

approach for fabricating multicellular tissue constructs

utilizing DNA-templated assembly of 3D cell-laden

hydrogel microtissues demonstrates robust patterning of

cellular hydrogel constructs containing numerous cell

types [86]. Also, the additional combination of photopat-

terning with dielectrophoresis-mediated cell patterning

enabled the construction of hepatocellular hydrogel

742 PART | ELEVEN Gastrointestinal system

Page 7: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

structures organized at the cellular scale. Overall, the abil-

ity to dictate scaffold architecture coupled with other

advances in scaffold material properties, chemistries, and

the incorporation of bioactive elements (discussed further

in the “Extracellular matrix for cell therapies” section)

will serve as the foundation for the future development of

improved tissue-engineered liver constructs that can be

customized spatially, physically, and chemically.

In vivo models

While there have been impressive advances in cell culture

models of the human liver, experimental animal models

still play an important role in the effort to engineer liver

therapies. Commonly performed surgeries such as bile

duct ligation and partial hepatectomy are experimentally

tractable models of acute liver injury, yet they are of little

clinical relevance. Drug-induced (e.g., carbon tetrachlo-

ride, acetaminophen, or thioacetamide) hepatotoxicity to

induce necrotic lesions is more recapitulative of human

pathophysiology, but the phenotype is difficult to repro-

duce [87]. In addition, modeling chronic liver injury in

animal models is problematic because they tend to rapidly

correct severe hepatic damage after a few days, which is

not representative of human disease progression and reso-

lution [88].

In order to model a human-like context for liver inju-

ries, it is necessary to develop improved, controlled mod-

els of human liver injury. Such animal models can be

useful for the evaluation of human liver biology and the

preclinical performance of candidate therapies and drugs

[89,90]. To accomplish this, human hepatocytes can be

transplanted orthotopically in immunocompromised mice

with no liver injury via the injection of a cell solution and

are useful for modeling human-specific drug metabolism,

liver injury, and hepatotropic infections. However, on

average, HT exhibits poor levels of engraftment

(B10%�30%) [91].

Transplanted hepatocytes have the ability to expand

preferentially if the host is compromised by injury or

genetic modification. The first genetically engineered

mouse model to demonstrate this was the Alb-uPA mouse

that carries a uroplasminogen activator under an albumin

promoter, causing liver injury and failure [92]. Aiming to

improve on the Alb-uPA system, a transgenic model of

hereditary tyrosinemia I was developed, in which a

genetic knockout of FAH leads to the hepatotoxic accu-

mulation of fumarylacetoacetate [93]. FAH knockout

mouse injury initiation and duration can be controlled

through the administration of a small molecule drug

[2-(2-nitro-3-trifluoro-methylbenzoyl)-1,3-cyclohexane-

dione] in the drinking water. Another inducible model

called TK-NOG, which expresses thymidine kinase

under an albumin promoter, causes hepatocyte ablation

following activation by ganciclovir treatment [94]. In

the AFC8 injury model, induction by the small mole-

cule AP20187 drives caspase-8-initiated apoptosis of

hepatocytes modified to express FK506 under an albu-

min promoter [95]. For all of the above models,

engraftment rates surpassing 70% have been observed.

A classical study in parabiotic rats in the 1960s

revealed that hepatic injury results in the expression of

systemic, soluble signals that have the potential to drive

liver regeneration [96]. Despite decades of research, the

complex signaling cascade driving liver regeneration is

still not well understood but has found utility in ectopic

humanized mouse models. Chronic liver injury often pre-

sents with high portal pressures, which can reduce

engraftment levels during an orthotopic transplantation.

Thus transplantation to ectopic sites is clinically attrac-

tive. Ectopic grafts that anastomose to the host vascula-

ture can interact with regenerative stimuli from the host

liver, causing expansion and proliferation of the trans-

planted human hepatocytes. Initially, ectopic transplanta-

tion was demonstrated in the LN [40] and spleen [97],

and later in the subcutaneous space and mesenteric fat

pad, both of which offer ease of accessibility for manipu-

lation and noninvasive imaging [58,59,98].

Taken together, the range of liver injury animal mod-

els are an essential tool for studying various perturbations

to normal liver biology and building implantable tissue

constructs to address acute and chronic liver failure. The

field is just beginning to uncover mechanisms that control

liver regeneration in various disease and injury contexts.

The discovery of new soluble regenerative signals will be

central to advancing therapies that have the potential to

improve the supply of donor tissue.

Cell sourcing

Cell number requirements

The development of cell-based therapies poses myriad

challenges, partially stemming from the scale of the liver.

An adult human liver is estimated to possess 241 billion

hepatocytes, 24 billion stellate cells, and 96 billion

Kupffer cells [99]. Sourcing such enormous cell numbers

using current technologies is not feasible.

However, many human HT studies suggest that clini-

cal intervention is possible with a fewer number of cells

and offer critical insights to help us determine minimum

cell numbers. In a review, Fisher and Strom cataloged 78

different human HT studies, detailing both the input cell

number and a qualitative description of functionality [30].

Correlating these, we can broadly surmise that to correct

inborn errors of metabolism, at least 1�10 billion hepato-

cytes are needed. However, for ALF, that number grows

to 5�20 billion cells. For liver cirrhosis, HTs have largely

been unsuccessful (discussed further in the “Hepatocyte

transplantation” section); therefore it is unclear what the

Hepatic tissue engineering Chapter | 40 743

Page 8: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

cellular requirements for cirrhosis are. While injection of

hepatocytes is not the same as implantation within a scaf-

fold, these studies serve as useful inputs into more com-

plex physiological models.

In order to get us closer to these numbers, many dif-

ferent cell sources have been explored.

Immortalized cell lines

Immortalized hepatocyte cell lines can be derived from

liver tumor tissue or directly from primary hepatocytes

in vitro. The prominent lines utilized today are HepG2,

derived from hepatocellular carcinoma; HepaRG [100], a

human bipotential progenitor cell line; C3A, derived from

HepG2s; and Huh7, derived from liver tumor [101].

Several other fetal and adult hepatic cell lines have also

been established, typically using a combination of viral

oncogenes and the human telomerase reverse transcriptase

protein [102]. However, these cell lines lack the full func-

tional capacity of primary adult hepatocytes and there is a

risk that oncogenic factors could be transmitted to the

patient, limiting their use as a cell source for transplanta-

tion therapies.

Primary cells

Unlike immortalized lines, PHHs can provide a whole

host of human liver-specific function. PHHs, however, are

limited in supply, and their phenotype is difficult to main-

tain in vitro. Many methods have been developed for

maintaining long-term functionality of hepatocytes

through the use of a variety of configurations and bioma-

terial constructs, which are further discussed in the “In

vitro models” section. Due to limitations in the supply of

mature human hepatocytes, many groups have attempted

to promote the expansion and proliferation of PHHs

in vitro. Peng et al. have shown that TNFα promotes the

expansion of hepatocytes in 3D cultures and enables serial

passaging and long-term culture for more than 6 months

[103]. In a similarly notable study, Hu et al. identified an

optimal cell culture cocktail consisting of B27 supplement

(without vitamin A), R-spondin, CHIR99021 (a Wnt ago-

nist), NAC, nicotinamide, gastrin, epidermal growth fac-

tor (EGF), TGFα, fibroblast growth factor (FGF)7,

FGF10, HGF, a TGFβ inhibitor (A83-01), and ROCK

inhibitor that led to long-term 3D organoid culture of

PHHs [104].

Fetal and adult progenitors

Given their ability to differentiate into diverse lineages

both in vitro and in vivo, iPSC and human embryonic

stem cell cultures can also be utilized to generate HLCs.

Various differentiation protocols have been applied to

these cultures to yield cell populations that exhibit some

phenotypic and functional characteristics of hepatocytes

[62,105�108]. These populations are termed HLCs

because of their expression of fetal proteins and fetal-like

cytochrome P450 profiles [109]. While they are distinct

from mature adult hepatocytes, HLCs can still serve as a

potential cell source in very specific contexts.

In addition to pluripotent cells, bipotential progenitor

cells can also serve as a source for hepatocytes. Huch

et al. delineated conditions that allow for long-term

expansion of adult bile duct-derived EpCAM1 bipoten-

tial progenitor cells from the human liver [110]. The

expanded cell population attained using their protocol is

stable at the chromosomal level and can be converted into

functional HLCs in vitro and in vivo [110].

Reprogrammed hepatocytes

HLCs can also be generated using direct reprogramming

of mature cell types. For example, several groups have

demonstrated the feasibility of reprogramming fibroblasts

into HLCs without a pluripotent intermediate [111�113].

Cheng et al. demonstrated that a combination of nuclear

factors can stimulate the conversion of hepatoma cells to

HLCs [101]. These findings raise the future possibility of

deriving human HLCs directly from another adult cell

type.

Extracellular matrix for cell therapies

The ECM of the liver provides a structural scaffold with

bioactive cues that modulate hepatic function and pro-

mote vascularization. Collagen and fibronectin are the

major structural components of the liver ECM. Along

with other nonstructural proteins, these components par-

ticipate in integrin-mediated signaling between cells and

their surrounding matrix. Hepatocytes are sensitive to

their ECM, and it has been demonstrated that the presence

of abnormal amounts and/or types of ECM components

correlates with the onset and progression of liver fibrosis

[114].

ECM scaffolds for hepatic tissue engineering are use-

ful for constructing 3D tissue models and as a delivery

vehicle for implants. Polymeric biomaterial hydrogels

gained popularity as an engineering tool for recapitulating

a physiologically relevant 3D tissue niche. Aside from

creating a permissive environment for hepatocyte survival

and growth, ECM scaffolds for hepatic tissue engineering

also enable the formation of biliary and vascular networks

that will be further discussed in the “Vascular and biliary

tissue engineering” section. Broadly speaking, ECM scaf-

folds can be constructed using synthetic and/or naturally

derived polymers.

744 PART | ELEVEN Gastrointestinal system

Page 9: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

Natural scaffold chemistry and modifications

A wide range of natural biomaterial polymers spanning

polysaccharides (e.g., dextran and chitosan), peptides

(e.g., collagen and fibrin), decellularized ECM (dECM),

and composites of these have been employed as hepatic

tissue�engineering scaffolds [55,57,58,115�121]. The

advantages of biologically derived materials include their

biocompatibility; naturally occurring cell adhesive moie-

ties; and, in the case of decellularization, native architec-

tural presentation of ECM molecules. However, naturally

derived biomaterials have several barriers to use in the

clinic, primarily due to lot-to-lot variability and xenoge-

neic origin.

The choice of material determines the physicochemi-

cal and biological properties of the scaffold. For

example, early efforts in developing implantable hepatic

constructs utilized collagen-coated dextran microcarriers

that enabled hepatocyte attachment since hepatocytes

are known to be anchorage-dependent cells. The intra-

peritoneal transplantation of these hepatocyte-attached

microcarriers resulted in successful replacement of

liver functions in two different rodent models of genetic

liver disorders [122]. Subsequently, collagen-coated or

peptide-modified cellulose [120,123], gelatin [124], and

gelatin�chitosan composite [125] microcarrier chemis-

tries have also been explored for their capacity to promote

hepatocyte attachment. On the other hand, materials that

are poorly cell adhesive such as alginate [115] have been

exploited for their utility in promoting hepatocy-

te�hepatocyte aggregation (i.e., spheroid formation) and

phenotypic stabilization within these scaffolds.

Collectively, the size of engineered tissues created by

these approaches is limited by oxygen and nutrient diffu-

sion to only a few hundred microns in thickness.

To address this constraint, recent work has sought to

use decellularized whole organ tissue as a matrix for liver

tissue engineering. The decellularization process utilizes

perfusion-based technologies to remove cells from donor

tissues but preserve the structural and functional charac-

teristics of the native underlying tissue. Recent advances

in decellularization protocols have yielded scaffolds with

native liver ECM composition, growth factor presentation,

vascular structure, and biliary network architecture

[126�128]. To date, seeding protocols have achieved up

to 95% efficiency of recellularization with relevant cell

populations (e.g., hepatocytes, vascular cells and bipotent

hepatic progenitors); resulting recellularized grafts exhib-

ited liver-specific function, and survival after transplanta-

tion in rodents [121,126,129]. Furthermore, cell-laden,

xeno-derived dECM scaffolds are compatible with immu-

nocompetent animal models [128]. However, given the

shortage of donor tissue, the wide use of dECM scaffolds

is unlikely.

Synthetic scaffold chemistry

In contrast to biologically derived material systems, syn-

thetic materials enable precisely customized architecture

(porosity and topography), mechanical and chemical

properties, and degradation modality and kinetics, which

are known to drive cell behavior. Synthetic materials that

have been explored for liver tissue engineering include

poly(L-lactic acid) (PLLA), PLGA, poly(ε-caprolactone),and PEG [85,98,130�135]. Polyesters such as PLLA and

PLGA are the most common synthetic polymers utilized

in the generation of porous tissue-engineering constructs.

These materials are biocompatible, biodegradable, and

have been used as scaffolds for HT [132,136]. A key

advantage of PLGA is the potential to finely tune its deg-

radation time due to differences in susceptibility to hydro-

lysis of the ester groups of its monomeric components

(lactic acid and glycolic acid). However, the accumulation

of hydrolytic degradation products has been shown to pro-

duce an acidic environment within the scaffold which

initiates peptide degradation and stimulates inflammation,

which may affect hepatocyte function [137].

Consequently, as alternatives to macroporous scaffold

systems, approaches aimed at the efficient and homoge-

neous encapsulation of hepatocytes within a fully 3D

structure have been explored. In particular, hydrogels that

exhibit high water content and thus similar mechanical

properties to tissues are widely utilized for various tissue-

engineering applications, including hepatocellular plat-

forms. Synthetic, PEG-based hydrogels have been

increasingly utilized in liver tissue-engineering applica-

tions due to their high water content, hydrophilicity, resis-

tance to protein adsorption, biocompatibility, ease of

chemical modification, and the ability to be polymerized

in the presence of cells, thereby enabling the fabrication

of 3D networks with uniform cellular distribution [138].

PEG-based hydrogels have been used for the encapsula-

tion of diverse cell types, including immortalized and pri-

mary hepatocytes and hepatoblastoma cell lines

[85,98,135]. The encapsulation of primary hepatocytes

requires distinct material modifications [e.g., 10% w/v

PEG hydrogel, inclusion of RGD adhesive motifs] as

detailed below, as well as, analogous to 2D coculture sys-

tems, the inclusion of nonparenchymal supporting cell

types such as fibroblasts and endothelial cells [135].

Modifications in scaffold chemistry

The relatively inert nature of synthetic scaffolds allows

for the controlled incorporation of chemical/polymer moi-

eties or biologically active factors to regulate different

aspects of cellular function. Chemical modifications such

as oxygen plasma treatment or alkali hydrolysis of PLGA

[139,140], or the incorporation of polymers such as

Hepatic tissue engineering Chapter | 40 745

Page 10: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

poly(vinyl alcohol) or poly(N-p-vinylbenzyl-4-O-β-D-galactopyranosyl-D-glucoamide) (PVLA) into PLGA or

PLLA scaffolds [132,141,142] have improved hepatocyte

adhesion by modulating the hydrophilicity of the scaffold

surface [143]. Biological factors may include whole bio-

molecules or short bioactive peptides. Whole biomole-

cules are typically incorporated by nonspecific adsorption

of ECM molecules such as collagen, laminin, or fibronec-

tin [139,144]; covalent conjugation of sugar molecules

such as heparin [145,146], galactose [131,147], lactose

[145] or fructose [148]; or growth factors such as EGF

[149]. Alternatively, short bioadhesive peptides that inter-

act with cell surface integrin receptors have been exten-

sively utilized to promote hepatocyte attachment in

synthetic scaffolds. For example, conjugation of the RGD

peptide to PLLA has been shown to enhance hepatocyte

attachment [150], whereas RGD modification signifi-

cantly improved the stability of long-term hepatocyte

function in PEG hydrogels [98,135]. The additional incor-

poration of adhesive peptides that bind other integrins

may serve as a way to further modulate and enhance

hepatocyte function within synthetic polymer substrates.

Moreover, Stevens et al. demonstrated that integration of

matrix metalloproteinase-sensitive peptide sequences into

hydrogel networks as degradable linkages has been shown

to enable cell-mediated remodeling of the hydrogel [151].

The capacity to modify biomaterial scaffold chemistry

through the introduction of biologically active factors will

likely enable the finely tuned regulation of cell function

and interactions with host tissues important for

implantable systems.

Porosity

A common feature of many implantable tissue-

engineering approaches is the use of porous scaffolds that

provide mechanical support, often in conjunction with

cues for growth and morphogenesis. Collagen sponges,

various alginate and chitosan composites, and PLGA are

the most commonly used porous scaffolds for hepatocyte

culture and are generally synthesized using freeze-dry or

gas-foaming techniques. Pore size has been found to regu-

late cell spreading and cell�cell interactions, both of

which can influence hepatocyte functions [116], and may

also influence angiogenesis and tissue ingrowth [152].

Porous, acellular scaffolds are normally seeded using

gravity or centrifugal forces, capillary action, convective

flow, or through cellular recruitment with chemokines,

but hepatocyte seeding is generally heterogeneous in these

scaffolds [153,154].

Vascular and biliary tissue engineering

Beyond compatibility with hepatic cell types, scaffolds

should also be conducive to vasculature formation.

Relying on vascularization by the host is not sufficient for

large tissue constructs required for the clinic, because

cells that are not near capillary structures

(. 150�200 μm) are at a risk for necrosis after a matter

of hours due to a lack of oxygen, nutrient availability, and

waste transport. In this section, we discuss composite

approaches toward building scalable, vascularized

constructs.

Vascular engineering

Approaches to engineering vessels can generally be cate-

gorized as bottom-up induction of vascular assembly and

top-down fabrication of vascular conduits [155]. Bottom-

up vascular engineering approaches are built upon the

idea of neovascularization, or new vessel formation.

Vessel formation can occur by angiogenic sprouting, the

formation of vessels branching off of an existing blood

vessel, or vasculogenesis, the self-assembly of single

endothelial cells or progenitor cells into lumenized ves-

sels. Despite the ability of single vascular cells to coa-

lesce to enable self-assembly, vasculogenesis is

accelerated by coculture with supporting stromal cells,

such as fibroblasts, mesenchymal stem cells, and pericytes

[155�157]. Studies exploring angiogenesis and sprouting

events suggest that chemical gradients, fluid-driven shear

stress, and a hypoxia are key players in vessel formation

[158�161].

Top-down fabrication approaches dictate geometry

and architecture, rather than driving self-assembly.

Polymer molding using microetched silicon has been

shown to generate extensive channel networks with capil-

lary dimensions, though it is not amenable to high-

throughput manufacturing [162]. While direct printing of

cells can be cytotoxic, 3D printing has become a popular

approach for fabricating hollow channels that enable vas-

cular cell seeding. The challenge lies in using this

approach to build patent capillary beds, which are

5�10 μm in diameter. While two-photon polymerization

has an impressively high feature size resolution at

100 nm, the trade-off between build volume (i.e., building

constructs large enough for clinical impact), build speed

(i.e., impacting manufacturability), and printer resolution

renders it inappropriate for most applications in tissue

engineering. Alternate printers using direct-ink writing

(DIW) of viscoelastic materials have emerged as a power-

ful tool for the fabrication of patterned hydrogel con-

structs. DIW can achieve minimum feature size

resolutions from 1 to 250 μm, depending on the ink

“building block” size [163]. However, DIW printing

requires yield-stress fluid inks with restrictive viscosities

(102�106 mPa s), such that they fluidize under stress but

regain the original shear elastic modulus after printing

[163]. Kolesky et al. demonstrated that DIW and fugitive

inks were useful for multimaterial printing as well as

746 PART | ELEVEN Gastrointestinal system

Page 11: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

construction of thick (. 1 cm), vascularized tissues

[164,165]. Miller et al. demonstrate the use of thermal

microextrusion approach to create sacrificial sugar glass

lattices that can be embedded in various cell-laden bioma-

terials, evacuated and subsequently lined with vascular

cells [166].

Taken together, self-assembly driven formation of a

capillary bed can be combined with printing of larger ves-

sels to enable the fabrication of a fully vascularized tissue

construct. Song et al. used 3D printing to create curved

vascular channels using a fugitive ink [159]. The vascular

cells lining the channel were able to degrade the sur-

rounding hydrogel matrix and undergo sprouting when

exposed to angiogenic factors. They further demonstrated

that the curvature and complexity of printed vasculature

impacted the extent of sprouting, which will be an impor-

tant consideration for vessel patterning in future clinical

applications [159].

Host integration

For vascularized constructs to survive after implantation

the engineered vessels must anastomose with the host vas-

culature. To date, the exact mechanism that drives inte-

gration with the host is not well understood [155].

A number of studies have elucidated the contribution of

cytokines important in angiogenesis and recruitment of

host vasculature in implant constructs, such as vascular

EGF (VEGF) [136], basic FGF [167], and VEGF in com-

bination with platelet-derived growth factor [168].

Furthermore, preimplantation of VEGF releasing alginate

scaffolds prior to hepatocyte seeding was demonstrated to

enhance capillary density and improve engraftment [169].

In addition, while building an AL tissue construct,

Stevens et al. demonstrated that the coimplantation of par-

allel endothelial cell cords with PHH and fibroblast spher-

oids led to better hepatic performance and survival when

compared to an implant with nonpatterned endothelial

cells, suggesting that there may be an optimal templated

endothelial geometry that enables vascularization and

host integration in vivo [58]. Surgical anastomosis poses

an alternative to biologically driven anastomosis, though

this approach requires invasive surgery and access to

suture-able vessels both in the graft and in the host.

Strategies to incorporate vasculature into engineered con-

structs include the microfabrication of vascular units with

accompanying surgical anastomosis during implantation

[75,170].

In addition to interactions with the vasculature, inte-

gration with other aspects of host tissue will constitute

important future design parameters. For instance, incorpo-

ration of hydrolytic or protease-sensitive domains into

hepatocellular hydrogel constructs could enable the degra-

dation of these systems following implantation [151]. Of

note, liver regeneration proceeds in conjunction with a

distinctive array of remodeling processes such as protease

expression and ECM deposition. Interfacing with these

features could provide a mechanism for the efficient inte-

gration of implantable constructs. Similarly to whole liver

or cell transplantation, the host immune response follow-

ing the transplant of tissue-engineered constructs is also a

major consideration. Immunosuppressive treatments will

likely play an important role in initial therapies, although

stem cell�based approaches hold the promise of

implantable systems with autologous cells. Furthermore,

harnessing the liver’s unique ability to induce antigen-

specific tolerance [171] could potentially represent

another means for improving the acceptance of engi-

neered grafts.

Biliary network engineering

Importantly, future iterations of hepatic grafts should

include a biliary system, which is responsible for excre-

tory functions. In a similar vein, we envision that a com-

bination of “top-down” manufacturing, such as the

aforementioned technologies for generating patent vascu-

lar conduits, and “bottom-up” approaches, which could

involve leveraging biological phenomena that drive bili-

ary morphogenesis, will be useful for building a biliary

network.

The biliary tree is a complex, 3D network of tubular

conduits of various sizes and properties. The liver con-

tains an intrahepatic compartment that is lined by biliary

epithelial cells, termed cholangiocytes, that aid in the

modification and removal of hepatocyte-secreted bile.

Even though the blood vessel�fabrication approaches

delineated above have not yet been applied to engineering

bile networks for implantable liver constructs, advance-

ments in cholangiocyte sourcing methods have made it

feasible. Sampaziotis et al. identified a protocol for

directed differentiation of human iPSCs into

cholangiocyte-like cells [172]. In 2017 the same group

provided the first proof-of-concept study to reconstruct

the gallbladder wall and repair the biliary epithelium

using human primary cholangiocytes expanded in vitro

[173]. Furthermore, current studies are focused on the

development of in vitro models that exhibits biliary mor-

phogenesis and recapitulates appropriate polarization and

bile canaliculi organization [174�176], as well as plat-

forms for the engineering of artificial bile duct structures

[177,178].

Conclusion and outlook

Traditionally, hepatic tissue�engineering research has

focused on designing the microenvironment to support a

stable hepatic phenotype. As concomitant advances in

pluripotent cell research and polymer chemistry have

been actualized, new cell sources and extracellular

Hepatic tissue engineering Chapter | 40 747

Page 12: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

matrices have been added to the pipeline. This interdisci-

plinary synergy has been the driving force behind the

development of tissue-engineered grafts with long-term

survival and growth. In order to inch closer to the regen-

erative medicine north star of an ex vivo engineered graft

that can serve as a replacement for the native organ, there

are several areas to consider for improvement: (1) vascu-

larization of thick, dense grafts through a combination of

self-assembly and bioprinting; (2) engineering of the

hepatic graft to prevent immune rejection in allogeneic

and xenogeneic settings; (3) improved understanding of

metabolic and cellular requirements of various liver dis-

eases; (4) development of scalable, renewable cell sources

that do not compromise the functional capabilities of

cells; (5) leveraging of animal injury models as bioreac-

tors for cell sourcing; and (6) upscaling of grafts in a

manner that is compatible with FDA’s Good

Manufacturing Practice standards.

References

[1] Mokdad AA, Lopez AD, Shahraz S, Lozano R, Mokdad AH,

Stanaway J, et al. Liver cirrhosis mortality in 187 countries

between 1980 and 2010: a systematic analysis. BMC Med

2014;12:145.

[2] Murali AR, Menon KVN. Acute liver failure. 2017, ,http://www.

clevelandclinicmeded.com/medicalpubs/diseasemanagement/hepa-

tology/acute-liver-failure/., 2017 (accessed 16.01.2019).

[3] Asrani SK, Devarbhavi H, Eaton J, Kamath PS. Burden of liver

diseases in the world. J Hepatol 2018;70:151�71. Available from:

https://doi.org/10.1016/j.jhep.2018.09.014.

[4] O’Grady JG, Schalm SW, Williams R. Acute liver failure : rede-

fining the syndromes. Lancet 1985;342:273�5.

[5] Bernal W, Wendon J. Acute liver failure. N Engl J Med

2013;369:2525�34.

[6] Bhatia SN, Underhill GH, Zaret KS, Fox IJ. Cell and tissue engi-

neering for liver disease. Sci Transl Med 2014;6:245sr2.

[7] Sokal EM. Liver transplantation for inborn errors of liver metabo-

lism. J Inherit Metab Dis 2006;29:426�30. Available from:

https://doi.org/10.1007/s10545-006-0288-x.

[8] Donati G, Stagni B, Piscaglia F, Venturoli N, Morselli-Labate

AM, Rasciti L, et al. Increased prevalence of fatty liver in arterial

hypertensive patients with normal liver enzymes: role of insulin

resistance. Gut 2004;53:1020�3. Available from: https://doi.org/

10.1136/gut.2003.027086.

[9] Farrell GC, Van Rooyen D, Gan L, Chitturi S. NASH is an

inflammatory disorder: pathogenic, prognostic and therapeutic

implications. Gut Liver 2012;6:149.

[10] Cheung O, Sanyal AJ. Abnormalities of lipid metabolism in non-

alcoholic fatty liver disease. Semin Liver Dis 2008;28:351�9.

[11] Estes C, Razavi H, Loomba R, Younossi Z, Sanyal AJ. Modeling the

epidemic of nonalcoholic fatty liver disease demonstrates an expo-

nential increase in burden of disease. Hepatology 2018;67:123�33.

Available from: https://doi.org/10.1002/hep.29466.

[12] Kwong GA. Mass-encoded synthetic biomarkers for multiplexed

urinary monitoring of disease. Nat Biotechnol 2013;31:63�70

,https://doi.org/10.1038/nbt.2464..

[13] Carpentier B, Gautier A, Legallais C. Artificial and bioartificial

liver devices: present and future. Gut 2009;58:1690�702.

Available from: https://doi.org/10.1136/gut.2008.175380.

[14] Ellis AJ, Hughes RD, Wendon JA, Dunne J, Langley PG, Kelly JH,

et al. Pilot-controlled trial of the extracorporeal liver assist device

in acute liver failure. Hepatology 1996;24:1446�51. Available

from: https://doi.org/10.1053/jhep.1996.v24.pm0008938179.

[15] Custer L, Mullon CJ-P. Oxygen delivery to and use by primary

porcine hepatocytes in the HepatAssistt 2000 system for extracor-

poreal treatment of patients in end-stage liver failure. Oxygen

transport to tissue XX. Springer; 1998. p. 261�71.

[16] Sauer IM, Gerlach JC. Modular extracorporeal liver support. Artif

Organs 2002;26:703�6.

[17] Van De Kerkhove M-P, Di Florio E, Scuderi V, Mancini A, Belli

A, Bracco A, et al. Phase I clinical trial with the AMC-

bioartificial liver. Int J Artif Organs 2002;25:950�9.

[18] Makin AJ, Williams R. Acetaminophen-induced hepatotoxicity:

predisposing factors and treatments. Adv Intern Med

1997;42:453�83.

[19] Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM,

Bass NM, et al. Pioglitazone, vitamin E, or placebo for nonalco-

holic steatohepatitis. N Engl J Med 2010;362:1675�85.

[20] Starzl TE, Marchioro TL, Von Kaulla KN, Hermann G, Brittain

RS, Waddell WR. Homotransplantation of the liver in humans.

Surg Gynecol Obstet 1963;117:659.

[21] Starzl TE, Groth CG, Brettschneider L, Penn I, Fulginiti VA,

Moon JB, et al. Orthotopic homotransplantation of the human

liver. Ann Surg 1968;168:392.

[22] Zarrinpar A, Busuttil RW. Liver transplantation: past, present and

future. Nat Rev Gastroenterol Hepatol 2013;10:434�40. Available

from: https://doi.org/10.1038/nrgastro.2013.88.

[23] Fan S-T, Lo C-M, Liu C-L, Yong B-H, Chan JK-F, Ng IO-L.

Safety of donors in live donor liver transplantation using right

lobe grafts. Arch Surg 2000;135:336�40.

[24] Busuttil RW, Goss JA. Split liver transplantation. Ann Surg

1999;229:313.

[25] Tector AJ, Mangus RS, Chestovich P, Vianna R, Fridell JA,

Milgrom ML, et al. Use of extended criteria livers decreases wait

time for liver transplantation without adversely impacting post-

transplant survival. Ann Surg 2006;244:439.

[26] Niu D, Wei H-J, Lin L, George H, Wang T, Lee I-H, et al.

Inactivation of porcine endogenous retrovirus in pigs using

CRISPR-Cas9. Science 2017;357:1303�7. Available from:

https://doi.org/10.1126/science.aan4187.

[27] Langin M, Mayr T, Reichart B, Michel S, Buchholz S, Guethoff

S, et al. Consistent success in life-supporting porcine cardiac

xenotransplantation. Nature 2018;564:430�3. Available from:

https://doi.org/10.1038/s41586-018-0765-z.

[28] Matas AJ, Sutherland DE, Steffes MW, Mauer SM, Sowe A,

Simmons RL, et al. Hepatocellular transplantation for metabolic

deficiencies: decrease of plasma bilirubin in Gunn rats. Science

1976;192(80):892�4.

[29] Mito M, Kusano M. Hepatocyte transplantation in man. Cell

Transplant 1993;2:65�74.

748 PART | ELEVEN Gastrointestinal system

Page 13: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

[30] Fisher RA, Strom SC. Human hepatocyte transplantation: world-

wide results. Transplantation 2006;82:441�9. Available from:

https://doi.org/10.1097/01.tp.0000231689.44266.ac.

[31] Horslen SP, McCowan TC, Goertzen TC, Warkentin PI, Cai HB,

Strom SC, et al. Isolated hepatocyte transplantation in an infant

with a severe urea cycle disorder. Pediatrics 2003;111:1262�7.

[32] Dhawan A, Mitry RR, Hughes RD, Lehec S, Terry C, Bansal S,

et al. Hepatocyte transplantation for inherited factor VII defi-

ciency. Transplantation 2004;78:1812�14.

[33] Muraca M, Gerunda G, Neri D, Vilei M-T, Granato A, Feltracco

P, et al. Hepatocyte transplantation as a treatment for glycogen

storage disease type 1a. Lancet 2002;359:317�18.

[34] Sokal EM, Smets F, Bourgois A, Van Maldergem L, Buts J-P,

Reding R, et al. Hepatocyte transplantation in a 4-year-old girl

with peroxisomal biogenesis disease: technique, safety, and meta-

bolic follow-up1. Transplantation 2003;76:735�8.

[35] Dhawan A, Puppi J, Hughes RD, Mitry RR. Human hepatocyte

transplantation: current experience and future challenges. Nat Rev

Gastroenterol Hepatol 2010;7:288�98.

[36] Beck BB, Habbig S, Dittrich K, Stippel D, Kaul I, Koerber F,

et al. Liver cell transplantation in severe infantile oxalosis—a

potential bridging procedure to orthotopic liver transplantation?

Nephrol Dial Transplant 2012;27:2984�9.

[37] Bilir BM, Guinette D, Karrer F, Kumpe DA, Krysl J, Stephens J,

et al. Hepatocyte transplantation in acute liver failure. Liver

Transplant 2000;6:32�40.

[38] Sutherland DE, Numata M, Matas AJ, Simmons RL, Najarian JS.

Hepatocellular transplantation in acute liver failure. Surgery

1977;82:124�32.

[39] Hoppo T, Komori J, Manohar R, Stolz DB, Lagasse E. Rescue of

lethal hepatic failure by hepatized lymph nodes in mice.

Gastroenterology 2011;140:656�66.

[40] Komori J, Boone L, DeWard A, Hoppo T, Lagasse E. The mouse

lymph node as an ectopic transplantation site for multiple tissues.

Nat Biotechnol 2012;30:976�83. Available from: https://doi.org/

10.1038/nbt.2379.

[41] Younossi ZM, Loomba R, Rinella ME, Bugianesi E, Marchesini

G, Neuschwander-Tetri BA, et al. Current and future therapeutic

regimens for nonalcoholic fatty liver disease and nonalcoholic

steatohepatitis. Hepatology 2018;68:361�71.

[42] Gebhardt R. Metabolic zonation of the liver: regulation and impli-

cations for liver function. Pharmacol Ther 1992;53:275�354.

Available from: https://doi.org/10.1016/0163-7258(92)90055-5.

[43] Godoy P, Hewitt NJ, Albrecht U, Andersen ME, Ansari N,

Bhattacharya S, et al. Recent advances in 2D and 3D in vitro sys-

tems using primary hepatocytes, alternative hepatocyte sources

and non-parenchymal liver cells and their use in investigating

mechanisms of hepatotoxicity, cell signaling and ADME. Arch

Toxicol 2013;87:1315�530. Available from: https://doi.org/

10.1007/s00204-013-1078-5.

[44] Swift B, Pfeifer ND, Brouwer KLR. Sandwich-cultured hepato-

cytes: an in vitro model to evaluate hepatobiliary transporter-

based drug interactions and hepatotoxicity. Drug Metab Rev

2010;42:446�71. Available from: https://doi.org/10.3109/

03602530903491881.

[45] Dunn JCY, Tompkins RG, Yarmush ML. Long-term in vitro func-

tion of adult hepatocytes in a collagen sandwich configuration.

Biotechnol Prog 1991;7:237�45. Available from: https://doi.org/

10.1021/bp00009a007.

[46] Guguen-Guillouzo C, Clement B, Baffet G, Beaumont C, Morel-

Chany E, Glaise D, et al. Maintenance and reversibility of active

albumin secretion by adult rat hepatocytes co-cultured with

another liver epithelial cell type. Exp Cell Res 1983;143:47�54.

Available from: https://doi.org/10.1016/0014-4827(83)90107-6.

[47] Bale SS, Geerts S, Jindal R, Yarmush ML. Isolation and co-

culture of rat parenchymal and non-parenchymal liver cells to

evaluate cellular interactions and response. Sci Rep 2016;6:25329.

Available from: https://doi.org/10.1038/srep25329.

[48] March S, Ramanan V, Trehan K, Ng S, Galstian A, Gural N, et al.

Micropatterned coculture of primary human hepatocytes and sup-

portive cells for the study of hepatotropic pathogens. Nat Protoc

2015;10:2027�53. Available from: https://doi.org/10.1038/

nprot.2015.128.

[49] Khetani SR, Berger DR, Ballinger KR, Davidson MD, Lin C,

Ware BR. Microengineered liver tissues for drug testing. J Lab

Autom 2015;20:216�50. Available from: https://doi.org/10.1177/

2211068214566939.

[50] Khetani SR, Bhatia SN. Microscale culture of human liver cells

for drug development. Nat Biotechnol 2008;26:120�6. Available

from: https://doi.org/10.1038/nbt1361.

[51] Davidson MD, Kukla DA, Khetani SR. Microengineered cultures

containing human hepatic stellate cells and hepatocytes for drug

development. Integr Biol 2017;9:662�77. Available from: https://

doi.org/10.1039/C7IB00027H.

[52] Yuasa C, Tomita Y, Shono M, Ishimura K, Ichihara A.

Importance of cell aggregation for expression of liver functions

and regeneration demonstrated with primary cultured hepatocytes.

J Cell Physiol 1993;156:522�30. Available from: https://doi.org/

10.1002/jcp.1041560311.

[53] Lin R-Z, Chang H-Y. Recent advances in three-dimensional multi-

cellular spheroid culture for biomedical research. Biotechnol J

2008;3:1172�84. Available from: https://doi.org/10.1002/

biot.200700228.

[54] Griffith LG, Swartz MA. Capturing complex 3D tissue physiology

in vitro. Nat Rev Mol Cell Biol 2006;7:211�24. Available from:

https://doi.org/10.1038/nrm1858.

[55] Liaw C-Y, Ji S, Guvendiren M. Engineering 3D hydrogels for per-

sonalized in vitro human tissue models. Adv Healthc Mater

2018;7:1701165. Available from: https://doi.org/10.1002/

adhm.201701165.

[56] Bell CC, Hendriks DFG, Moro SML, Ellis E, Walsh J, Renblom

A, et al. Characterization of primary human hepatocyte spheroids

as a model system for drug-induced liver injury, liver function

and disease. Sci Rep 2016;6:25187. Available from: https://doi.

org/10.1038/srep25187.

[57] Stevens KR, Ungrin MD, Schwartz RE, Ng S, Carvalho B,

Christine KS, et al. InVERT molding for scalable control of tissue

microarchitecture. Nat Commun 2013;4:1847. Available from:

https://doi.org/10.1038/ncomms2853.

[58] Stevens KR, Scull MA, Ramanan V, Fortin CL, Chaturvedi RR,

Knouse KA, et al. In situ expansion of engineered human liver tis-

sue in a mouse model of chronic liver disease. Sci Transl Med

2017;9:eaah5505. Available from: https://doi.org/10.1126/sci-

translmed.aah5505.

[59] Takebe T, Sekine K, Enomura M, Koike H, Kimura M, Ogaeri T,

et al. Vascularized and functional human liver from an iPSC-

derived organ bud transplant. Nature 2013;499:481�4. Available

from: https://doi.org/10.1038/nature12271.

Hepatic tissue engineering Chapter | 40 749

Page 14: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

[60] Takebe T, Sekine K, Kimura M, Okamoto S, Ueno Y, Taniguchi

H. Massive and reproducible production of liver buds entirely

from human pluripotent stem cells. Cell Rep 2017;21:2661�70

,https://doi.org/10.1016/j.celrep.2017.11.005..

[61] Huh D, Hamilton GA, Ingber DE. From 3D cell culture to organs-

on-chips. Trends Cell Biol 2011;21:745�54. Available from:

https://doi.org/10.1016/j.tcb.2011.09.005.

[62] Gouon-Evans V, Boussemart L, Gadue P, Nierhoff D, Koehler CI,

Kubo A, et al. BMP-4 is required for hepatic specification of

mouse embryonic stem cell-derived definitive endoderm. Nat

Biotechnol 2006;24:1402.

[63] Bhatia SN, Ingber DE. Microfluidic organs-on-chips. Nat

Biotechnol 2014;32:760�72. Available from: https://doi.org/

10.1038/nbt.2989.

[64] Huh D, Torisawa Y, Hamilton GA, Kim HJ, Ingber DE.

Microengineered physiological biomimicry: organs-on-chips. Lab

Chip 2012;12:2156. Available from: https://doi.org/10.1039/

c2lc40089h.

[65] Kim HJ, Huh D, Hamilton G, Ingber DE. Human gut-on-a-chip

inhabited by microbial flora that experiences intestinal peristalsis-

like motions and flow. Lab Chip 2012;12:2165. Available from:

https://doi.org/10.1039/c2lc40074j.

[66] Kim P, Kwon KW, Park MC, Lee SH, Kim SM, Suh KY. Soft

lithography for microfluidics: a review. Biochip J 2008;2:1�11.

[67] Smith Q, Gerecht S. Going with the flow: microfluidic platforms in

vascular tissue engineering. Curr Opin Chem Eng 2014;3:42�50.

Available from: https://doi.org/10.1016/J.COCHE.2013.11.001.

[68] Lee S-A, No DY, Kang E, Ju J, Kim D-S, Lee S-H. Spheroid-based

three-dimensional liver-on-a-chip to investigate hepatocyte�hepa-

tic stellate cell interactions and flow effects. Lab Chip

2013;13:3529. Available from: https://doi.org/10.1039/c3lc50197c.

[69] Skardal A, Murphy SV, Devarasetty M, Mead I, Kang H-W, Seol

Y-J, et al. Multi-tissue interactions in an integrated three-tissue

organ-on-a-chip platform. Sci Rep 2017;7:8837. Available from:

https://doi.org/10.1038/s41598-017-08879-x.

[70] Edington CD, Chen WLK, Geishecker E, Kassis T, Soenksen LR,

Bhushan BM, et al. Interconnected microphysiological systems for

quantitative biology and pharmacology studies. Sci Rep

2018;8:4530. Available from: https://doi.org/10.1038/s41598-018-

22749-0.

[71] Groger M, Rennert K, Giszas B, Weiß E, Dinger J, Funke H, et al.

Monocyte-induced recovery of inflammation-associated hepatocel-

lular dysfunction in a biochip-based human liver model. Sci Rep

2016;6:21868. Available from: https://doi.org/10.1038/srep21868.

[72] Tsang VL, Bhatia SN. Three-dimensional tissue fabrication. Adv

Drug Deliv Rev 2004;56:1635�47.

[73] Kim SS, et al. Survival and function of hepatocytes on a novel

three-dimensional synthetic biodegradable polymer scaffold with

an intrinsic network of channels. Ann Surg 1998;228:8�13.

[74] Petronis S, Eckert KL, Gold J, Wintermantel E. Microstructuring

ceramic scaffolds for hepatocyte cell culture. J Mater Sci Mater

Med 2001;12:523�8.

[75] Kaihara S, et al. Silicon micromachining to tissue engineer

branched vascular channels for liver fabrication. Tissue Eng

2000;6:105�17.

[76] Ogawa K, Ochoa ER, Borenstein J, Tanaka K, Vacanti JP. The

generation of functionally differentiated, three-dimensional

hepatic tissue from two-dimensional sheets of progenitor small

hepatocytes and non-parenchymal cells. Transplantation

2004;77:1783�9.

[77] Vozzi G, Flaim C, Ahluwalia A, Bhatia SN. Fabrication of PLGA

scaffolds using soft lithography and microsyringe deposition.

Biomaterials 2003;24:2533�40.

[78] Tan W, Desai TA. Microfluidic patterning of cells in extracellular

matrix biopolymers: effects of channel size, cell type, and matrix

composition on pattern integrity. Tissue Eng 2003;9:255�67.

[79] Wang X, et al. Generation of three-dimensional hepatocyte/gelatin

structures with rapid prototyping system. Tissue Eng

2006;12:83�90.

[80] Hahn MS, et al. Photolithographic patterning of polyethylene gly-

col hydrogels. Biomaterials 2006;27:2519�24.

[81] Revzin A, et al. Fabrication of poly(ethylene glycol) hydrogel

microstructures using photolithography. Langmuir

2001;17:5440�7.

[82] Beebe DJ, et al. Functional hydrogel structures for autonomous

flow control inside microfluidic channels. Nature

2000;404:588�90.

[83] Liu VA, Bhatia SN. Three-dimensional photopatterning of hydro-

gels containing living cells. Biomed Microdevices 2002;4:257�66.

[84] Tan W, Desai TA. Layer-by-layer microfluidics for biomimetic

three-dimensional structures. Biomaterials 2004;25:1355�64

S0142961203006756 [pii].

[85] Liu Tsang V, et al. Fabrication of 3D hepatic tissues by additive

photopatterning of cellular hydrogels. FASEB J

2007;21:790�801. Available from: https://doi.org/10.1096/fj.06-

7117com.

[86] Li CY, Wood DK, Hsu CM, Bhatia SN. DNA-templated assembly

of droplet-derived PEG microtissues. Lab Chip 2011;11:2967�75.

[87] Palmes D, Spiegel H-U. Animal models of liver regeneration.

Biomaterials 2004;25:1601�11.

[88] Forbes SJ, Newsome PN. Liver regeneration—mechanisms and

models to clinical application. Nat Rev Gastroenterol Hepatol

2016;13:473�85. Available from: https://doi.org/10.1038/nrgastro.

2016.97.

[89] Grompe M, Strom S. Mice with human livers. Gastroenterology

2013;145:1209�14. Available from: https://doi.org/10.1053/J.

GASTRO.2013.09.009.

[90] Tan AKY, Loh KM, Ang LT. Evaluating the regenerative potential

and functionality of human liver cells in mice. Differentiation

2017;98:25�34. Available from: https://doi.org/10.1016/j.diff.2017.

09.003.

[91] Gupta S, Gorla GR, Irani AN. Hepatocyte transplantation: emerg-

ing insights into mechanisms of liver repopulation and their rele-

vance to potential therapies. J Hepatol 1999;30:162�70.

Available from: https://doi.org/10.1016/S0168-8278(99)80022-1.

[92] Rhim JA, Sandgren EP, Palmiter RD, Brinster RL. Complete

reconstitution of mouse liver with xenogeneic hepatocytes. Proc

Natl Acad Sci USA 1995;92:4942�6.

[93] Azuma H, Paulk N, Ranade A, Dorrell C, Al-Dhalimy M, Ellis E,

et al. Robust expansion of human hepatocytes in Fah2/2/Rag22/

2/Il2rg2/2 mice. Nat Biotechnol 2007;25:903�10. Available

from: https://doi.org/10.1038/nbt1326.

[94] Hasegawa M, Kawai K, Mitsui T, Taniguchi K, Monnai M,

Wakui M, et al. The reconstituted ‘humanized liver’ in TK-NOG

mice is mature and functional. Biochem Biophys Res Commun

2011;405:405�10. Available from: https://doi.org/10.1016/j.

bbrc.2011.01.042.

[95] Bility MT, Zhang L, Washburn ML, Curtis TA, Kovalev GI, Su

L. Generation of a humanized mouse model with both human

immune system and liver cells to model hepatitis C virus infection

750 PART | ELEVEN Gastrointestinal system

Page 15: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

and liver immunopathogenesis. Nat Protoc 2012;7:1608�17.

Available from: https://doi.org/10.1038/nprot.2012.083.

[96] Bucher NLR, Scott JF, Aub JC. Regeneration of the liver in para-

biotic rats. Cancer Res 1951;11:457�65.

[97] Ponder KP, Gupta S, Leland F, Darlington G, Finegold M,

DeMayo J, et al. Mouse hepatocytes migrate to liver parenchyma

and function indefinitely after intrasplenic transplantation. Proc

Natl Acad Sci USA 1991;88:1217�21.

[98] Chen AA, Thomas DK, Ong LL, Schwartz RE, Golub TR,

Bhatia SN. Humanized mice with ectopic artificial liver tissues.

Proc Natl Acad Sci USA 2011;108:11842�7. Available from:

https://doi.org/10.1073/pnas.1101791108.

[99] Bianconi E, Piovesan A, Facchin F, Beraudi A, Casadei R,

Frabetti F, et al. An estimation of the number of cells in the

human body. Ann Hum Biol 2013;40:463�71. Available from:

https://doi.org/10.3109/03014460.2013.807878.

[100] Guillouzo A, Corlu A, Aninat C, Glaise D, Morel F, Guguen-

Guillouzo C. The human hepatoma HepaRG cells: a highly dif-

ferentiated model for studies of liver metabolism and toxicity of

xenobiotics. Chem Biol Interact 2007;168:66�73.

[101] Cheng Z, He Z, Cai Y, Zhang C, Fu G, Li H, et al. Conversion

of hepatoma cells to hepatocyte-like cells by defined hepatocyte

nuclear factors. Cell Res 2018;1. Available from: https://doi.org/

10.1038/s41422-018-0111-x.

[102] Tsuruga Y, Kiyono T, Matsushita M, Takahashi T, Kasai H,

Matsumoto S, et al. Establishment of immortalized human hepa-

tocytes by introduction of HPV16 E6/E7 and hTERT as cell

sources for liver cell-based therapy. Cell Transplant

2008;17:1083�94.

[103] Peng WC, Logan CY, Fish M, Anbarchian T, Aguisanda F,

Alvarez-Varela A, et al. Inflammatory cytokine TNFα promotes

the long-term expansion of primary hepatocytes in 3D culture.

Cell 2018;175:1607�1619.e15. Available from: https://doi.org/

10.1016/j.cell.2018.11.012.

[104] Hu H, Gehart H, Artegiani B, LOpez-Iglesias C, Dekkers F,

Basak O, et al. Long-term expansion of functional mouse and

human hepatocytes as 3D organoids. Cell 2018;175:1591�1606.

e19. Available from: https://doi.org/10.1016/j.cell.2018.11.013.

[105] Si-Tayeb K, Noto FK, Nagaoka M, Li J, Battle MA, Duris C,

et al. Highly efficient generation of human hepatocyte�like cells

from induced pluripotent stem cells. Hepatology

2010;51:297�305.

[106] Rambhatla L, Chiu C-P, Kundu P, Peng Y, Carpenter MK.

Generation of hepatocyte-like cells from human embryonic stem

cells. Cell Transplant 2003;12:1�11.

[107] Cai J, Zhao Y, Liu Y, Ye F, Song Z, Qin H, et al. Directed dif-

ferentiation of human embryonic stem cells into functional

hepatic cells. Hepatology 2007;45:1229�39.

[108] Song Z, Cai J, Liu Y, Zhao D, Yong J, Duo S, et al. Efficient

generation of hepatocyte-like cells from human induced pluripo-

tent stem cells. Cell Res 2009;19:1233.

[109] Schwartz RE, Fleming HE, Khetani SR, Bhatia SN. Pluripotent

stem cell-derived hepatocyte-like cells. Biotechnol Adv

2014;32:504�13.

[110] Huch M, Gehart H, van Boxtel R, Hamer K, Blokzijl F,

Verstegen MMA, et al. Long-term culture of genome-stable

bipotent stem cells from adult human liver. Cell

2015;160:299�312. Available from: https://doi.org/10.1016/j.

cell.2014.11.050.

[111] Zhu S, Rezvani M, Harbell J, Mattis AN, Wolfe AR, Benet LZ,

et al. Mouse liver repopulation with hepatocytes generated from

human fibroblasts. Nature 2014;508:93.

[112] Huang P, Zhang L, Gao Y, He Z, Yao D, Wu Z, et al. Direct

reprogramming of human fibroblasts to functional and expand-

able hepatocytes. Cell Stem Cell 2014;14:370�84.

[113] Du Y, Wang J, Jia J, Song N, Xiang C, Xu J, et al. Human hepa-

tocytes with drug metabolic function induced from fibroblasts by

lineage reprogramming. Cell Stem Cell 2014;14:394�403.

[114] Wells RG. Cellular sources of extracellular matrix in hepatic

fibrosis. Clin Liver Dis 2008;12:759�68. Available from: https://

doi.org/10.1016/j.cld.2008.07.008 viii.

[115] Glicklis R, Shapiro L, Agbaria R, Merchuk JC, Cohen S.

Hepatocyte behavior within three-dimensional porous alginate

scaffolds. Biotechnol Bioeng 2000;67:344�53 3, 344::AID-

BIT11. 3.0.CO;2�2 [pii].

[116] Ranucci CS, Kumar A, Batra SP, Moghe PV. Control of hepato-

cyte function on collagen foams: sizing matrix pores toward

selective induction of 2D and 3D cellular morphogenesis.

Biomaterials 2000;21:783�93.

[117] Semino CE, Merok JR, Crane GG, Panagiotakos G, Zhang S.

Functional differentiation of hepatocyte-like spheroid structures

from putative liver progenitor cells in three-dimensional peptide

scaffolds. Differentiation 2003;71:262�70. Available from:

https://doi.org/10.1046/j.1432-0436.2003.7104503.x

S0301�4681(09)60357�1 [pii].

[118] Bruns H, et al. Injectable liver: a novel approach using fibrin gel

as a matrix for culture and intrahepatic transplantation of hepato-

cytes. Tissue Eng 2005;11:1718�26.

[119] Fan J, Shang Y, Yuan Y, Yang J. Preparation and characteriza-

tion of chitosan/galactosylated hyaluronic acid scaffolds for pri-

mary hepatocytes culture. J Mater Sci Mater Med

2010;21:319�27.

[120] Kasai S, et al. Cellulose microcarrier for high-density culture of

hepatocytes. Transplant Proc 1992;24:2933�4.

[121] Uygun BE, et al. Organ reengineering through development of a

transplantable recellularized liver graft using decellularized liver

matrix. Nat Med 2010;16:814�20. Available from: https://doi.

org/10.1038/nm.2170.

[122] Demetriou AA, et al. Replacement of liver function in rats by

transplantation of microcarrier-attached hepatocytes. Science

1986;233:1190�2.

[123] Kobayashi N, et al. Development of a cellulose-based microcar-

rier containing cellular adhesive peptides for a bioartificial liver.

Transplant Proc 2003;35:443�4. Available from: https://doi.org/

10.1016/s0041-1345(02)03783-1 [pii].

[124] Tao X, Shaolin L, Yaoting Y. Preparation and culture of hepato-

cyte on gelatin microcarriers. J Biomed Mater Res A

2003;65:306�10.

[125] Li K, et al. Chitosan/gelatin composite microcarrier for hepato-

cyte culture. Biotechnol Lett 2004;26:879�83 5274932 [pii].

[126] 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 2011;53:604�17.

Available from: https://doi.org/10.1002/hep.24067.

[127] Soto-Gutierrez A, Zhang L, Medberry C, Fukumitsu K, Faulk D,

Jiang H, et al. A whole-organ regenerative medicine approach

for liver replacement. Tissue Eng, C: Methods 2011;17:677�86.

Available from: https://doi.org/10.1089/ten.tec.2010.0698.

Hepatic tissue engineering Chapter | 40 751

Page 16: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

[128] Mazza G, Rombouts K, Rennie Hall A, Urbani L, Vinh Luong T,

Al-Akkad W, et al. Decellularized human liver as a natural 3D-

scaffold for liver bioengineering and transplantation. Sci Rep

2015;5:13079. Available from: https://doi.org/10.1038/

srep13079.

[129] Ogiso S, Yasuchika K, Fukumitsu K, Ishii T, Kojima H,

Miyauchi Y, et al. Efficient recellularisation of decellularised

whole-liver grafts using biliary tree and foetal hepatocytes. Sci

Rep 2016;6:35887. Available from: https://doi.org/10.1038/

srep35887.

[130] Cima LG, Ingber DE, Vacanti JP, Langer R. Hepatocyte culture

on biodegradable polymeric substrates. Biotechnol Bioeng

1991;38:145�58. Available from: https://doi.org/10.1002/

bit.260380207.

[131] Chua KN, et al. Stable immobilization of rat hepatocyte spher-

oids on galactosylated nanofiber scaffold. Biomaterials

2005;26:2537�47. Available from: https://doi.org/10.1016/j.bio-

materials.2004.07.040 S0142�9612(04)00681�7 [pii].

[132] Mooney DJ, et al. Biodegradable sponges for hepatocyte trans-

plantation. J Biomed Mater Res 1995;29:959�65.

[133] Mooney DJ, et al. Long-term engraftment of hepatocytes trans-

planted on biodegradable polymer sponges. J Biomed Mater Res

1997;37:413�20 3, 413::AID-JBM12. 3.0.CO;2-C [pii].

[134] Itle LJ, Koh WG, Pishko MV. Hepatocyte viability and protein

expression within hydrogel microstructures. Biotechnol Prog

2005;21:926�32.

[135] Underhill GH, Chen AA, Albrecht DR, Bhatia SN. Assessment

of hepatocellular function within PEG hydrogels. Biomaterials

2007;28:256�70. Available from: https://doi.org/10.1016/j.bio-

materials.2006.08.043 S0142-9612(06)00760-5 [pii].

[136] Smith MK, Peters MC, Richardson TP, Garbern JC, Mooney DJ.

Locally enhanced angiogenesis promotes transplanted cell sur-

vival. Tissue Eng 2004;10:63�71.

[137] Houchin ML, Topp EM. Chemical degradation of peptides and

proteins in PLGA: a review of reactions and mechanisms. J

Pharm Sci 2008;97:2395�404.

[138] Peppas NA, Bures P, Leobandung W, Ichikawa H. Hydrogels in

pharmaceutical formulations. Eur J Pharm Biopharm

2000;50:27�46.

[139] Hasirci V, et al. Expression of liver-specific functions by rat

hepatocytes seeded in treated poly(lactic-co-glycolic) acid biode-

gradable foams. Tissue Eng 2001;7:385�94.

[140] Nam YS, Yoon JJ, Lee JG, Park TG. Adhesion behaviors of

hepatocytes cultured onto biodegradable polymer surface modi-

fied by alkali hydrolysis process. J Biomater Sci Polym Ed

1999;10:1145�58.

[141] Karamuk E, Mayer J, Wintermantel E, Akaike T. Partially

degradable film/fabric composites: textile scaffolds for liver cell

culture. Artif Organs 1999;23:881�4. Available from: https://

doi.org/10.1046/j.1525-1594.1999.06308.x [pii].

[142] Lee JS, Kim SH, Kim YJ, Akaike T, Kim SC. Hepatocyte adhesion

on a poly[N-p-vinylbenzyl-4-O-beta-D-galactopyranosyl-D-glucoa-

mide]-coated poly(L-lactic acid) surface. Biomacromolecules

2005;6:1906�11.

[143] Catapano G, Di Lorenzo MC, Della Volpe C, De Bartolo L,

Migliaresi C. Polymeric membranes for hybrid liver support

devices: the effect of membrane surface wettability on

hepatocyte viability and functions. J Biomater Sci Polym Ed

1996;7:1017�27.

[144] Fiegel HC, et al. Influence of flow conditions and matrix coat-

ings on growth and differentiation of three-dimensionally cul-

tured rat hepatocytes. Tissue Eng 2004;10:165�74.

[145] Gutsche AT, Lo H, Zurlo J, Yager J, Leong KW. Engineering of

a sugar-derivatized porous network for hepatocyte culture.

Biomaterials 1996;17:387�93. Available from: https://doi.org/

10.1016/0142-9612(96)85577-3 [pii].

[146] Kim M, Lee JY, Jones CN, Revzin A, Tae G. Heparin-based

hydrogel as a matrix for encapsulation and cultivation of primary

hepatocytes. Biomaterials 2010;31:3596�603. Available from:

https://doi.org/10.1016/j.biomaterials.2010.01.068 S0142-9612

(10)00100-6 [pii].

[147] Park TG. Perfusion culture of hepatocytes within galactose-

derivatized biodegradable poly(lactide-co-glycolide) scaffolds

prepared by gas foaming of effervescent salts. J Biomed Mater

Res 2002;59:127�35.

[148] Li J, Pan J, Zhang L, Yu Y. Culture of hepatocytes on fructose-

modified chitosan scaffolds. Biomaterials 2003;24:2317�22.

Available from: https://doi.org/10.1016/S0142-9612(03)00048-6.

[149] Mayer J, Karamuk E, Akaike T, Wintermantel E. Matrices for

tissue engineering-scaffold structure for a bioartificial liver sup-

port system. J Control Release 2000;64:81�90.

[150] Carlisle ES, et al. Enhancing hepatocyte adhesion by pulsed

plasma deposition and polyethylene glycol coupling. Tissue Eng

2000;6:45�52.

[151] Stevens KR, Miller JS, Blakely BL, Chen CS, Bhatia SN.

Degradable hydrogels derived from PEG-diacrylamide for

hepatic tissue engineering. J Biomed Mater Res A 2015;103

(10):3331�8.

[152] Oates M, Chen R, Duncan M, Hunt JA. The angiogenic potential

of three-dimensional open porous synthetic matrix materials.

Biomaterials 2007;28:3679�86. Available from: https://doi.org/

10.1016/j.biomaterials.2007.04.042 S0142�9612(07)00364-X

[pii].

[153] Yang TH, Miyoshi H, Ohshima N. Novel cell immobilization

method utilizing centrifugal force to achieve high-density hepa-

tocyte culture in porous scaffold. J Biomed Mater Res

2001;55:379�86 3, 379::AID-JBM1026. 3.0.CO;2-Z [pii].

[154] Lee KY, Peters MC, Anderson KW, Mooney DJ. Controlled

growth factor release from synthetic extracellular matrices.

Nature 2000;408:998�1000.

[155] Song H-HG, Rumma RT, Ozaki CK, Edelman ER, Chen CS.

Vascular tissue engineering: progress, challenges, and clinical

promise. Cell Stem Cell 2018;22:340�54. Available from:

https://doi.org/10.1016/j.stem.2018.02.009.

[156] Takebe T, Koike N, Sekine K, Fujiwara R, Amiya T, Zheng Y-

W, et al. Engineering of human hepatic tissue with functional

vascular networks. Organogenesis 2014;10:260�7. Available

from: https://doi.org/10.4161/org.27590.

[157] Davis GE, Speichinger KR, Norden PR, Kim DJ, Bowers SLK.

Endothelial Cell Polarization During Lumen Formation,

Tubulogenesis, and Vessel Maturation in 3D Extracellular

Matrices. Cell Polarity 1. Cham: Springer International

Publishing; 2015. p. 205�20. Available from: http://doi.org/

10.1007/978-3-319-14463-4_9.

752 PART | ELEVEN Gastrointestinal system

Page 17: Hepatic tissue engineering › sites › default › files › images › ... · the case of α1-antitrypsin ZZ deficiency, hemochromato-sis, Wilson’s disease, and hereditary tyrosinemia

[158] Montesano R, Vassalli JD, Baird A, Guillemin R, Orci L, Galie

PA, et al. Basic fibroblast growth factor induces angiogenesis

in vitro. Proc Natl Acad Sci USA 1986;83:7297�301. Available

from: https://doi.org/10.1073/pnas.83.19.7297.

[159] Song KH, Highley CB, Rouff A, Burdick JA. Complex 3D-

printed microchannels within cell-degradable hydrogels. Adv

Funct Mater 2018;28:1801331. Available from: https://doi.org/

10.1002/adfm.201801331.

[160] Galie PA, Nguyen D-HT, Choi CK, Cohen DM, Janmey PA,

Chen CS. Fluid shear stress threshold regulates angiogenic

sprouting. Proc Natl Acad Sci USA 2014;111:7968�73.

Available from: https://doi.org/10.1073/pnas.1310842111.

[161] Park KM, Gerecht S. Hypoxia-inducible hydrogels. Nat Commun

2014;5:4075. Available from: https://doi.org/10.1038/ncomms5075.

[162] Borenstein JT, et al. Microfabrication technology for vascular-

ized tissue engineering. Biomed Microdevices 2002;4:167�75.

[163] Truby RL, Lewis JA. Printing soft matter in three dimensions.

Nature 2016;540:371�8. Available from: https://doi.org/10.1038/

nature21003.

[164] Kolesky DB, Truby RL, Gladman AS, Busbee TA, Homan KA,

Lewis JA. 3D bioprinting of vascularized, heterogeneous cell-

laden tissue constructs. Adv Mater 2014;26:3124�30. Available

from: https://doi.org/10.1002/adma.201305506.

[165] Kolesky DB, Homan KA, Skylar-Scott MA, Lewis JA. Three-

dimensional bioprinting of thick vascularized tissues. Proc Natl

Acad Sci USA 2016;113:3179�84. Available from: https://doi.

org/10.1073/pnas.1521342113.

[166] Miller JS, Stevens KR, Yang MT, Baker BM, Nguyen D-HT,

Cohen DM, et al. Rapid casting of patterned vascular networks

for perfusable engineered three-dimensional tissues. Nat Mater

2012;11:768�74. Available from: https://doi.org/10.1038/

nmat3357.

[167] Lee H, et al. Local delivery of basic fibroblast growth factor

increases both angiogenesis and engraftment of hepatocytes in

tissue-engineered polymer devices. Transplantation

2002;73:1589�93.

[168] Richardson TP, Peters MC, Ennett AB, Mooney DJ. Polymeric

system for dual growth factor delivery. Nat Biotechnol

2001;19:1029�34.

[169] Kedem A, et al. Vascular endothelial growth factor-releasing

scaffolds enhance vascularization and engraftment of hepatocytes

transplanted on liver lobes. Tissue Eng 2005;11:715�22.

[170] Griffith LG, et al. In vitro organogenesis of liver tissue. Ann NY

Acad Sci 1997;831:382�97.

[171] Racanelli V, Rehermann B. The liver as an immunological

organ. Hepatology 2006;43:S54�62.

[172] Sampaziotis F, De Brito MC, Madrigal P, Bertero A, Saeb-Parsy

K, Soares FAC, et al. Cholangiocytes derived from human

induced pluripotent stem cells for disease modeling and drug val-

idation. Nat Biotechnol 2015;33:845�52. Available from:

https://doi.org/10.1038/nbt.3275.

[173] Sampaziotis F, Justin AW, Tysoe OC, Sawiak S, Godfrey EM,

Upponi SS, et al. Reconstruction of the mouse extrahepatic bili-

ary tree using primary human extrahepatic cholangiocyte orga-

noids. Nat Med 2017;23:954�63. Available from: https://doi.org/

10.1038/nm.4360.

[174] Sudo R, Mitaka T, Ikeda M, Tanishita K. Reconstruction of 3D

stacked-up structures by rat small hepatocytes on microporous

membranes. FASEB J 2005;19:1695�7.

[175] Ishida Y, et al. Ductular morphogenesis and functional polariza-

tion of normal human biliary epithelial cells in three-dimensional

culture. J Hepatol 2001;35:2�9.

[176] Auth MK, et al. Morphogenesis of primary human biliary epithe-

lial cells: induction in high-density culture or by coculture with

autologous human hepatocytes. Hepatology 2001;33:519�29.

[177] Miyazawa M, et al. A tissue-engineered artificial bile duct grown

to resemble the native bile duct. Am J Transplant

2005;5:1541�7.

[178] Du Y, et al. A bile duct-on-a-chip with organ-level functions.

Biorxiv 2019; http://dx.doi.org/10.1002/hep.30918.

Hepatic tissue engineering Chapter | 40 753