Sie sind auf Seite 1von 16

 

Volume 26, Issue 6, Pages 1379-1392 (July 1997)

In vitro evaluation of a novel bioreactor based on an


integral oxygenator and a spirally wound nonwoven
polyester matrix for hepatocyte culture as small aggregates
Leonard M. Flendrig, John W. la Soe, George G.A. Jörning,
Arie Steenbeek, Ole T. Karlsen, Wim M.M.J. Bovée, Nita C.J.J. Ladiges,
Anje A. te Velde, Robert A.F.M. Chamuleau

Received 7 May 1996; received in revised form 18 December 1996; accepted 18 December
1996.

Abstract

Background/Aims: The development of custom-made bioreactors for use as a bioartificial


liver (BAL) is considered to be one of the last challenges on the road to successful temporary
extracorporeal liver support therapy. We devised a novel bioreactor (patent pending) which
allows individual perfusion of high density cultured hepatocytes with low diffusional
gradients, thereby more closely resembling the conditions in the intact liver lobuli.

Methods: The bioreactor consists of a spirally wound nonwoven polyester matrix, i.e. a sheet-
shaped, three-dimensional framework for hepatocyte immobilization and aggregation, and of
integrated hydrophobic hollow-fiber membranes for decentralized oxygen supply and CO2
removal. Medium (plasma in vivo) was perfused through the extrafiber space and therefore in
direct hepatocyte contact. Various parameters were assessed over a period of 4 days including
galactose elimination, urea synthesis, lidocaine elimination, lactate/pyruvate ratios, amino
acid metabolism, pH, the last day being reserved exclusively for determination of protein
secretion.

Results: Microscopic examination of the hepatocytes revealed cytoarchitectural characteristics


as found in vivo. The biochemical performance of the bioreactor remained stable over the
investigated period. The urea synthesizing capacity of hepatocytes in the bioreactor was twice
that of hepatocytes in monolayer cultures. Flow sensitive magnetic resonance imaging (MRI)
revealed that the bioreactor construction ensured medium flow through all parts of the device
irrespective of its size.
Conclusions: The novel bioreactor showed encouraging efficiency. The device is easy to
manufacture with scale-up to the liver mass required for possible short-term support of
patients in hepatic failure.

Keywords: Aggregates, Bioartificial liver, Bioreactor, 3D-matrix, Hepatocytes, Liver


support, MRI, Oxygenator, Polyester
No full text is available. To read the body of this article, please view the PDF online.

Department of Experimental Internal Medicine, University of Amsterdam, Academic


Medical Center, Amsterdam, The Netherlands

Department of Technical Research and Development, University of Amsterdam,


Academic Medical Center, Amsterdam, The Netherlands

Department of Physics, University of Delft, The Netherlands

Correspondence: Leonard M. Flendrig, Academic Medical Center, Department of


Experimental Internal Medicine G2-130, PO Box 22700, 1100 DE Amsterdam, The
Netherlands. Tel: (0) 20-5665910. Fax: (0) 20-6977192 (or 5664440).

PII: S0168-8278(97)80475-8

© 1997 Published by Elsevier Inc.

 
AFSENDT ~ - a51961/he6018Oa

- ~ JAN 199i

Journa/ of Hepal% gy 1997; 26: 000-00O Copy right 0 Europemr Assorimion


Printed in Denmark . All rights reserved for the Stud)" of the Liv., 1997
Munksgaard' Copenhagen
Journal of Hepato1ogy
ISSN 0168-8278

In vitro evaluation of a novel bioreactor based on an integral oxygenator


and aspirally wound nonwoven polyester matrix for hepatocyte culture
as small aggregates
Leonard M. Flendrig 1, John W. la Soe 1, George G . A. Jörning l , Arie Steenbeek2 , Ole T. Karlsen 3 ,
Wim M. M. 1. Bovée 3 , Nita C. 1. 1. Ladiges l , Anje A. te Veldel and Robert A. F. M. Chamuleau l
I Department of Experimenta/ Interna/ Medicine. 2Department of Technica/ Research and Deve/opment. Vniversity of Amsterdam. Academie

Medica/ Center. Amsterdam. and the. 3Department of Physics. Vniversity of Delft. The Nether/ands

Background!Aims: The development of custom made last day being reserved exclusively for determination

bioreactors for use as a bioartificial üver (BAL) is of protein secretion.

considered to be one of the last cbaUenges on tbe road Results: Micro~opic examination of the bepatocytes

to successful temporary extracorporeal üver support revealed cytoarchitectural characteristics as found in

tberapy. \\é devised a novel bioreactor (patent pend­ .,i.,o. The biochemical performance of tbe bioreactor

ing) wbich allows individual perfusion of high density remained stabie over tbe investigated period. The ure­

cultured bepatocytes witb low diffusional gradients, asynthesizing capacity of bepatocytes in the bioreac­

tbereby more closely resembüng tbe conditions in the tor was twice that of bepatocytes in monolayer cul­

intact üver lobuli. tures. Flow sensitive magnetic resonance imaging

Methods: The bioreactor consists of aspirally wound (MRn revealed that the bioreactor construction en­

nonwoven polyester matrix, i.e. a sbeet-shaped, th ree­ sured medium flow through aU parts of the device ir­

dimensional framework for bepatocyte immobiüzation respective of its size.

and aggregation, and of integrated hydrophobic hol­ Conclusions: The novel bioreactor showed encourag­

low-fiber membranes for decentraüzed oxygen supply ing efficiency. The device is easy to manufacture ~itb

and CO2 removal. Medium (plasma ;n vivo ) was per­ scale-up to the liver mass required for possible short­

fused through the extrafiber space and therefore in term support of patients in hepatic failure.

direct hepatocyte contact. Various parameters were


assessed over a period of 4 days including galactose Key words: Aggregates; Bioartificial üver; Bioreactor;

eümination, urea synthesis, lidocaine eümination, lac­ 3D-matrix; Hepatocytes; Liver support; MRI; Oxy­

tate/pyruvate ratios, amino acid metaboüsm, pH, the genator; Polyester.

Tm DEVELOPMENT of a liver support system for the been reported , none achieved long-term survival (3,4) .
.1 treatment of patients with fulminant hepatic fail­ It was therefore concluded that an effective liver sup­
ure and as a bridge to liver transplantation is a major port system should be able to perform the li"er's
challenge. Many early attempts focused on blood de­ multiple synthetic and metabolic functions, including
toxification based on the assumption that liver failure detoxification and excretion. The most logical ap­
could be reversed if the associated toxins were removed proach to this problem is the introduction of active
from the circulation of the patient (1 ,2). Although im­ functioning hepatocytes (5). The state-of-the-art em­
provement of the neurological status of patients has bodiment of this theory is presented in the bioartificial
liver (BAL), an extracorporeal device comprising weil
Received 7 May: revised /8 December: accepted /8 December /996 nourished and oxygenated via bie hepatocytes inunobil­
Correspondence: Leonard M. Flendrig, Academic Medical
ized on a mechanical support and separated from the
Center, Department of Experimental Internal Medicine
blood circulation by semipermeable membranes.
G2-l30, PO Box 22700, 1100 DE Amsterdam,
Different bioreactor systems are currently under in­
The Netherlands.
vestigation. They can be classified according to the im­
L. M. Flendrig et al.

mobilization technique used: artificial substrates, such


as glass plates (6), microcarriers (7,8), hollow-fiber
membranes (9-13), biological matriees (14--17), encap­
sula tion (l8,19) and three-dimensional carrier ma­
terials (20). Objectives like biocompatibility, mainten­
anee of functional capacity and practicality, important
aspects in the development of a BAL, have been dis­
cussed in recent reviews (12,21-24). However, an ideal
BAL system has not yet been invented. In this respect,
the impact of bioreactor construction on hepatocyte
function has been undervalued. Every hepatocyte in
the intact liver functions under perfusion conditions
and close blood contact. The majority of the current
bioreactor designs do not meet these conditions, which
D
are essential for optimal substrate and metabolite ex­ B c
change to hepatocytes. This often results in non­
physiological gradients, which impair the metabolic ac­
tivity of the cultured eells.
Therefore, an important aim of this study was to
develop a bioreactor configuration that allows high
density hepatocyte culture and simultaneously ensures
@
I"
·tI
elF
~ -=[;;;: I IJ 1;;;; {lJlJt>>>

E
'" E
Fig. 1. Schematic drawing of a transverse and longitudinal
that every hepatocyte operates under in vivo like per­
cross-section of the bioreactor. The system is composed ofa
fusion conditions and direct medium contact. In ad­
polysulfon dialysis housing (A) comprising a three-dimen­
dition, we wanted to culture hepatocytes as small ag­ sional nonwoven polyester matrix (B) for high density he­
gregates, known to maintain many of the cytoarchitec­ patocyte culture as small aggregates and hydrophobic poly­
tural characteristics found in vivo and to exhibit higher propylene hollow-fiber membranes (C) for oxygen supply
and more prolonged functional activity than hepato­ and CO 2 removal. Medium is perfused through the extrafiber
cytes cultured in monolayers (25--27). Special attention bioreactor space via the side ports (F) and is in direct cell
was paid to optimal oxygenation of the cells, since oxy­ contact. Despite high density culturing there is sufficient
gen plays an important role in hepatocyte attachment space between the aggregatesfor unhindered transport ofme­
(28) and function (29,30). Therefore, the bioreactor dium (plasma in vivo) to and from the hepatocytes. The bi­
was equipped with an integrated oxygenator. This en­ oreactor is perfusedwirh culture gas via theendcaps (E). The
spirally wound polyeszer matrix guarantees a homogeneous
ables on site oxygenation of the hepatocytes with low
distribution of the oxygenation hollow-fibers throughout the
gradients, irrespective of the plasma perfusion rate,
bioreactor compartment, thereby ensuring every hepatocy -".
which can not be realized with a separate, external oxy­ of an oxygenation souree within its direct surroundings.
genator as found in most BAL systems. Another goal Moreover, the hollOlI'-fibers act as spacers between the layers
was to develop a bioreactor that could be scaled-up to of rhe 3D-matrix, creating numerous channels (D) which
incorporate sufficient cell mass for possible therapeutic form a defined space for uniformflow and distribution ofme­
liver support. These aspects resulted in a novel bioreac­ dium to all parts of the solid support.
tor comprising a spirally wound 3D non woven poly­
ester matrix with integrated oxygenation hollow fiber
membranes (Fig. 1) in which hepatocytes immobilize of Dermatology, and alocal slaughterhouse. The
and spontaneously reorganize as small aggregates. hepatocytes were isolated from pigs with a body ma ss
In this article, we present the characteristics of our of 20-25 kg using a simple two-step collagenase per­
novel culture device and the results of its assessment in fusion technique as descri bed previously (31). The vi­
vitro. The bioreactors were investigated over a period ability of the isolated eells, based on trypan blue ex­
of 3 days, as their future application as a BAL will be clusion, varied from 71 to 96% (n=8, mean 89%). The
shortly after hepatocyte immobilization. yield varied from 8· 106 to 30· 106 hepatocytes per g
wet liver weight.
Materials and Methods
Hepatocyte isolation Bioreactor
Pig livers were kindly provided by the Department of The bioreactor (patent pending) consists of a 3D no'- '--""
Clinical and Experimental Cardiology, the Department woven polyester matrix specifically designed for cultUI­

2
Navel bioreactor

ing anchorage-dependent cells (Fibra Cell, Bibby Steri­ sity wiJl increase. Fluid flow at higher velocities than 2
lin Ltd, Stone, Staffordshire, UK) and hydrophobic cmls wiJl not result in an increased signa!, as the detec­
polypropylene hoJlow fiber membranes donated by Dr. tion sliee is then completely refreshed. Therefore, the
J. Vienken of AKZO-NOBEL (plasmaphan, AKZO­ flow was calibrated such that the maximum fluid vel­
NOBEL, Wuppertal, Germany) for oxygenation and ocity in most flow channels (D in Fig. I) did not exeeed
carbon dioxide removal. The 3D matrix (dimensions: 2 cmls.
length 140 mm, width 90 mm, thickness 0.5 mm, fiber
diameter 13 /lm) provides a scaffold for hl.:patocyte im­ Hepatocyte culture
mobilization and self-aggregation. lts surface for Hepatocytes suspended in ice-cold Williams' E me­
attachment is about 15 times its projected area, which dium (Gibco BRL Life Technologies, European Divi­
enables high density hepatocyte culture. The oxygen­ sion) supplemented with heat inactivated FCS (10%,
ation hollow-fibers (external diameter 630 /1ffi, internal Boehringer Mannheim), glutamine (2 mM, BDH Lab­
diameter 300 /lm) are fixed to the 3D-carrier in a paral­ oratory Supplies Ltd.), insulin (20 miE, Novo Nordisk,
lel fashion by weaving, spaced at an average distance Denmark), dexamethasone (l/lM) and antibioticlanti­
of 2 mmo This polyester-polypropylene composite is mycotic solution (Gibco) at a concentration of 20· 106
spirally wound like a Swiss roil with the help of an viabie eells per mi were injected into two precooled
acrylic core (Fig. I) and placed in a polysulfon dialysis WC) dry bioreactors until each of the units contained
housing (Minifilter, Amicon Ltd, Ireland, ID 1.32 cm, 220· 106 ceJls per unit. The cooled bioreactors were in­
ED 1.7 cm, totallength 15.5 cm). The oxygenation hol­ tegrated into two separate eeJl perfusion circuits to ob­
low-fibers are embedded in polyurethane resin (pUR­ tain results in duplicate. The whole apparatus was put
system 725 A and 725 BF, Morton International, Bre­ in a temperature regulated (37°C) cabinet (Stuart
men, Germany) using dialyzer potting techniques and Scientific, model SI60, GB) where the bioreactors were
fitted with gas inlet and outlet endcaps. The bioreactor clamped onto a custom made rotation device and con­
is sterilized by autoclaving (20 min at 121°C). Hepato­ nected to "culture gas" (95% air and 5% CO 2 , flow
cyte seeding in the extrafiber space (volume II mi, suit­ rate: 30 mI/min). The reactors were rotated horizon­
abIe for in vivo experiments in the rat) is achieved by taJly along their longitudinal axis at 1 revolutionlmin
injecting the cell suspension via the side ports normaUy for a period of 120 min to secure an even distribution
used for dialysate flow. The same ports are used for of the ceJls throughout the reactor and to accelerate
medium perfusion after ceU immobilization. immobilization by entrapment, attachment, and self­
aggregation of via bie hepatocytes. After tbis immobil­
Flow sensitive Magnetic Resonance lmaging ization period, old medium was replaced by fresh me­
The flow distribution in a cross-section of a smaJl (lD dium (60 mi) intermittently for 15 h to flush dead and
1.32 cm, volume 11 mi, 46 hoJlow-fiber membranes, unattached eeUs out of the reactor, to supply nutrients
diameter acrylic core 0.4 cm) and a scaled-up bioreac­ to and to remove toxins from the eeUs, and to allow
tor (ID 2.2 cm, volume 33 mi, 138 hollow-fiber mem­ the hepatocytes to recover from the isolation pro­
branes, diameter acrylic core 0.4 cm) of equal length cedure. The devices were then considered to be ready
was investigated. The bioreactors were first flushed for use.
with ethanol and subsequently water to remove air
bubbles, which can block the medium flow and/or can Hepatocyte function tests
distort the homogeneity of the magnetic field, resulting General description. Bioreactors with and without
in a decreased signal intensity. A bioreactor was then hepatocytes were studied. Those without hepatocytes
placed in a birdcage coil and positioned horizontally served as controls. Both groups received identical treat­
in a 6.3 Tesla/20 cm bore home-built spectrometer. ment and monitoring. Hepatocyte function tests were
Cell-free devices were used as the spectrometer was not performed while supplemented Williarns' E medium
equipped to support viabie hepatocytes. Transaxial (30 mi) was recirculated through the extrafiber bioreac­
flow sensitive MRI's were taken from the middle of the tor spaee at a flow rate of 5 mI/min. Various par­
bioreactor using a novel per fusion imaging technique ameters were assessed over a period of 4 days, the last
(32). Briefly, the water signal in a detection slice (width day being reserved exclusively for determination of
2 mm, perpendicular to the flow direction) is sup­ protein secretion under serum free conditions. On
pressed. During an in-flow time of 100 ms, part of the every day during the first 3 days a battery of tests was
slice is refreshed, resulting in an increase in signa I in­ carried out including, galactose elimination, urea syn­
tensity. Thus, the higher the flow, and the more the thesis, lidocaine metabolism, and a subsequent 14-h in­
detection slice is refreshed, the more the signal inten- cubation with sup plemented Williams' E medium to

3
L. M. Flendrig el al.

evaluate the amino acid metabolism, lactate/pyruvate standard solution (EMGX 5 lig/mi in distilled water)
ratio, enzyme leakage, glucose levels, and pH. Every and 150 lil distilled water to a 150 lil sample. Analysis
test was preceded by a fresh medium waste wash. of the much higher lidocaine concentrations required a
Samples collected from the closed loop circuit were 20-fold dilution of the sample in supplemented Willi­
snap frozen in liquid nitrogen and stored at -70°C ams' E medium. The isolation of lidocaine and its
prior to analysis. It was not feasible to quantify the metabolites was performed by extraction. For th is, 150
hepatocytes in the device by evaluating the total pro­ lil sodium carbonate (0.1 M) and 600 lil chloroform
tein and/or DNA content, as the ceUs were too en­ were added to the sample preparation. After 1 min vor­
trapped within the nonwoven polyester matrix for total texing and 4 min centrifugation at 8000 rpm the aque­
harvesting. Therefore, quantification relied on micro­ ous supernatant was removed and 150 lil distilled water
scopic cell counting before inoculation. and 350 lil HCI (0.1 M) were added to the organic
Galactose elimination. D-Galactose (Sigma Chemi­ phase. The vortexing and centrifugation procedures
cal Co., St Louis, MO) was administered to the closed were repeated and the supernatant removed. A cooled
loop circuit at a concentration of 1 mg/ml and incu­ sample storage compartment kept the residu es at 4°C
bated for 3 h. Media samples were coUected at different prior to analysis. The mobile phase (0.5 M phosphate
time points every day for 3 days. The galactose concen­ buffer, pH 4.5) was pumped at a flow rate of 1.7 mV
tration was measured at 340 nm (Cobas Bio, Roche, min (Perkin Elmer 250, Norwalk, USA) and pretreat( """"'
Switzerland) using enzymatic test kits (Boehringer by a Quard-column (Superspher 60 RP 8, length 10
Mannheim, Wiesbaden, Germany, kit no. 124273). The cm, 4 J1II1 particles, Bischoff Chromatography, Ger·
amount of galactose eliminated was calculated from many). An auto sampler (Gilson Sample Injector
these data. model 231, France) injected 50 lil aliquots onto a tem­
Urea synthesis from NH4 Cl. The urea-synthesizing perature regulated (55°C, Chrompac Column Thermo­
capacity of hepatocytes cultured in the bioreactor was stat, The Netherlands) HPLC column (Superspher 60
compared to that of hepatocytes in monolayer cultures. RP 8, length 20 cm, i.d. 4.6 mm, 4 lim particles, Bi­
Hepatocytes of the same isolation were seeded in the schoff Chomatography, Germany). Detection was at
bioreactor and on two wells of a 6-weU tissue culture 198 nm (Schoeffel SF 770 UV-spectrophotometer, Ger­
plate (Becton Dickinson La bwa re, MA) at 3· 106 vi­ many) and peak areas were calculated with the aid of
able cells per weil. 10 mM NH 4 Cl was added to the an Olivetti M250 computer utilizing integration soft­
closed loop circuit and the monolayer cultures and in­ ware (Chrompac PCI, version 5.12, The Netherlands).
cubated for 2 h. Media samples were collected at differ­ The samples were quantified by comparing the peak
ent time points every day for 3 days. To quantify the area ratio of the component of interest to that of the
hepatocytes of the monolayer cultures, the DC protein internal standard. Standard curves were obtained for
assay from Bio-Rad (Hercules, CA) was used. Urea ni­ lidocaine (5-80 lig/mi), MEGX (0.5-16 lig/mi), xyli­
trogen was determined colorimetricaUy at 525 nm dine (5-80 lig/mI), and GX (1-32 lig/mi) and showe
(Zeiss MQ3 UV spectrophotometer, Germany) with linearity (r=0.996, n=6). The detection limit was 0.
Sigma Chemical Co. kit no. 535. The amount of urea lig/mi for GX, 0.3 lig/mi for xylidine, 0.2 lig/mi for
synthesized was calculated from these urea nitrogen MEGX, 0.4 lig/mi for EMGX and 0.5 lig/mI for lido­
data. caine. The retention times for these components were
Lidocaine metabolism. Lidocaine-HCl (Sigma) was 2.2 min, 2.4 min, 3.2 min, 4.1 min, and 5.8 min, respec­
administered to the closed loop circuit at a concen­ tively. Column stabilization time Was limited to 20 min
tration of 500 lig/mi and incubated for 1 h. Media by washing with a phosphate-acetonitrile-phosphoric
samples were collected at different time points every acid buffer (50 mM, pH= 1.7) and an acetonitrile solu­
day for 3 days. The samples were analyzed for lido­ tion (distilled water: ACN = 1: 1) to remove the chloro­
caine and three lidocaine metabolites, mono-ethyl-gly­ form peak.
cine-xylidide (MEGX), 2,6-xylidine-HCI, glycine-xyli­ Amino acid metabolism. The metabolic turnover of a
dide (GX), by reversed phase high performance liquid wide range of amino acids was investigated. The amino
chromatography (HPLC). Lidocaine-HCl was ob­ acid concentrations were determined by a fuUy auto­
tained from Sigma Chemica I Co. and MEGX, xylidine, mated precolumn derivatization with o-phthaldial­
GX , and ethyl-methyl-glycine-xylidide (EMGX) were dehyde (OPA), followed by high-performance liquid
gifts from Dr. R. Sandberg of Astra Pain Con trol (Sö­ 'chromatography as described previously (33).
dertälje, Sweden). Lactate/pyruvate ratio. Levels of lactate and pyruv­
Sample preparation for the analysis of MEGX, xyli­ ate were determined at 340 run (Co bas Bio, Roer .........
dine and GX involved addition of an 75 lil internal Switzerland) using erizymatic test kits (Boehringe,

4
Novel bioreaClor

Mannheim Wiesbaden, Germany, lactate kit no. were washed in water, dehydrated in graded ethanols,
149993 and pyruvate kit no. 124982). and embedded in paraffin. Ultrathin (8 j.lm) sections
Enzyme leakage. Lactate dehydrogenase (LDH), glu­ were cut from this block. These were deparaffinated
tamic oxaloacetic transaminase (GOD and glutamic with xylol and stained with hematoxylin-eosin. The
pyruvic transaminase (GPT) levels were measured by preparations were examined under an Olympus Vamox
routine clinical analyzers. light microscope (type AHBT3, Tokyo, Japan) .
Glucose. Glucose levels were measured using glucose Scanning electron microscopy. SEM studies were per­
test strips (hemoglucotest 1-44 R, Boehnnger Mann­ formed after fixation of a 5-day-old hepatocyte culture
heim, Wiesbaden, Germany) and the accessory Reflo­ by flushing one bioreactor with 4% glutaraldehyde in
lux-S readout device. phosphate buffer, pH 7.3 (Fluka Chem A.G., Buchs,
pH. The pH was measured by sampling 1 mi of me­ Switzerland). The bioreactor was cut through in the
dium in a bloodgas syringe (Marz-175, Sherwood middle and one part was dehydrated in graded etha­
Medical, Ireland) which was determined on a bloodgas nols and finally dried in hexamethyldisilazane (Sigma,
analyzer (Radiometer, model ABL 300, Copenhagen). Munich, Germany) . The cut surface was coated with
Protein secretion. On day 4, the entire bioreactor cul­ gold in a sputter coater and examined under a scan­
ture system was washed with 250 mi supplemented Wil­ ning electron microscope (ISI SS40, Japan).
liams' E medium without FCS and incubated in the Transmission electron microscopy. A 4-day-old he­
same medium. The same procedure was performed for patocyte culture was fixed by flus hing one bioreactor
control bioreactors (which do not incorporate hepato­ with 4% paraformaldehyde. Cellular aggregates were
cytes) to investigate the possible contribution of pro­ mechanically stripped from the nonwoven polyester
tein release from the bioreactor as a result of the 3-day matrix. The specimens were postfixed in 1% OS04 (in
perfusion with supplemented Williams' E medium with cacody!ate buffer) for 15 min, block-stained with 1%
FCS. Media samples were collected after 24 hand dial­ uranyl acetate, dehydrated in 2,2-dimethoxypropane,
yzed extensively against a 50 mM NH 4HC0 3 solution and embedded in epoxy resin. Ultrathin sections of the
and then freeze-dried. The dry residues were reconsti­ hepatocyte aggregates were examined with an EM 10
tuted in an electrophoresis bufTer (Tris-barbital buffer, electron microscope (Philips, The Netherlands).
pH=8.6, ionic strength 0.1) to a concentration 20 times
greater than the culture supernatant. Statistical analysis

To visualize the individual serum proteins secreted An unpaired Student's t-test was used, and p<0.05 was

by the pig hepatocytes we performed crossed-over im­ considered to be statistically significant. Data are pre­

munoelectrophoresis as described previously (31). sented as mean:!:SEM.

Briefly, proteins in the concentrated culture super­


natant were separated electrophoretically in a 1% agar­ Results
ose gel (80 h, 10 V/cm) in the first dimension. In the Flow sensitive magnetic resonance imaging
second dimension the separated proteins were elec­ Fig. 2 displays the flow distribution in a cross-section
trophorized into an antiserum raised against pig serum of a sni.all (A) and a scaled-up bioreactor (B). The fiuid
proteins containing agarose gel (12 h, 80 Vlcm), result­ velocity was detected only in the axial direction and
ing in a number of precipitating peaks, each peak re­ ranged from zero (black) to around 2 crnls (white).
presenting an individual protein. When equa! amounts When compared with Fig. I several components of the
of the same antiserum are used in each plate, the rela­ bioreactor can be identified, such as the nonwoven
tive concentrations of the various proteins in the polyester fabric, the oxygenation hollow-fiber mem­
sample can be determined, as the area contained by branes, the flow channels, and the acrylic core. The
each peak is directly proportional to the amount of black representation of the nonwoven polyester fabric
antigen in the sample. The antiserum used was pre­ indicates only that medium flow within the 3D-matrix
pared in a rabbit (New Zealand White, 1.5 kg) by sev­ was not in the axial direction. The perfusion of the
eral injections of pig serum. fabric in other directions was not investigated. The
homogeneous distribution of the gray spots demon­
Microscopie examination strates that all flow channels in both devices were per­
Light microscopy. Hepatocytes from 5-day-old cultures fused . The shades of gray indicate that the fluid vel­
were fixed by flushing the bioreactors (n=3) with 4% ocity could differ per flow channel (ranging from 0.5
formalin . After 24 h the bioreactors were cut open and to 2 crnls, but mostly around 1.5 crnls). The arrows in
12 matrix samples (1 cm 2) were taken from various Fig. 2B show spots of decreased signal intensity as a
parts of the nonwoven polyester matrix. The samples result of entrapped air bubbles. Spin echo images (not

5
L. M. Flendrig el al.

trometer only allowed a horÎzontal orientation of the


bioreactor. Normally, the device is positioned verti­
cally, which facilitates the removal of air bubbles.

Hepatocyte culture
The study involved the construction of 22 bioreactors,
of which 16 devices (n=8 in duplicate) were used to
culture hepatocytes and six devices without cells (n=
3 in duplicate) served as controls. The results of the
hepatocyte function tests in two bioreactors with eells
from the same isolation procedure never differed more
than 10%, indicating reproducible cell immobilization
and cultivation. The culture system remained sterile
throughout the study and no leaking of medium into
the lumen of the oxygenation hollow-fibers was ob­
served (34).

Hepatocyte function tests


Galactose elimination. Fig. 3 shows that the galactose
elimination capacity after incubation for 180 min with
a standard dose of galactose remained constant over a
period 0[3 days.
Urea synthesis. Since high levels of ammonia are

0.5
-
-
-
E
C)
E 0.4
c
T
0
:;:;
as 0.3
c
:êQ)
CD 0.2
-
I/)
0
0
as
Fig. 2. Transaxialflow sensitive M Rls ofa smal/ (A, inter­ (ij 0.1
(!)
nal diameter 1.32 cm) and a scaled-up bioreactor (B, internal
diameter 2.2 cm). The fluid velocity ranged from zero
(black ) to around 2 cmls (white). When compared with Fig. o
I several components ofthe bioreactor can be identified, such 30 60 120 180 180 180
~~--~--~~------
as the nonwoven polyester fabric, the oxygenation hol/ow­ day 1 day 2 day 3
fiber membranes, the flow channels, and the acrylic core. The
images ofboth devices show that al/flow channels were per­ time (minutes)
fused. Differences in the fluid velocity ofthe flow channels can Fig. 3. Galactose elimination by porcine hepatocytes cul­
be observed. The arrows in Fig. 2B indicate spots of de­ tured for 3 h in the bioreactor inoculated with 220· /rf> vi­
creased signal intensity as a result ofentrapped air bubbles. able cel/s. I mg/ml galactose was added to the c/osed loop
circuit and incubatedfor 3 h every day for 3 days. Medium
samples were col/ected at different time points on day I
and after 3 h on days 2 and 3, after which the galactose
shown) revealed that the sÎze of the air bubbles was concentration was determined. Results are expressed a:
much smaller than the resulting distortion. The spec­ mean of seven experiments in duplicate±SEl'll.

6
Novel bioreactor

5
--a;
(J)
ual experiments, lidocaine clearance correlated better
with xylidine than MEGX formation. Porcine hepato­


cytes did not produce detectable levels of the metabo­
0
c 4
BAL lite GX during incubation with lidocaine for 1 h.
.2
Amino acid metabolism. Table I shows the changes
~ Monolayer in the medium concentration of some amino acids that
E
tn 3 are relevant for liver function (36,37). A decrease in

-::i.
( J)
.e;;

-Cl)
.r:.
c

(J)
2

1
500

ca 400
Cl)
~

-E
::::>
o
15 30
~~--~~--~~~~~
60 120 120 120
-~300
Cl)
dav 1 daV 2 daV 3 c
.ä; 200
time (minutes) 0
0
Fig. 4. Urea synthesis by porcine hepatocytes .cultured for "'0
2 h in the bioreactor inoculated with 220· lef viabie cel/s :.J
100
and as monolayers at 3· lef viabie ce/ls per weil. JO mM
NH4CL was added to both culture systems and incubated
for 2 h every day for 3 days. Medium samples of the biore­ 0
actor were collected at different time points on day land 40
after 2 h on days 2 and 3, and in monolayer cultures after
f:LI MEGX
T
2 h on days I to 3, after which the urea concentration was
determined. Results are expressed as mean of eight experi­
ments in duplicate±SEM:
-E 30
~
Cl>
.6
considered to play a role in hepatic encephalopathy :2
>­ 20
(35), we tested the bioreactor's ability to synthesize X

"t:J

urea from ammonia. The efficacy of the device to sup­ C


port hepatocytes was evaluated by comparing the urea­ as

X
10
synthesizing capacity of the hepatocytes in the bioreac­ (!)
tor to that of hepatocytes in monolayer cultures. The w
~
results in Fig. 4 show that the urea synthesis in both
culture systems, after a 120 min incubation with 10
mM NH 4 Cl, did not vary over a period of 3 days. The
urea-synthesizing capacity of the hepatocytes cultured
o 10 20 30 60 60 60
in the bioreactor was twice as high as hepatocytes cu 1­ dav 1 daV 2 day 3
tured as monolayers. time (minutes)
Lidocaine metabolism. The cytochrome P450 activity
of the hepatocytes was assessed by determining lido­ Fig. 5. Lidocaine elimination and subsequent MEGX and
xylidine format ion by porcine hepatocytes cultured for I h
caine and its metabolites (Fig. 5). The lidocaine elimin­
in the bioreactor inoculated with 220· lef viabie ceiIs. 500
ation and subsequent MEGX and xylidine production
Jlglml lidocaine was added to the c10sed loop circuit and
after a 60 min lidocaine incubation did not significant­ incubated for I h every day for 3 days. Medium samples
Iy change over a period of 3 days. Xylidine was the were collected at different time points on day I and after I
main lidocaine metabolite during the first 2 days. h on days 2 and 3, after which Ihe lidocaine, MEGX, and
There was no significant difference in xylidine and xylidine concentralion were delermined. Resulls are ex­
MEGX production on day 3. When looking at individ­ pressed as mean of seven experiments in duplicale±SEM.

7
L. M. Flendrig el al.

glutamine concentration was associated with an in­ day of culture (Tabie 1). A significant decrease in the
creased glutamate concentration. Liver metabolism of glucose concentration was observed on days 2 and 3.
aromatic amino acids was refiected by a decrease in the pH. The pH in the studied bioreactor was kept con­
concentrations of phenylalanine, tyrosine, and trypto­ stant (Tabie 1) by an integrated oxygenator which en­
phan. Decreased arginine concentrations and synthesis sured stabie CO 2 partial pressures (32.6::t0.4 mmHg,
of ornithine are indicative of arginase activity. A de­ n=8) in the sodium bicarbonate bufIered medium.
crease in alanine concentration, a precursor of liver Protein secretion. Cultured hepatocytes secrete pro­
gluconeogenesis, was observed. teins into their culture medium. On day four a two­
Other amino acids concentrations which decreased dimensional crossed immunoelectrophoresis was per­
statistically significantly were asparagine, glycine, histi­ formed after a 24 hour incubation with supplemented
dine, valine, methionine, isoleucine, leucine, and lysine Wil1iams'E medium without FCS. The result of one
(data not shown in Table 1). The observed changes in representative experiment (out of three) is shown in
amino acid concentrations were similar on days 1, 2, Fig. 6. Each precipitation peak represents an individual
and 3. protein. No proteins were detected in identical experi­
Lactate/pyruvate ratio. The lactate/pyruvate ratio is ments with con trol bioreactors (without cells).
an index of the functional state of cellular oxidation
and aerobic metabolism (38). Table 1 shows a drop in Microscopie examination
the lactate/pyruvate ratio, which was solely due to a Light microscopy studies. Fig. 7 shows a microscopic
decline in the lactate concentration. The lactate/pyruv­ photograph of a cross-section of the 3D-matrÎx from
ate ratio of 5 to 7 refiected physiological oxygenation a bioreactor at 5 days in culture. The hepatocytes from
of the culture system over a period of 3 days. the single-cell suspension reorganized into small irregu­
En:yme leakage. To assess hepatocyte viability, the lar shaped aggregates that were immobilized on and
appearance of enzyme activity, namely LDH, GOT, entrapped within the polyester fiber framework. De­
and GPT, was determined in the culture medium. LDH spite high density culturing there is sufficient space be­
release was only significant on day 1 (Tab Ie I). GOT tween the aggregates for medium perfusion. The aggre­
liberation was significant and tended to fal1 over the 3­ gates are so small (one diameter never being larger
day period. Low but significant quantities of GPT were than five cells, mostly two to three cells) that most
released on days 1 and 2. hepatocytes function in direct contact with the me­
Glucose. Glucose levels did not change during first dium. As the 3D-matrix is relatively empty, there is the

TABlE I
Results of a 14-h incubation (every day for 3 days) of 220· 1()6 bioreactor cultured hepatocytes in supplemented Williams' E medium concerni
changes in amino acid concentrations. lactate and pyruvate concentrations and lactateJpyruvate ratios. enzyme leakage. glucose concentratio nl>.
and pH
Evaluation Unit Comrol· day I day 2 day 3
Glutamate·· tlM 402.9::6.6 851.3::80.4 971.6::62.6 1038.2::94.6
Glutamine tl M 1893.0::47.1 881.4:: 106.6 809 .3:: 119.0 784.0:: 124.0
Phenylalanine tlM 155.1 ::2.2 62.1 ::6.2 59.1=6.7 68.2:: 5.03
Tyrosine tl M 181.7::2.3 58.0:: 12.6 51.8:: 18.2 50.6:: 14.0
Tryptophan tlM 50.6::0.7 20.6::4.1 13.5::3.7 11.8:: 1.5
Arginine tlM 306.9::9.3 15.0::2.0 15.0::4.4 18.4::6.1
Ornithine tlM 28.2::3.8 231.5::24.1 229.8::27.7 250.4::28.3
Alanine tlM 1088.4::20.9 408.8::89.0 457.7::82.0 424.6::64.4
Lactate··· mM 1.44::0.02 0.34::0.07 0.26::0.06 0.27::0.05
Pyruvate mM 0.08::0.004 0.05::0.01 0.05::0.01 0.05::0.004
lactJPyr ratio 18.0::0.8 6.7::0.8 5.4::0.5 5.6::0.8
LDH··· UIL 14.3::0.9 35.2::3.5 21.0::2 .7" 14.8:: 1.3"
GaT UIL 4.2::0.2 169::40 120::41.9 93::34
GPT UIL 0.95::0.2 2. 1::0.3 1.7::0.2 1.4=0.2­
Glucose··· mM 12.0::0.1 12.4::0.7" 10.9::0.4 9.6::0.6
pH··· 7.46::0.03 7.36::0.02" 7.39::0.01­ 7.40::0.01­
• Mean of three experiments in duplicate::SEM .

•• Mean of six experiments in duplicate::SEM .

... Mean of eight experiments in duplicate::SEM.


p<0.05 versus control. except for data indicated with a.

8
Navel bioreactor

micrograph of isolated hepatocytes after 5 days in cul­


ture in the 3D-matrix of the bioreactor. A hepatocyte
aggregate immobilized between the polyester fibers of
the 3D-matrÎx is displayed. The hepatocytes are com­
pletely embedded in a matrix material, the compo­
sition of which is currently under investigation.

Fig. 6. Prolein secrelion by porcine hepatocyles cullured Fig. 7. Lighl microscopic pholomicrograph of a cross-sec­
for 72-96 h in Ihe bioreactor inoculaled wilh 220· /(15 viabie lion of lhe 3D-malrix from a bioreactor in which 20· /(15
cells. On day 4 Ihe enlire bioreactor culture syslem was viabie hepalocyles/ml had been cullured for 5 days. One
washed wilh 250 mi supplemenled Wil/iams' E medium represenlalive specimen (oulof Ihree bioreactor experi­
withoul FeS and incubaled in Ihe same medium. The me­ menIs) is shown. The hepatocyles sponlaneously form small
dium sample was collecled afler 24 h and subjecled 10 aggregales which inunobilize on and belween lhe poiyesIer
crossed immunoeleclrophoresis analysis using a polyspecific fibers (Iranslucent circles, /3 f.U7l in diameIer) of Ihe non­
anliserum 10 pig serum proleins. The resull ofone represen­ woven fabric. There is plenly of space belween Ihe aggre­
/alive experimenl (oulof Ihree) is shown. Each peak rep re­ gates for unhindered perfusion of the hepalocytes.
senls an individual prolein. No proleins were delecled in
identical experimenls using cell-free bioreactors (nol
shown) .

potential for culturing hepatocytes at even higher den­


si ties than the present 20· 106 viabie cells per mI.
Examination of 3D-matrÎx samples taken near the
inlet and outlet port and in the middle of the non­
woven fabric revealed that the hepatocytes are evenly
distributed in the bioreactor device (results not shown).
Cel! counts in 12 microscopic preparations (dimen­
sions: length 10 mm, width 0.5 mm, thickness 8 f.1ID),
chosen from various parts of the 3D-matrÎx (dimen­
sions: length 140 mm, width 90 mm, thickness 0.5 mm)
of one bioreactor, revealed an average number of Fig. 8. Scanning electron micrograph of isolaled porcine
hepatocytes cultured for five days in the 3D-malrix of a
1379:: 135 (mean::SD) hepatocytes/preparation. If
bioreactor device at 20 · /(15 viabie ce lis per mi. A small
220· 106 viabie hepatocytes would immobilize within
hepatocyte aggregate entrapped between the poiyesIer
the nonwoven fabric, one can calculate that every prep­ fibers of the nonwoven polyester matrix is shown. The
aration should contain about 1400 viabie cells. So on hepatocytes are completely embedded in a matrix material
average, 98% of the seeded viabie hepatocytes were im­ which composition is currently wuJer investigation. Ten­
mobilized in this experiment. tacles, possibly ofprotein origin, cleave the interfiber space
SEM studies. Fig. 8 shows a scanning electron (arrows). The polyester fibres are /3 f.U7l in diameIer.

9
L. M. Flendrig el al.

Firstly, clinical treatment of hepatic failure requires


large scale, high density hepatocyte culture. In many bi­
oreactors such high coneentrations give rise to the for­
mation of non-physiological sized clusters of ceUs.
Hepatocytes in the center of these large aggregates have
poor metabolic activity and may even show necrosis, as
most of the oxygen and nutrients win be consumed after
passing the surrounding een-Iayers, also hindering the
removal of carbon dioxide, toxins, and cell products
from these cens (9,41,42). This redueed ra te of transport
inside large cen aggregates (internal mass transfer) is not
observed in the in vivo liver where the cell orientation is
such that every hepatocyte operates in close contact
with the blood . Secondly, in most of these capillary
Fig. 9. Transmission electron micrograph of a hepatocyte
membrane bioreactors, substrate and metabolite ex­
aggregate from the 3D-matrix of a bioreactor at 4 days in
change between the culture medium and the eeU surfa
culture (bar represents 1 fJl1l). Neighboring cells reconsti­
tute bile canaliculus-like structures (BC) with typical (external mass transfer) depends on diffusion, which I.
microvilli andjunctional complexes including tightjwrctions known to be a strongly limiting transport mechanism
(arrows) and desmosomes (D). Other cell structures dis­ (42). This is in contrast to the in vivo situation where
played are: mitochondria (M), Goigi complexes (G), hepatocytes function under perfusion conditions \liith
rough endoplasmatic reticulum (RER), a peroxisome (P), correspondingly low diffusional gradients.
and a nucleus ( N). We developed a novel bioreactor deviee (Fig. 1) for
potential use as a BAL, which addresses the above­
mentioned requirements for physiological mass trans­
TEM studies. Transmission electron microscopy on fer. This new system has the following features:
hepatocyte aggregates at 4 days in culture showed the Three-dimensional nonwoven polyester fabric. Lght
distinctive ultrastructure of viabie hepatocytes (Fig. 9). microscopic and scanning electron microscopic exam­
The hepatocytes exhibited extensive eell-cell contact. ination has shown that the polyester fibers of the non­
Adjacent cells reconstituted bile canaliculus-like struc­ woven fabric provide a framework for high density he­
tures with typical microvilli and junctional complexes patocyte immobilization (20· 106 cen per mi) and
including tight junctions and desmosomes. Mitochon­ spontaneous reorganization into small aggregates (one
dria, nuclei, Golgi complexes, rough endoplasmatic diameter never being larger than five eells, mostly :wo
reticulum, and peroxisomes appeared norma!. to three eells) with space between the aggregates. This
allows individual perfusion of hepatocytes with direc-""
Discussion supply of oxygen and substrates to and removal of cat­
The development of bioreactor devices specifically de­ abolites and eell produets away from the eells, resulting
signed for use as an extracorporeal bioartificial liver is in low diffusional gradients that more closely mimic
a field that deserves more attention. Standard hollow­ physiological conditions.
fiber membrane deviees, as known in dialysis practice, No extracellular matrix materiais. In a pre\ious
have been used for hepatocyte cultivation since the be­ study we eompared the functional activity of porcine
ginning of the seventies (9). Uchino et al. (6) and Ger­ hepatocytes attaehed to hydrophillie tissue culture
lach et al. (10) were the first to devise custom-made plastic, to ceUs attached to several extracellular matrix
bioreactors. Most researchers continued culturing constituents: eoUagen land rv, laminin, fibronectin,
hepatocytes in the intraluminal (15) and extrafiber Engelbreth-Holm-Swarm Natrix, and in the presence
space (12,13,17,39,40) of common hollow-fiber mem­ of Matrigel (31). With the exception of Matrigel, nei­
brane units. The popularity of this method of cell cu1­ ther of the extracellular matrix substrates enhanced
turing can be easily understood, as it is the simplest porcine hepatocyte function compared to tissue culture
way of achieving a BAL. However, it remains to be plastic. Matrigel has the disadvantages that it is \-ery
seen whether these systems have a future, as they do expensive and that relatively large amounts of murine
not meet the essential conditions for optimal substrate proteins of tumor origin leak out of the gel and might
and metabolite exchange to hepatocytes as present in get into the eireulation of the patient. We therefore de­
the intact liver. As a consequence, hepatocyte meta­ eided to inject the cell suspension directly into the d.r .--,.
bolic activity is impaired for the foUowing reasons: bioreactor. No prerinsing of the bioreactor with me­

10
Novel bioreactor

dium or coating of the polyester fibers with common the oxygenator in the bioreactor. The spirally wound
extracellular matrix materials such as Matrigel (10) construction of our device creates a homogeneous dis­
and collagen (6,7) was necessary. The result appeared tribution of the oxygenation hollow-fibers throughout
to be a safer, cheaper and more convenient device. the bioreactor, thereby ensuring every hepatocyte of an
Low substrate and metabolite gradients. On acellular oxygenation source within its direct surroundings. This
level, low substrate and metabolite gradients in a high results in optimal oxygenation of the hepatocytes,
density hepatocyte culture can be realized by a cell which was confirmed by a sta bie, physiologicallactate/
orientation that allows every hepatocyte to function in pyruvate ratio (38), and stabie pH, indicating constant
direct contact with the medium, e.g. the formation of CO 2 partial pressures in the sodium bicarbonate buf­
smal! hepatocyte aggregates in the nonwoven polyester fered medium.
matrix. When looking at the entire bioreactor, low sub­ Bioeompatibility. The bioreactor is constructed of
strate and metabolite gradients can be obtained either materials that have been FDA approved and withstand
by reducing the perfusion distance between the inlet the high thermal stress of autoclaving. As far as we
port and outlet port or by increasing the medium flow know this is the first bioreactor for hepatocyte culture
rate. Gerlach et al. (lO),came up with an interesting but that can be steam sterilized. This procedure is biologic­
complicated bioreactor design that realized the former ally much safer than the commonly used ethylene-ox­
option by culturing the hepatocytes between four inde­ ide sterilization, which is very toxic. Ethylene-oxide
pendently woven hollow-fiber membrane bundies, residues leak out of polymers for many weeks and may
among one for medium inflow and another for me­ cause sensitiza tion and allergic reactions in patients
dium outflow. This allows decentralized perfusion of (43,44).
the cells between these capillaries with low diffusional 6. Easy sealing-up. As can be concluded from Fig.
gradients. A technically much simp Ier solution is the 2, sealing-up simply imp lies increasing the number of
latter option of increasing medium flow rate through windings of the hollow-fiber/3D-matrix composite un­
the bioreactor. In contrast to other bioreactors, our til the required immobilization capacity has been ob­
novel device has been constructed to benefit from this tained. The length of the bioreactor remains the same
principle as much as possible. Firstly, the module does while its diameter increases. A bigger device will there­
not comprise semi-permeable hollow-fiber membranes fore have more flow channels and oxygenation hollow­
for plasma or blood perfusion, which can foul and act fibers. The bioreactor configuration is not affected by
as a diffusional barrier to the hepatocytes. In our sys­ this, since the dimensions of the flow channels, the hol­
tem, plasma can come in direct contact with the low-fibers, and the thickness of the 3D-matrix remain
hepatocytes. Secondly, macroscopic and microscopic the same. Hence, scaling-up will not influence the
evaluation of the bioreactor revealed that the majority plasma distribution in the bioreactor. This was con­
of the hepatocytes were immobilized within the 3D­ firmed by flow sensitive MRI, which showed perfusion
matrix and thereby protected from shear stress. This of all flow channels in a small and a scaled-up bioreac­
was confirmed by a pilot experiment in which the me­ tor. The fluid velocity could differ per flow channel,
dium flow rate was increased stepwise from 5 mVrnin which is a result of the fact that the bioreactors were
(standard flow) to 15 mlJmin. No signs of shear stress, hand-made. Industrial production techniques are cur­
such as a decrease in hepatocyte functional activity or rently evaluated to solve this. The use of standard di­
an increase in enzyme leakage, were observed. The in­ alysis housings and potting techniques enable easy
creased flow rates only slightly increased the inlet manufacturing of a wide range of bioreactor sizes. Bi­
pressure, as the resistance of the bioreactor to fluid oreactors, with a volume of 400 mi that can hold up
flow is very low. The non-invasive technique of flow to 20· 10 9 hepatocytes, are currently being constructed
sensitive MRI revealed that medium transport was en­ (Microgon, Laguna Hills, CA) for experiments in large
su red to all parts of the 3D-matrix by numerous flow animal models.
channels, which are evenly distributed throughout the A bioartiiicialliver support system for the treatment
bioreactor space (D in Fig. I). These channels also pro­ of fulminant hepatic failure and as a bridge to liver
vide a homogeneous supply of the injected hepatocyte transplantation requires large amounts of viabie and
suspension to the 3D-matrix. actively functioning hepatocytes. Porcine hepatocytes
Deeentralized oxygenation. The oxygen carrying ca­ are considered to be the best alternative to human
pacity of plasma is very limited. It is therefore not un­ hepatocytes. Human hepatocytes for culture are scarce
likely that hypoxic regions might occur in bioreactors and transformed human hepatocytes may lack critica I
with an external oxygenator. In line with Gerlach et al. hepatocyte functions (45,46). Pig livers can be readily
(30) we believe that this can be overcome by integrating obtained from laboratory animals or from slaughter

11
L. M. Flendrig el al.

houses, and porcine hepatocytes can be easily isolated tering the rat plasma at the inlet and the ou tiet of the
in large quantities with a simple two-step collagenase bioreactor. Possible humoral irnmunological compli­
perfusion technique. cations may be circumvented by choosing filters with a
Long-term in vitro studies are important to evaluate proper membrane cut-off (48,49). An in vivo study on
the ability of the bioreactor to maintain the functional the effects of toxic plasma of LIS-rats on the viability
integrity of the hepatocytes (10,47). In a pilot experi­ of the porcine hepatocytes in the BAL is in progress.
ment, two of our bioreactors were loaded with hepato­ In conclusion, we have devised a novel bioreactor
cytes of one cell isolation and cultured over a period configuration which shows promising efficiency and
of two weeks. Lidocaine clearance was monitored as ensures maintenance of various liver specific functions
an index of cytochrome P450 activity, which has been over the investigated period at a high density of cul­
suggested to be the critical function that must be pro­ tured hepatocytes. This system, which is easy to manu­
vided bya successful BAL (5). In both devices the P450 facture, use and scale up, appears to have considerable
activity was sustained over the investigated 14 days potential for short term support of patients in hepatic
(300±18 ,ug'ml-1'h- 1) with a gradually decreasing failure.
trend in the second week to around 70% of the initia I
activity (213±27 ,ug' mi-I. h- 1). Although these re­ Acknowledgements
su lts demonstrate that the functional integrity of the We thank our colleagues from the Department of Ex
hepatocytes was maintained, at least during the first perimental Cardiology and the Department of Derma­
week, this is no guarantee that the initial functional tology for providing the pig livers. We further wish to
activity of the hepatocytes in the bioreactor would ex­ thank Mrs. 1. Maathuis for performing the light micro­
ceed that of hepatocytes in other types of culture sys­ scopic preparations, Or. 1. van Marle and Mr. H. van
tems. So, the efficacy of a bioreactor construction Veen for performing the scanning EM, Dr. K.P. Dinge­
should not only be judged by its potential to maintain mans and Mr. M.A. van den Bergh Weerman for per­
hepatocyte function and viability, but also include a forming the transmission EM, and Dr. E.A . Jones and
study on its efficiency. For the latter, hepatocyte func­ Dr. W. Boers for critical reading of the manuscript.
tion in the bioreactor should be compared with stan­ This research has been made possible by a grant
dardized hepatocyte culture techniques. Such a com­ from the Netherlands Digestive Diseases Foundation.
parison does not necessarily require long-term studies.
We chose for a 3-day period, since the future appli­
cation of our bioreactor as a BAL will be shortly after References
I. Takahashi T, Malchesky PS, Nosé Y. Artificial liver. State of
hepatocyte immobilization. Ultimately, the different the art. Dig Dis Sci 199\ ; 36: \327-40.
bioreactor constructions currently under investigation 2. Malchesky PS. Nonbiological Iiver support: historie over­
should be tested in one center, under identical con­ view. Artif Organs 1994; 18: 342-7.
ditions, to elucidate the design characteristics that are 3. Davies E, Hodgson HJF. Artificial livers - what's keepir"-"'"
them? Gut 1995; 36: 168-70.
essential to efficiency.
4. O'Grady JG, Grinson AES, O'Brien CJ, Puclmell A, Hughes
The urea-synthesizing capacity of the hepatocytes in RD, Williams R. Controlled trials of charcoa! hemoperfusion
our bioreactor was twice that of hepatocytes in mono­ and prognostic factors in fulminant hepatic failure. Gastro­
layer cultures. This encouraging efficiency indicates enterology 1988; 94: 1186-92.
that the features of our device resulted in an environ­ 5. Jauregui HO, Gann KL. Mammalian hepatocytes as founda­
ment for hepatocyte cultivation that more closely re­ tion for treatment in human liver failure. J Cell Biochem
1991; 45: 359-ó5.
sembles the in vivo situation. Other liver-specific func­ 6. Uchino J, Tsuburaya T, Kumagai F, Hase T, Hamada T, Ko­
tions like galactose elimination and amino acid metab­ mai T, Funatsu A et al. Hybrid bioartificial liver composed
olism were weil maintained over the experimental of multiplated hepatocyte monolayers. ASAIO Trans 1988;
period. 34: 972-7.
In order to evaluate the full potentialof the bioreac­ 7. Arnaout WS, Moscioni AD, Barbour RL, Demetriou AA.
Development of a bioartificial Iiver: Bilirubine conjugation
tor as a bioartificialliver, in vivo experiments in experi­ in gunn rats. J Surg Res 1990; 48: 379-82.
mental animal models and clinical trials need to be 8. Shnyra A, Bocharov A, Bochlc:ova N, Spirov V. Bioartificial
performed. In vivo experiments in rats with liver is­ Iiver using hepatocytes on biosilon microcarriers: treatment
chemia (LIS), treated with porcine hepatocyte based of chemically induced acute hepatic failure in rats. Artif Or­
bioreactors (40· 106 eeUs per ml), are nearly completed. gans 1991; 15: 189-97.
9. Wolf CF, Munkelt BE. Bilirubine conjugation by an artificial
So far, the results show statistically significantly im­ liver composed of cultured eells and synthetic capillaries
proved survival compared to control experiments. Trans Am Soc Artif Intern Organs 1975; 21: 16-27.
Cellular immunological problems are banned by fil­ 10. Gerlach JC, Enclc:e J, Hole 0, Maller C, Ryan CJ, Neuhaus

12
Navel bioreactor

P. Bioreactor for larger scale hepatocyte in vi/ra perfusion. Formation of porcine hepatocyte spheroids for use in a bi­
Transplantation 1994; 58: 984-8. oartificialliver. Cell Transplant 1995; 4: 259-68.
11. Sussman NL, Chong MG, Koussayer T, He D, Shang T, Whi­ 28. Rotem A, Toner M, Bhatia S, Foy BD, Tompkins RG, Yar­
sennand H, Kelly JH. Reversal of fulminant hepatic failure mush ML. Oxygen is a factor determining in vi/ra tissue as­
using an extracorporeal liver assist device. Hepatology 1992; sembly: Effects on attachment and spreading of hepatocytes.
16: 60-5. Biotech Bioeng 1994; 43: 654-60.
12. Dixit V. Development of a bioartificial liver using isolated 29. Catapano G, De Bartoio L, Lombardi Cp, Drioli E. The effect
hepatocytes. Artif Organs 1994; 18: 371-84. of oxygen transport resistances on the viability and functions
13. Jauregui HO, Mullon CJp, Trenkler D, Naik S, Santangini H, ofisolated rat hepatocytes. Int J ArtifOrgans 1996; 19: 61-71.
Press p, Muller TE et al. In vivo evaluation of a bollow fiber 30. Gerlach J, Klöppel K, Stol! p, Vienken J, Müller C. Gas sup­
liver assist device. Hepatology 1995; 21: 460-9. ply across membranes in bioreactors for hepatocyte culture.
14. Yanagi K, Ookawa K, Mizuno S, Oshima N. Performance of Artif Organs 1990; 14: 328-33.
a new hybrid artificial liver support system using hepatocytes 31. te Velde AA, Ladiges NClJ, Flendrig LM, Chamuleau
entrapped within a hydrogel. Trans Am Soc Artif Intern Org RAFM. Functional activity of isolated pig hepatocytes at­
1989; 35: 570-2. tached to different extracel!ular matrix substrates. Impli­
15. Nyberg SL, Shirabe K, Peshwa MY, Sielaff TD, Crotty PL, cations for application of pig hepatocytes in a bioartificial
Mann HJ, Remmei RP et al. Extracorporeal application of liver. J Hepatol 1995; 23: 184-92.
a gel-entrapment, bioartificial liver: Demonstration of drug 32. Karlsen OT, Creyghton JHN, Mehlkopf AF, Bovée WMMJ.
metabolism and other biochemical functions. Cel! Transplant A steady state perfusion experiment. Proc ISMRM, 4 th
1993; 2: 441-52. Scientific Meeting 1996: 1308.
16. Koebe HG, Wick M, Cramer U, Lange Y, Schildberg FW. 33. van Eijk HMH, van der Heijden MAH, van Serlo CLH, Soe­
Collagen gel immobilisation provides a suitable cel! matrix ters PB. Fully. automated liquid-chromatographic determi­
for long term human hepatocyte cultures in hybrid reactors. nation of amino acids. Clin Chem 1988; 34: 2510-3.
Int J ArtifOrgans 1994; 17: 95-106. 34. Montoya Jp, Shanley Cl, Merz SI, Bartlett RH. Plasma leak­
17. Takeshita K, Ishibashi H, Suzuki M, Yamamoto T, Akaike T, age through microporous membranes. Role of phospho­
Kodama M. High cell-density culture system of hepatocytes lipids. ASAIO J 1992; 38: M399-405.
entrapped in a three-dimensional hollow fiber module with 35. Butterworth RF, Giuère JI:; Michaud J, Lavoie J, Pomeir­
collagen gel. Artif Organs 1995; 19: 191-3. Layrargues G. Ammonia: key factor in the pathogenesis of
18. Fremond B, Malandain C, Guyomard C, Chesne C, Guillou­ hepatic encephalopathy. Neurochem Pathol 1987; 6: 1-12.
zo A, Campion J. Correction of bilirubin conjugation in the 36. Meijer AJ, Lamers WH, Chamuleau RAFM. Nitrogen me­
gunn rat using hepatocytes immobilized in alginate gel beads tabolism and ornithine cycle function. Physiol Rev 1990; 70:
as an extracorporeal bioartificialliver. Cell Transplant 1993; 701-48.
2: 453-60. 37. Fuchs M, Gerlach J, Encke J, Unger J, Smith M, Neuhaus p,
19. Matthew HWT, Basu S, Peterson WD, Salley SO, Klein MD. Riedel E. Amino acid metabolism by hepatocytes in a hybrid
Performance of plasma perfused, microencapsulated hepato­ bioreactor. Int J Artif Org 1994; 17: 663-9.
cytes: Prospects for extracorporeal liver support. J Pediatr 38. Gudbjarnason S, Bing RJ. The redox-potential or the lactate­
Surg 1993; 28: 1423-8. pyruvate system in blood as an indicator of the functional
20. Miyoshi H, Yanagi K, Fukuda H, Ohshima N. Long-term state of cell ular oxidation. Biochim Biophys Acta 1962; 60:
continuous culture of hepatocytes in a packed-bed reactor I 58-ó2.
utilizing porous resin. Biotech Bioeng 1994; 43: 635-44. 39. Demetriou AA, Rozga J, Podestra L, Lepage E, Morsiani E,
21. Nyberg SL, Madhusudan Vp' William DP, Hu W-C, Cerra Moscioni AD, Hoffman A et al. Early clinical experience
FB. Evolution of the bioartificial liver: The need for ran­ with a hybrid bioartificial liver. Scand J Gastroenterol 1995;
domized clinical trials. Am J Surg 1993; 166: 512-21. 30: 111-7.
22. Sussman NL, Gislason GT, Kelly JH. Extracorporeal liver 40. Sussman NL, Gislason GT, Conlin CA, Kelly JH. The hepa­
support. Application to fulminant hepatic failure. J Clin tix extracorporeal liver assist device: Initial clinical experi­
Gastroenterol 1994; 18: 320-4. ence. Artif Organs 1994; 18: 390-{).
23. Rozga J, Morsiani E, Lepage E, Moscioni AD, Giorgio T, 41. Ho CS, Wang IC. Animal Cel! Bioreactors. Boston: Butter­
Demetriou AA. Isolated hepatocytes in a bioartificial liver: a worth- Heinemann, 1991: 13, 14, 196, 197,358.
single group view and experience. Biotech Bioeng 1994; 43: 42. Willaert RG, Baron GY, De Backer L. Mass transfer in im­
645-53. mobilised cell systems. In: Willaert RG, Baron GY, De
24. Chamuleau RAFM, Flendrig LM, la Soe JW, Ladiges NClJ, Backer L. Immobilised Living Cell Systems. Chichester: John
te Velde AA. Bioartificial liver and its clinical significance Wiley & Sons, 1996; 21-45.
anno 1994. Transplantology 1994; 5: 166-70. 43. Röckel A. Klinke B, Hertel J, Baur X, Thiel C, Abdelhamid
25. Landry J, Sernier D, Oue1let C, Goyette R, Marceau N. S, Fiegel P et al. A1lergy to dialysis materiais. Nephrol Dial
Spheroidal aggregate culture of rat liver cells: Histotypic re­ Transplant 1989; 4: 646-52.
organization, biomatrix deposition and maintenance of func­ 44. Rumpf KW, Seubert S, Seubert A, Jaeger M, Lorvitz HO,
tional activities. J Cell Biol 1985; lOl: 914-23. Valentin R, Schunemann B et al. Ethylene-oxide induced IgE
26. Koide N, Shinji T, Tanabe T, Asano K, Kawaguchi M, Saka­ antibodies and symptomatology in dialysis patients. Contr
guchi K, Koide Y et al. Continued high albumin production Nephrol 1987; 59: 145-53.
by multicellular spheroids of adult rat hepatocytes formed in 45. Rozga J, Williams F, Ro M-S, Neuzil DF, Giorgio TD,
the presence of liver-derived proteoglycans. Biochern Biophys Backfish G, Mosconi AD et al. Development of a bioartificiaJ
Res Commun 1989; 161; 385-91. liver: Properties and function of a hollow-fiber module inocu­
27. Lazar A, Peshwa MY, Wu FJ, Chi C, Cerra FB, Hu W-c. lated with liver cells. Hepatology 1993; 17: 258-65.

13
L. M. Flendrig el al.

46. Nyberg SL, Remmei Rp, Mann HJ, I\:shwa MY, Hu W-S, 48. Nyberg SL, Platt JL, Shirabe K, Payne WO, Hu W-S. Cerra
Cerra FB. Primary hepatocytes outperform HepG2 cells as FB. Immunoprotection of xenocytes in a hollow fiber bioarti­
the source of biotransformation functions in a bioartificial ficialliver. ASAIO J 1992; 38: M463-7.
liver. Ann Surg 1994; 220: 59-67. 49. te Yelde AA, Flendrig LM, Ladiges NCJJ, Chamuleau
47. Gerlach JC, Schnoy N, Encke J, Smith MD, MUller C, Neu­ RAFM. Immunological consequences of the use of xeno­
haus P. Improved hepatocyte in vilra maintenance in a culture geneic hepatocytes in a bioartificial liver for acute hepatic
model with woven multicompartment capillary systems: elec­ failure. Int J Artif Organs 1996; in press.
tron microscopy studies. Hepatology 1995; 22: 546-52.

14

Das könnte Ihnen auch gefallen