in vitro microscale systems for systematic drug toxicity study

15
MINI REVIEW In vitro microscale systems for systematic drug toxicity study Jong Hwan Sung Michael L. Shuler Received: 28 May 2009 / Accepted: 4 August 2009 / Published online: 23 August 2009 Ó Springer-Verlag 2009 Abstract After administration, drugs go through a complex, dynamic process of absorption, distribution, metabolism and excretion. The resulting time-dependent concentration, termed pharmacokinetics (PK), is critical to the pharmacological response from patients. An in vitro system that can test the dynamics of drug effects in a more systematic way would save time and costs in drug devel- opment. Integration of microfabrication and cell culture techniques has resulted in ‘cells-on-a-chip’ technology, which is showing promise for high-throughput drug screening in physiologically relevant manner. In this review, we summarize current research efforts which ulti- mately lead to in vitro systems for testing drug’s effect in PK-based manner. In particular, we highlight the contri- bution of microscale systems towards this goal. We envi- sion that the ‘cells-on-a-chip’ technology will serve as a valuable link between in vitro and in vivo studies, reducing the demand for animal studies, and making clinical trials more effective. Keywords Microfluidics Pharmacokinetics ADME Introduction Drug development is an expensive, time consuming pro- cess. Despite continuing effort to improve the productivity of the drug development process, only one out of ten drug candidates entering the clinical trial reaches the final approval stage [1, 2]. The number of new molecular enti- ties (NMEs) that are approved by US FDA has been declining over the last decade, from 53 in 1996 to 21 in 2008 [3]. The main reasons for such a low success rate are primarily unforeseen lack of efficacy (accounting for about 30% of failures) and toxicity (accounting for another 30%) [1]. When the lack of efficacy or unexpectedly high toxi- city is revealed in later stages of clinical trials, it increases the total cost, since the cost of clinical testing is consid- erably higher than the cost of preclinical testing, increasing with successive phases [4]. Improving the ability of in vitro assay systems to predict the efficacy and toxicity of drug candidates earlier in the drug discovery process will greatly enhance productivity. Consequently, there has been a sig- nificant amount of effort geared towards improvement of in vitro systems for assessing drug efficacy and toxicity [5, 6]. Typically, cell-based assessment of drug toxicity and efficacy is evaluated in multi-well microtiter plate systems. Cells are cultured in multi-well plates, incubated with drugs for a specific amount of time, and the viability of the cells are measured. A large number of human cell lines intended for specific target organ toxicity are available [7]. Although this methodology has been well-established and has been used for several decades as a standard method for evaluation of drugs, many drugs move onto successive stages only to fail, due to unforeseen toxicity or lack of efficacy. The predictability of multi-well, cell-based drug assay is not satisfactory because this system differs from the human body in many aspects. First, in a cell-based J. H. Sung M. L. Shuler (&) Department of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA e-mail: [email protected] J. H. Sung e-mail: [email protected] M. L. Shuler Department of Biomedical Engineering, Cornell University, Ithaca, USA 123 Bioprocess Biosyst Eng (2010) 33:5–19 DOI 10.1007/s00449-009-0369-y

Upload: jong-hwan-sung

Post on 15-Jul-2016

214 views

Category:

Documents


1 download

TRANSCRIPT

MINI REVIEW

In vitro microscale systems for systematic drug toxicity study

Jong Hwan Sung Æ Michael L. Shuler

Received: 28 May 2009 / Accepted: 4 August 2009 / Published online: 23 August 2009

� Springer-Verlag 2009

Abstract After administration, drugs go through a

complex, dynamic process of absorption, distribution,

metabolism and excretion. The resulting time-dependent

concentration, termed pharmacokinetics (PK), is critical to

the pharmacological response from patients. An in vitro

system that can test the dynamics of drug effects in a more

systematic way would save time and costs in drug devel-

opment. Integration of microfabrication and cell culture

techniques has resulted in ‘cells-on-a-chip’ technology,

which is showing promise for high-throughput drug

screening in physiologically relevant manner. In this

review, we summarize current research efforts which ulti-

mately lead to in vitro systems for testing drug’s effect in

PK-based manner. In particular, we highlight the contri-

bution of microscale systems towards this goal. We envi-

sion that the ‘cells-on-a-chip’ technology will serve as a

valuable link between in vitro and in vivo studies, reducing

the demand for animal studies, and making clinical trials

more effective.

Keywords Microfluidics � Pharmacokinetics �ADME

Introduction

Drug development is an expensive, time consuming pro-

cess. Despite continuing effort to improve the productivity

of the drug development process, only one out of ten drug

candidates entering the clinical trial reaches the final

approval stage [1, 2]. The number of new molecular enti-

ties (NMEs) that are approved by US FDA has been

declining over the last decade, from 53 in 1996 to 21 in

2008 [3]. The main reasons for such a low success rate are

primarily unforeseen lack of efficacy (accounting for about

30% of failures) and toxicity (accounting for another 30%)

[1]. When the lack of efficacy or unexpectedly high toxi-

city is revealed in later stages of clinical trials, it increases

the total cost, since the cost of clinical testing is consid-

erably higher than the cost of preclinical testing, increasing

with successive phases [4]. Improving the ability of in vitro

assay systems to predict the efficacy and toxicity of drug

candidates earlier in the drug discovery process will greatly

enhance productivity. Consequently, there has been a sig-

nificant amount of effort geared towards improvement of in

vitro systems for assessing drug efficacy and toxicity [5, 6].

Typically, cell-based assessment of drug toxicity and

efficacy is evaluated in multi-well microtiter plate systems.

Cells are cultured in multi-well plates, incubated with

drugs for a specific amount of time, and the viability of the

cells are measured. A large number of human cell lines

intended for specific target organ toxicity are available [7].

Although this methodology has been well-established and

has been used for several decades as a standard method for

evaluation of drugs, many drugs move onto successive

stages only to fail, due to unforeseen toxicity or lack of

efficacy. The predictability of multi-well, cell-based drug

assay is not satisfactory because this system differs from

the human body in many aspects. First, in a cell-based

J. H. Sung � M. L. Shuler (&)

Department of Chemical and Biomolecular Engineering,

Cornell University, Ithaca, NY, USA

e-mail: [email protected]

J. H. Sung

e-mail: [email protected]

M. L. Shuler

Department of Biomedical Engineering,

Cornell University, Ithaca, USA

123

Bioprocess Biosyst Eng (2010) 33:5–19

DOI 10.1007/s00449-009-0369-y

assay, only a single cell type is cultured at a time, which

means that multi-organ interaction is absent. On the other

hand, in the human body, drugs go through a complex,

dynamic process of absorption, distribution, metabolism

and excretion (collectively known as ADME) [8]. Conse-

quently, the effective drug concentration at a target site is

often different from what is expected from the adminis-

tered dosage. Pharmacokinetics (PK), which describes the

concentration profile of a drug after administration, has a

profound effect on the observed efficacy and toxicity of a

drug [2], but current in vitro cell-based assays lack the

ability to reproduce the PK of drugs. The second issue is

that the cell culture in multi-well format is different from

cells in their native environment. Cells are typically cul-

tured in 2-D monolayer, immersed in an excess amount of

liquid medium. In their tissue environment, cells are sur-

rounded by extracellular matrix (ECM) and other sup-

porting cells, with nutrients supplied by diffusion from

blood vessels. The microenvironment surrounding the cells

in 2-D culture is drastically different from that of native

tissue in terms of nutrient transport, shear stress, chemical

and mechanical signaling. As a result, cell behavior is often

not ‘authentic’, which means that cells behave differently

from the manner they would behave in their native envi-

ronment. This could be the reason behind a drug candidate

seeming to have a promising efficacy/toxicity profile in the

preclinical stage, but showing a different profile in a human

clinical testing. Thirdly, often there are individual varia-

tions in patients’ responses to a drug, resulting in unac-

ceptably high toxicity or lack of efficacy in a group of

patents while the same dosage works well for others [5].

Such individual variations are often due to genotypic or

phenotypic variations, which cause alterations in the PK of

a drug. For example, the rate at which a drug is metabo-

lized can vary depending on the enzyme level in a patient,

resulting in a different PK and toxicity profile [9]. Current

in vitro assay systems are not suitable for tackling these

issues, since these systems cannot reproduce the PK of a

drug. A mathematical approach called PK–pharmacody-

namic (PK–PD) modeling can help determine the effect of

such genotypic and phenotypic differences on the observed

pharmacological effect of a drug [2]. However, it has

limitations in that accurate parameter estimation is neces-

sary for model development, and unknown mechanisms of

action cannot be predicted with a model.

Such shortcomings of conventional cell-based assay

systems demand a more realistic in vitro system, which can

test the effect of a drug in a more systematic, that is,

PK-based way. Recently, integration of conventional cell

culture techniques and microfabrication technology

resulted in a new area called ‘cells-on-a-chip’ technology

[10, 11]. The area of cells-on-a-chip has grown rapidly

during the past few years, with the expectation that it can

offer advantages that cannot be provided by conventional

cell culture techniques. Among the many advantages of

microscale systems, the ability to fabricate complex

microscale structures and then to culture multiple cell types

inside them allows researchers to create novel microscale

systems mimicking multi-organ interactions. Such a device

allows one to observe the systematic, whole-body response

to drugs rather than the response of a single cell population.

Furthermore, the recent advance in microfluidic systems

has made it possible to precisely reproduce multi-organ

interaction with blood circulation, which has been termed

as a ‘body-on-a-chip’ approach [12]. Together with three-

dimensional cell cultures, such recent advances in micro-

scale systems can contribute to developing a more realistic,

in vitro system that can reproduce the whole-body response

to drugs (Fig. 1).

This paper reviews current research efforts for devel-

opment of microscale cell culture systems to overcome the

aforementioned limitations of conventional cell-based

assay systems. First, we summarize the recent development

in the area of cells-on-a-chip with a focus on high-

throughput implementation. Here we describe research

efforts to combine microfabrication and conventional cell

culture techniques, to create novel systems with advantages

that cannot be offered by conventional cell cultures. In the

Fig. 1 A diagram showing the concept of in vitro research efforts

towards achieving a more realistic, whole-body response to drugs.

Microfluidics and 3-D cell culture technology can be combined with

conventional cell culture techniques to enable reproduction of a

whole-body response. Microfluidics can contribute to mimicking the

dynamics of drug exposure to human body, and 3-D cell cultures can

help cells behave in a more authentic manner. Furthermore, the

typical length scale in microfluidics helps to reproduce the native

cellular microenvironment better, as it becomes easier to control

parameters such as a flow rate, shear stress, and liquid-to-cell ratio

6 Bioprocess Biosyst Eng (2010) 33:5–19

123

following section, we summarize microscale systems for

metabolism-dependent toxicity assay, and microscale sys-

tems based on the concept of physiologically based phar-

macokinetic (PBPK) modeling. Among many multi-organ

interactions, liver metabolism is the most critical element

for the realization of PK-based drug assay, and microscale

systems for reproducing the liver metabolism in vitro will

be reviewed. We will conclude with a summary of issues

that need to be overcome to achieve the PK-based drug

toxicity study, such as stable operation of microfluidic

devices and integration of real-time detection.

Advantages of combining microfabrication and cell

cultures

One obvious advantage of miniaturization is the possibility

of high-throughput implementation, and there have been

numerous examples of microscale systems for high-

throughput drug screening [13]. High-throughput imple-

mentation is necessary to screen a large number of drug

candidates in a given time, but also because it is often

required to perform a large number of tests per drug, to

obtain statistically meaningful data for various doses and

under various conditions. In case of microbial cells, we

have seen a significant amount of progress in high-

throughput and large-scale implementation in bioreactor

cultures with medium perfusion [14, 15]. With mammalian

cells, a high-throughput implementation has been achieved

at a high level in conventional multi-well assay systems,

but the area of microbioreactor systems for high-through-

put screening has begun to gain wide interest only recently.

There are unique advantages that can be offered only by

combining microfabrication/microfluidics and cell cultures.

Tailoring the architecture of microenvironment for cells is

possible, owing to the precise control over microscale

structures. The ability to easily manipulate the geometry

and the flow pattern gives researchers a control over the

transport of growth factors, reagents and oxygen inside the

system, as well as hydrodynamic shear stress upon cells

[16]. As will be reviewed further in this paper, such factors

can have an enormous influence on cell behavior and help

researchers create a more in vivo-like environment for

cells. It would be of a tremendous advantage if such novel

systems can be operated in high-throughput manner to

obtain large data set more easily.

Wang et al. reported a high-density microfluidic arrays

for cell cytotoxic assays (Fig. 2a) [17]. The device con-

sisted of fluidic channels for cell seeding, and another array

of fluidic channels for toxin exposure in an orthogonal

direction. 24 by 24 (=576) arrays of cell culture chambers

were fabricated, which were used to screen three cell types

against five toxins. Fluorescence live/dead staining was

used to evaluate the cell viability, and the result obtained

from this microfluidic array was comparable to a similar

experiment in a microtiter plate assay. This is one of the

early examples where relatively high-throughput toxicity

screening of multiple drugs against multiple cell types was

achieved on a single chip. Wu et al. developed a device

with a similar concept of orthogonal channels for cell

seeding and toxin application, but with 3-D cell culture in

agarose [18]. The device consisted of 15 microbioreactors,

which were perfused with medium using the same number

of pneumatic micropumps. Due to the use of integrated,

pneumatic pump, a significant amount of automation was

achieved with the device. An 8 by 5 microchamber array

with pressure-driven medium perfusion was developed,

and used to test seven toxins at a time [19]. A unique

terrace structure surrounding the microchamber prevented

air bubbles from invading the chambers and protected cells

inside. Microfluidic gene expression array with 16 by 16

(=256) 10-nl chambers was developed with microvalves for

row-cell seeding and column-stimulation (Fig. 2b) [20].

Using this device together with cells transfected with

fluorescent reporter genes, expression dynamics of eight

inflammatory genes were monitored in response to eight

stimuli.

As demonstrated by these examples, there has been a

significant amount of effort in the cells-on-a-chip area

during the past few years directed to automation of oper-

ation, such as easier cell loading and toxin application,

medium perfusion and evaluation of cell viability [21–24].

However, to compete with conventional microtiter plate

systems equipped with robotic liquid handling for high-

throughput experimentation, such microfluidic systems

need to demonstrate advantages other than high-throughput

processing to justify all the ‘hassle’ of using a microfluidic

system. Perhaps one of the most important and useful

advantages of a microfluidic cell culture system is that it

enables formation of well-controlled concentration gradi-

ent of chemicals inside the system. The laminar flow inside

the device restricts the transport process to be dominantly

diffusive. A concentration gradient can be formed by

simply having two parallel channels, one channel working

as a source and the other as a sink. Several other types of

more sophisticated devices have also been developed for

the formation of a more complex concentration gradient

[25]. Formation of concentration gradient offers an

advantage over multi-well plate system, since a multi-

dosage study can be performed in one shot, as opposed to

testing multiple doses individually in separate wells. In

addition, many drugs are administered in combination to

achieve a synergistic effect, but finding the optimal com-

bination ratio can be challenging. The concentration gra-

dient maker can be useful for testing various combination

ratios in a more efficient manner, rather than mixing and

Bioprocess Biosyst Eng (2010) 33:5–19 7

123

testing them individually. Recently a microfluidic gradient

maker was utilized to test the effect of various concentra-

tions of anesthetics, bupivacaine and lidocaine on myo-

blasts [26]. In this device, a gradient maker was integrated

with a cell culture chamber, so that cells are exposed to a

concentration gradient of toxins (Fig. 2c). By performing

cell viability assay after the exposure and correlating the

width across the chamber with the concentration predicted

by mathematical simulation, a quantitative dose–response

relationship was achieved. This type of study illustrates the

advantage of utilizing a microfluidic device to increase the

efficiency of drug testing. A similar concept was demon-

strated in a study by Hung, where 10 by 10 cell culture

chamber arrays were fabricated and a gradient maker was

attached to the columns [27, 28]. By introducing a flow in

horizontal direction in addition to the concentration gra-

dient in vertical direction, a series of combination ratios

were easily generated across columns and rows. The

potential of a microfluidic system for drug screening pur-

pose was demonstrated by testing the effect of a serum

concentration on cells. While these devices created a

continuous concentration gradient of chemicals, a micro-

fluidic device which generates a linear dilution of chemi-

cals on chip has been developed [29]. Nine dilutions were

generated by mixing two input fluid streams and control-

ling the geometry of fluidic channel network. Such a

controlled-microfluidic delivery of reagents illustrates the

potential of cells-on-a-chip technology to achieve novel

experimentation not possible with conventional multi-well

based platforms.

Another advantage of integrating microfabrication

technology with cell culture techniques is that the micro-

environment can be easily tailored by controlling param-

eters such as the geometry of the device structure and the

flow rate. Devices with various chamber volumes ranging

from nanoliters to several milliliters have been demon-

strated [30–32]. Such a wide spectrum of culture volumes

can have a profound effect on cell behavior by dramatically

altering the transport phenomena. To characterize cell

culture microenvironment, the concept of ‘effective culture

volume’ has been introduced, but the understanding of

microscale cell culture environment is still at its infancy

[16]. Another interesting and important parameter in a

microfluidic cell culture system is the flow rate. The effect

of flow rate on the growth kinetics of human mammalian

cells has been studied [33]. Fibroblasts were grown for up

to 2 weeks in a microbioreactor in various flow conditions,

from static (no flow) condition to high flow rate. The cells

Fig. 2 (a) High-density microfluidic array for cytotoxic assays.

24 9 24 (576) chambers are used to screen three cell types against

five toxins. The figure reproduced from [17] with permission of RSC

Publishing (b) 16 9 16 Microfluidic living cell array (shown in

yellow, layer 1). Fluid is controlled by two sets of PDMS barriers,

shown in green and purple. Cells are drawn from left to right, and

stimuli are drawn from top to bottom. Reproduced from [20] with

permission of RSC Publishing (c) A mathematical simulation of

gradient generation, reproduced from [26] with permission from

Elsevier (d) A microfluidic device for 3-D hydrogel-cell culture.

Human hepato-cellular carcinoma cells (HepG2) were encapsulated in

collagen composite, OPLA, and Puramatrix scaffolds inside the

device made of PDMS. Cellular activity (albumin production) and

cytotoxicity assay (Triton X-100) were performed using the device.

The image taken from [45] with permission from Springer

8 Bioprocess Biosyst Eng (2010) 33:5–19

123

did not grow well in either the no flow or high flow rate

(0.3 ml/h) condition, but grew best in low flow rate

(\0.2 ml/h) conditions. In a study with a similar purpose, a

microfluidic device which can generate a logarithmic range

of flow rates was developed and used to study the effect of

flow rate on the proliferation of embryonic stem cells [34].

In this study, cells exposed to a low flow rate (1 9 10-3 ll/

min, residence time of 319 min) grew considerably slower

than the cells exposed to a high flow rate (1.1 ll/min,

residence time of 0.29 min). Theses preliminary studies

have demonstrated that microfluidic environment in an

important factor in determining the behavior of culture

cells. It is also pertinent to toxicity assays, since it implies

that cell response to toxins can vary widely depending on

the microenvironment they are exposed to. A more para-

metric study on underlying mechanism of how cells

interact with surrounding microenvironment and how

microenvironment affects cell behavior is likely to yield

valuable information for creating more in vivo like

microscale cell culture systems.

It is believed and has been demonstrated that a 3-D cell

culture can offer a more in vivo-like environment for cells

than 2-D monolayer culture by providing a more realistic

cell–cell interaction, chemical and mechanical signals [35,

36]. Encapsulation of cells in 3-D matrix has been utilized

for various purposes, for both mammalian and microbial

cells [37]. It has been demonstrated that offering cell

adhesion, growth and organization in 3-D space can induce

authentic response of cells, leading to in vivo like differ-

entiation, proliferation, metabolic activity, and metastasis

potential of tumor cells [38–41]. It is also notable that the

response of tumor cell lines to chemotherapeutic agents can

change drastically (more than 1,000-fold) when they were

cultured in 3-D environment compared to 2-D monolayer

[42].

Although the importance of 3-D cell culture has been

well-recognized, one limitation of 3-D cell culture is an

inefficient transport into the matrix. With diffusive trans-

port being dominant in 3-D matrix, often nutrients and

oxygen are not able to reach the inside area of 3-D cell

culture matrix at sufficiently high level, especially in case

where the size of matrix is greater than a millimeter range.

Given the ability of microscale systems to precisely control

transport of molecules, it is likely that combination of 3-D

cell cultures and microfabrication techniques will yield an

in vitro system with more efficient and controlled transport.

In a recent study, hydrogel-cell matrix modules of sub-mm

size were made using a PMDS mold [43]. A PDMS

membrane with sub-mm size holes was fabricated, which

was filled with cell-hydrogel mixture in liquid. After the

gel formation the gel modules were removed from the

PDMS membrane, creating cell-hydrogel modules with

sub-mm size. The feasibility of the method was tested with

collagen, MatrigelTM

and agarose, and various cell densities

(105–108 cells/cm3) were obtained. The size of the fabri-

cated modules was determined by the thickness of the

PDMS membrane (down to 40 lm) and the size of holes

(200, 500, and 1,000 lm).

Incorporation of 3-D cell cultures into a microfluidic

device has been gaining increasing attention recently. Kim

et al. reported a 3-D microfluidic platform for a cell-based

assay (Fig. 2d) [44, 45]. A hepatoma cell line was cultured

inside a peptide hydrogel in the device, and the cytotoxicity

assay was performed using a linear concentration gradient

of Triton X-100. A microfluidic system with immobilized

hydrogel-encapsulated mammalian cells was developed

[46]. Poly(ethylene) glycol (PEG) hydrogel was photoli-

thographically polymerized in a microfluidic device and

the cytotoxicity assay was performed. Torisawa et al.

developed a 3-D cell culture array using collagen as a

matrix [47]. Human breast cancer (MCF-7) cells were

grown inside the 4 by 5 array of collagen matrix and

proliferation of cells was monitored for 5 days. In addition

to the cytotoxicity assay result, an interesting result was

observed when the proliferation kinetics of the cells was

compared with that of the same cells grown in 2-D

monolayer. The cells in 3-D matrix grew significantly

slower than the cells in monolayer, which again demon-

strates that 3-D environment causes a significant change in

the cell behavior.

Microscale systems for metabolism-dependent toxicity

study

Testing metabolism-dependent toxicity requires at least

two components, that is, activating system (metabolism)

and target cells. A simple and straightforward way to

achieve this at macroscale level is to incubate both systems

in the same cell culture well. Such an early attempt was

made in 1980 by Spielberg et al., who incubated human

lymphocytes with a drug in the presence of liver micro-

somes [48]. The metabolizing microsomes were removed

after 2 h of exposure, and cells were further incubated for

16 h before cell viability was measured. This system was

used to study the metabolism-dependent toxicity of several

antiepileptic drugs. Tabatabaei et al. developed a system

with a similar concept, using an external metabolizing

system (rabbit microsomes) and lymphocytes as target cells

[49]. The major modification made by the authors was

implementation of a membrane-impermeable fluorescent

dye and multiwell plate scanner, which gave the advantage

of high-throughput cell viability assay over the trypan blue

exclusion assay used in the previous study. In a more recent

study by Vignati et al., metabolism-dependent toxicity

was assessed by combining CYP3A4 bioactivation with

Bioprocess Biosyst Eng (2010) 33:5–19 9

123

hepatoma cell line (HepG2) [50]. Cytochrome P450 (CYP)

is the major metabolizing enzyme family in the liver, and

CYP3A4 is an isozyme that is involved in about 50% of

drugs currently on the market [51]. The activating system

was either human CYP3A4 cDNA expressed microsomes,

or HepG2 cell line transiently transfected with CYP3A4.

The HepG2 cell line was used as a target cell, since it is

widely used for evaluating hepatotoxicity and it also con-

tains coenzyme NADPH-cytochrome reductase, which is

required for CYP-mediated drug metabolism. Ten drugs in

various categories were tested, and the metabolism-induced

alteration in the cytotoxic effect was observed. Primary

human hepatocytes have also been gaining attention as an

in vitro system for assessing metabolism-dependent toxic-

ity [52]. Primary hepatocytes are known to have metabolic

activity profile similar to the liver, at least immediately

after isolation. However, the poor availability, the high

instability of enzyme activity (the enzyme level drops

drastically with increasing culture time) has prevented a

wider application of primary hepatocytes. All of these

approaches can mimic some aspect of metabolism-depen-

dent toxicity, but macroscale devices have limitations that

make high-throughput implementation inherently difficult,

and reproduction of the liver function is incomplete.

During the last few years, there have been several

attempts to develop an in vitro microscale system for

assessing metabolism-dependent toxicity. Microscale

systems offer advantages of easier high-throughput

implementation, and physiologically realistic environment

for more complete liver function, as will be discussed in

more detail later. One prominent example was reported by

Lee et al., who developed a 15 9 35 array of sol–gel

encapsulated P450 enzymes (Fig. 3a, denoted by the

authors as MetaChip). A prodrug was added onto each spot,

which would generate the active drug, and cytotoxicity was

assessed by overlapping with a layer of target cells (breast

cancer cell line MCF-7). Two main cytochrome P450

isozymes CYP 3A4 and 2B6 were prepared, and three

prodrugs (cyclophophamide, Tegafur, and acetaminophen)

were tested. All three drugs showed toxicity profiles similar

to a solution control (CYP enzymes in solution rather than

sol-gel droplets), which verified that the nanoliter scale sol-

gel arrays could produce comparable toxicity assay results,

but in a more high-throughput manner. The system was

further improved with the development of a 20 by 54 array

of hydrogel-encapsulated cells (called DataChip by

authors), which can be superimposed on the MetaChip with

a CYP enzyme array [53]. Using this system, three CYP

isozymes or mixtures of the three against nine compounds

were tested. The IC50 values obtained from this system

were also comparable to the results from a conventional

96-well plate assay.

As mentioned earlier, microfluidics provides an ideal

platform for fabricating and connecting several components

Fig. 3 Microscale systems for

metabolism-dependent toxicity

assay. a CYP enzymes are

spotted in an array (MetaChip),

and target drugs are incubated

with the enzymes, which

generates metabolites. DataChip

is provided by spotting an array

of target cells. By

superimposing the two and

incubating, the effect of the

target drugs and their

metabolites can be tested. The

artwork was drawn as described

in [53]. b A three-layer

microfluidic device consists of

cell culture chambers, human

liver microsomes (HLM), and

fluidic channels for medium

perfusion and metabolites

detection, as described in [54].

Via diffusion, the drugs and

their metabolites are transported

to the cell culture chamber.

c The concept of ‘wells-within-

a-well’, which allows

interaction of multiple cell types

through overlying cell culture

medium [59]. The artwork was

drawn as described

10 Bioprocess Biosyst Eng (2010) 33:5–19

123

in a single device. Recently, a microfluidic device for testing

metabolism-dependent drug toxicity was developed. This

microfluidic integrates a sol-gel human liver microsomes

(HLM) bioreactor with a cell culture chamber array [54].

This device consists of three layers (PDMS-quartz-PDMS),

quartz layer in the middle with microwells for HLM and

bottom PDMS layer for cell culture chambers. A drug con-

taining solution is introduced continuously into the top

PDMS layer and diffuses into HLM in the middle layer, and a

drug together with metabolites generated from the HLM

diffuse further down to the cell culture chamber (Fig. 3b).

Acetaminophen (AP) and its UDP-glucuronosyltransferase

(UGT) metabolism were tested using HepG2 as a target cell

line for hepatotoxicity. AP undergoes detoxification by

competing phase II conjugation reactions, glucuronidation

and sulfation, which convert AP to nontoxic conjugates for

elimination in bile or urine [55]. As expected, upon metab-

olism of AP to nontoxic glucuronidation metabolite (APG),

its cytotoxic effect was observed to be reduced. When

overdosed AP saturates the UGT detoxification pathway, it is

metabolized by CYP enzymes to a toxic metabolite, quinine

imine. When authors blocked the UGT detoxification path-

way by omitting uridine 50-diphosphate glucuronic acid

(UDPGA, coenzyme for UGT detoxification reaction) from

the reaction mixture, AP was metabolized by CYP and the

cytotoxicity of AP was increased. Furthermore, a metabo-

lism-based drug–drug interaction was tested. Addition of

UGT inhibitor such as phenotin would result in increased

drug toxicity, which was again observed experimentally.

Another notable feature of this device is that the microfluidic

device was integrated with a UV-absorbance detection sys-

tem previously developed by the authors, which enabled

simultaneous detection of the drug and its metabolite con-

centrations [56].

Achievement of multi-organ interaction in vitro inevi-

tably requires seeding multiple cell types in a device with a

precise control. A novel method of direct 3-D cell writing

for in vitro PK model has been developed [57]. A 3-D cell

culture in alginate was created by the syringe-based, layer

by layer bioprinting method, which is to be enclosed by a

housing made from a soft-lithography micropatterning

technique. The proliferation and urea synthesis rate of a

hepatoma cell line (HepG2) encapsulated in alginate matrix

was monitored. This method has a direct implication for in

vitro PK-based drug testing, since it will allow free-form

fabrication of multiple cell types onto a device, which can

be used to reproduce multi-organ interaction. Nevertheless,

direct-writing of only one cell type was demonstrated in

this study, and a further study of integrating the cell matrix

with the microfluidic device is required to achieve this

goal.

An in vitro platform with multiple cell types has been

developed by having small, separate inner-wells inside a

larger well [58, 59]. Termed as ‘‘wells-within-a-well’’

concept by the authors, multiple cell types are cultured

separately in small wells (Fig. 3c). This provides a simple,

rudimentary method to reproduce multi-organ interaction,

without different cell types physically contacting one

another. Cell culture medium is shared by flooding the

inner wells and filling the large well. A chemotherapeutic

agent tamoxifen was tested against six cell types, liver

(hepatocytes), kidney (kidney cortical cells), lung (small

airway epithelial cells), central nervous system (astro-

cytes), blood vessels (aortic endothelial cells), and breast

cancer (MCF-7). Using this device, EC50, EC90, EC99

(concentration to cause 50, 90, and 99% decrease in sur-

vival, respectively) values were obtained for the six cell

types. Unfortunately, multi-organ interaction was not

characterized by the authors, nor any comparison with

separate cell culture tests was made. However, it is plau-

sible that such a system will be able to mimic at least some

aspect of multi-organ interaction.

In an effort to combine a conventional perfusion

bioreactor with a microscale cell culture device, a hepa-

tocyte-bioreactor coupled was developed to assess hepato-

activated transformation of substrates [60]. In this device,

primary pig hepatocytes were cultured in a recirculating

50 ml perfusion bioreactor, which was connected to

a commercially available cell culture chamber called

microphysiometer at a flow rate of 150 ll/min. Inside the

microphysiometer the target cells, ZR 751, a breast cancer

cell line was cultured at 1.5 9 105 cells/chamber. As a

model drug, cyclophophamide (CYCL) was fed into the

bioreactor with liver cells, and after seven hours the via-

bility of the target cells were measured. The viability of ZR

751 cells dropped only in the presence of the prodrug

(CYCL) and the bioreactor activation.

All of the in vitro systems described so far attempt to go

beyond testing the direct effect of a drug, and reproduce

metabolism-dependent actions of the drug. One potentially

important aspect which these systems lack is the ability to

mimic the dynamics of multi-organ interaction; that is,

cells are not simply exposed to the drug or its metabolites

in a bolus dose, but the circulation of blood and transport

inside tissue environment alter the dynamics of drug

exposure.

Microscale cell culture analog (lCCA) based

on a PBPK model

Pharmacokinetics refers to the time-dependent concentra-

tions of a substance in a living system [61]. It examines the

ADME processes. A PK model is a mathematical model

which attempts to predict the concentration profile of a

drug in the blood or at the target site from a given dose.

Bioprocess Biosyst Eng (2010) 33:5–19 11

123

PK models can be categorized into several subtypes,

depending on their complexity of structure [62]. The most

comprehensive model, a physiologically based PK

(PBPK) model, is based on physiological consideration.

They comprise of several compartments, each represent-

ing tissue and organs. These compartments are connected

to one another with hypothetical blood flow, in the same

organization as the blood circulation. The volume of each

compartment is same as actual physical volume of a

corresponding organ. Physiological blood flow rates into

and out of each organ are given as the flow rates into and

out of each compartment, respectively. A set of simple

mass-balance equations accounting for flux in, flux out,

absorption, and reactions is constructed for each com-

partment, and the ordinary differential equations (ODEs)

are solved using numerical methods. A large number of

commercial mathematical software packages are also

available [63]. The output result of PBPK model is the

time-dependent concentrations of a drug and its metabo-

lites in each compartment, which can be directly com-

pared with experimental results.

The microscale cell culture analog (lCCA), also known

as a ‘body-on-a-chip’ or an ‘animal-on-a-chip’, was con-

ceived as a physical realization of a PBPK model, as an in

vitro system to emulate body’s dynamic response to drugs

[12]. Just like a PBPK model, compartments representing

different organs are connected with fluidic channels mim-

icking blood circulation, and in each chamber the corre-

sponding cell type is cultured. Before the advance of

microfabrication technology, the prototype cell culture

analog device was constructed with two milk dilution

bottles and a spinner flask, connected with Teflon tubing

and a peristaltic pump [64]. Lung and liver-related cell

lines were cultured in the two bottles separately, and

toxicity of naphthalene and its metabolites generated from

the liver cell line was observed. Since the first develop-

ment, several variations of the device were developed,

along with a mathematical PBPK model that described the

system [65–68].

Following the integration of microfabrication technol-

ogy with cell culture techniques, a microscale version of

cell culture analog device was developed (lCCA) [69].

Incorporation of microfabrication technology offers several

advantages; (1) fluidic residence times, which determine

the dynamics of drug exposure to each organ, can be easily

controlled to mimic physiological values. (2) fluidic shear

stress can be controlled to be in a physiological range for

specific cell types (3) microscale devices allow incorpo-

ration of authentic tissue samples, for example tumor

biopsy samples. As a proof-of-concept study, naphthalene

toxicity was assessed using the lCCA. It was demonstrated

that naphthaquinone, rather than naphthalene epoxide,

was the reactive metabolite generated from the liver

compartment causing glutathione depletion and loss of

viability in the lung compartment [70].

Since the initial development, several improvements

have been made. The device assembly and operation have

been optimized for long-term experimentation (several

days, rather than several hours), and multi-drug resistance

(MDR) and combination chemotherapy have been tested

using the device [71]. In this four-chamber lCCA, cells

representing the liver (HepG2/C3A), bone marrow (MEG-

01), uterine cancer (MES-SA), and MDR variant of uterine

cancer (MES-SA/DX-5) were cultured in the device

(Fig. 4a and (c)), and treated with various combinations of

drugs for either 24-h acute toxicity or 72-h proliferation. In

both studies, it was demonstrated that combining a che-

motherapeutic, doxorubicin, with modulators (cyclosporine

and nicardipine) resulted in greater efficacy than doxoru-

bicin or modulators alone. Furthermore, the combination

was shown to be particularly effective against MDR variant

cell line (MES-SA/DX-5), which was not observed in

96-well plate assays. This cell-specific synergy is believed

to be due to the pharmacokinetic interaction between

compartments. PBPK models for human and the lCCA

device were constructed with the predicted pharmacokinetic

profiles of a drug and its metabolites.

It is believed that 3-D hydrogel-cell cultures can elicit

more authentic behavior from culture cells [36]. The

lCCA device has been modified to accommodate 3-D

hydrogel-cell construct, and the metabolism-dependent

toxicity of Tegafur–uracil combination has been observed

[72]. In this three-chamber device, cells representing the

liver (HepG2/C3A), colon cancer (HCT-116), and bone

marrow (Kasumi-1) were cultured (Fig. 4b and d). Unlike

previous lCCA studies where cells were cultured in

monolayer, cells were encapsulated inside hydrogel

(alginate or MatrigelTM

). Tegafur, a drug widely used to

treat colon cancer, is a prodrug which needs activation in

the liver to become an active (toxic) metabolite 5-fluo-

rouracil (5-FU). Tegafur showed a cytotoxic effect in a

lCCA due to the metabolism by the liver cells, whereas it

had a negligible effect in a 96-well plate assay with colon

cancer cells only. The metabolism-dependent toxicity was

further verified by treating cells with Tegafur in a lCCA

with the liver chamber empty, in which case the cytotoxic

effect was significantly reduced. Uracil acts as an inhib-

itor of the liver enzyme dihydropyrimidine dehydrogenase

(DPD), which is responsible for the catabolism of 5-FU,

the active metabolite of Tegafur. Uracil is often added to

Tegafur as a modulator, since inhibition of 5-FU metab-

olism results in increased efficacy of the drug [73]. In a

lCCA, addition of uracil also enhanced the cytotoxic

effect of Tegafur on tumor cells. These results demon-

strate the strength of lCCA systems in testing the com-

bination of drugs in pharmacokinetic-based way, which

12 Bioprocess Biosyst Eng (2010) 33:5–19

123

would have been possible only in animal or human

studies.

Microscale systems for reproducing biotransformation

in vitro

Perhaps the most important prerequisite for the realization

of in vitro PK-based drug toxicity study is to reproduce the

liver metabolism in vitro. Biotransformation, especially in

the liver, has a significant effect on the pharmacokinetic

profile and the pharmacological effect of a drug [74].

Metabolism in the liver is responsible for not only detox-

ification, but also activation of a drug and drug–drug

interactions. Drug biotransformation is divided into two

types, phase I (hydrolysis, oxidation and reduction), and

phase II (conjugation) [75]. The cytochrome P450 (CYP)

enzymes are the major enzymes in the phase I reactions,

and are divided into several isoforms. Among many known

isoforms, the majority of metabolic reactions are carried

out by CYP 1A2, 2C9, 2D6, and 3A4 [74]. Drug–drug

interactions occur when one drug either induces or inhibits

the activity of an enzyme that is responsible for the

metabolism or absorption of another drug. Alternatively,

drugs can interact due to their pharmacodynamic proper-

ties, for example compete for the same receptor or

enzymes.

Prediction of drug metabolism and potential drug–drug

interactions is critical for successful drug development, and

there have been enormous amount of efforts directed

toward reproducing the liver metabolism in vitro [75].

Currently, there is a whole range of in vitro systems

available to researchers, from cDNA-expression systems to

perfused liver tissues. Unfortunately, each system has pros

and cons, and yet there is no single system that can

reproduce the liver function at a satisfactory level.

Advantages and disadvantages of various in vitro systems

are well documented in several review articles [75–77].

Fig. 4 Examples of microscale cell culture analog (lCCA) devices.

a A four-chamber lCCA for testing MDR cancer. Cell lines

representing the liver (HepG2/C3A), MDR-variant uterine cancer

(MES-SA/DX-5), normal uterine cancer (MES-SA), and bone marrow

(MEG-01) are cultured in chamber 1, 2, 3, and 4, respectively. The

image was reproduced from [71] with permission. Copyright 2009,

Wiley Periodicals, Inc. b A lCCA for testing colon cancer drugs with

three chambers. Cell lines cultured are 1: HCT-116 (colon cancer), 2:

HepG2/C3A (liver), and 3: Kasumi-1 (marrow) [72]c. A PBPK model

corresponding to the four-chamber lCCA for MDR cancer. d A

PBPK model corresponding to the three-chamber lCCA for colon

cancer. Note that the chambers are interconnected differently in the

two cases, to mimic the physiological blood circulation. The

chambers in a lCCA and the corresponding compartments in a

PBPK model are marked with numbers. The medium reservoir, which

corresponds to the plasma/other tissues, is not shown in the figure

Bioprocess Biosyst Eng (2010) 33:5–19 13

123

Brief descriptions, pros and cons of conventional in vitro

systems are summarized in Table 1.

Microscale systems offer unique advantages in control-

ling the transport of nutrients, mechanical stimulation,

surface interaction, and organizing cells in a defined

geometry. These advantages have been exploited by

researchers to develop an in vitro system with an improved

liver function. In a typical length scale of a microfluidic

system, a laminar flow is generated and diffusion plays a

dominating role in the transport of nutrient and oxygen.

With a combination of a mathematical modeling, concen-

tration of oxygen and nutrients can be predicted and con-

trolled [78, 79]. A microfluidic flat-bed bioreactor was

created, in which rat hepatocytes and non-parenchymal

cells were co-cultured (Fig. 5a) [80]. By controlling the

flow rate, it was possible to create a gradient of oxygen

concentration across the bioreactor, so that cells are

exposed to a heterogeneous oxygen environment. The

oxygen gradient in the bioreactor mimics the physiological

condition of the actual liver, where the localized function

of the liver is thought be controlled by the local oxygen

concentration [81]. For example, a gradient of oxygen

concentration is formed from the inlet at the periportal side

(high oxygen region), where glucose, albumin, urea bio-

synthesis and glutathione conjugation take places, to the

perivenous outlet (low oxygen region), where glucuroni-

dation and oxidation by CYP enzymes mainly occur. This

has an implication that a low oxygen tension might be

beneficial to achieve high CYP enzyme activity of cultured

hepatocytes. Indeed, the authors found that a differential

CYP activity was measured across the length of the bio-

reactor, and perfusion of acetaminophen, which is known

to have liver toxicity, caused the most damage to the cells

in the area of low oxygen concentration. This example

demonstrates the power of microfluidic system in control-

ling the transport process for more authentic cell function.

PDMS (Polydimethylsiloxane) has been the most com-

monly used material for microfluidic devices, owing its

popularity to several advantages it can offer, such as bio-

compatibility, gas permeability, and optical transparency.

One of the earliest attempts to culture liver-related cells in

a PDMS microfluidic device was demonstrated by a series

of papers by Leclerc et al., who developed a microfluidic

device for culturing HepG2 cells in 3-D by stacking two

layers of PDMS. Cells were maintained in the device for

several days, and cells were observed to attach and grow

successfully on the surface of PDMS. The measurement of

glucose consumption and albumin production confirmed

that the metabolic activity of the cells was maintained

during the culture period [82]. In a subsequent paper, the

authors attempted a high-density cell culture by stacking

10 layers of PDMS, and achieved the cell density of up to

4 9 107 cells/cm2, which is close to the density in a

macroscale bioreactor [83]. In addition to the HepG2 cell

line, authors also demonstrated culturing fetal human

hepatocytes (FHH) in the same device, and successfully

Table 1 In vitro systems for reproducing liver metabolism

System Description Advantages Disadvantages

cDNA-expression system

(supersomes or

baculosomes)

Human CYP and UGT can be

transfected into insect cells

and microsomes are

separated

Useful for characterizing

single enzyme reaction in

high-throughput manner

May lack in vivo relevance

since whole liver enzyme

repertoire is lacking

Human liver microsomes

(HLM)

Separated from liver tissues

by centrifugation, and

commercially available

Whole array of CYP and

UGT enzymes present

Cytosolic and phase II

enzymes are lacking

Relatively simple to use high-

throughput implementation

Can be recovered from frozen

tissue samples

Liver cell line Immortalized cell lines

originated from

hepatocellular carcinoma

Unlimited availability and

easy handling

Only a subset of in vivo liver

functions is retained

Hepatocytes Primary hepatocytes isolated

from the liver

Good reproduction of liver

metabolic profile

Limited culture time and

rapid loss of enzyme

activity (4 weeks in

maximum)

Relatively high-throughput No 3-D structure

Liver slices or perfused

liver

Best representation of in vivo

liver function

3-D cell–cell and cell–matrix

interactions are intact

Limited availability of human

models

Function is preserved for

only a few hours

14 Bioprocess Biosyst Eng (2010) 33:5–19

123

demonstrated that the perfusion culture in a microfluidic

device enhanced the metabolic function of cells in both

FHH and HepG2 cells [84].

One way to improve the liver-specific function of

hepatocytes is to co-culture them with various supporting

cell types, such as epithelial, Kupffer, fibroblasts [85]. In

conventional methods, those cells would be cultured in a

random mixture, with the only available control on the

ratio of mixing the cell populations. With the advent of

micropatterning techniques, it has become possible to

organize multiple cell types in a defined geometry [86].

One of the techniques has been used to create a micro-

patterned co-culture of rat hepatocytes and fibroblasts [87].

A PDMS stencil with through-holes was used to selectively

seed hepatocytes in a defined area of a polystyrene well,

where rest of the area was filled with fibroblasts. The

maximum liver function was observed at 500 lm size of

hepatocytes ‘islands’, with 1,200 lm spacing. Gene

expression profiles, metabolizing enzyme activity, secre-

tion of liver-specific proteins and response to hepatotoxins

were measured and the liver-specific functions were found

to be maintained for several weeks. The simplified proce-

dure of the cell patterning is shown in Fig. 5b.

Mechanical stimulation, a shear stress in particular, is

also thought play an important role in modulation of the

tissue-specific function [88, 89]. In addition, organization

of cells in 3-D, rather than 2-D monolayer culture, is

thought to provide essential chemical/mechanical stimula-

tions necessary for authentic cell behavior [36]. Microflu-

idic systems are ideal for providing both aspects with a

precise control. A microfluidic bioreactor with perfused

3-D liver cell culture was developed (Fig. 5c) [90]. Rat

primary hepatocytes were cultured in 3-dimensional con-

figuration made by through-holes in a silicon wafer, while

providing hydrodynamic shear at a physiological level

(\2 dyne/cm2). After up to 2 weeks of culture, cells rear-

ranged to form tissue-like structures. Preaggregation of

cells into spheroid-like form prior to seeding into the

device further improved the liver-specific cell behavior,

such as albumin secretion and urea synthesis [91]. This

concept was further developed into a scalable, multi-unit

structure and it was demonstrated from gene expression

profiles and biochemical activity measurements that the rat

primary hepatocytes cultured in this system was closer to

the human liver than 2-D collagen sandwich culture or 3-D

MatrigelTM

culture [92].

Recently, Vozzi et al. described a multicompartment

bioreactor (MCB), where multiple cell types are cultured in

separate chambers of a single device, connected with flu-

idic conduits (Fig. 5d) [93]. This is similar to the concept

Fig. 5 Microscale systems for

reproducing the liver function in

vitro. a A concentration

gradient of oxygen is formed

inside a flat-bed microreactor

with hepatocytes. The oxygen

concentration gradient mimics

the physiological situation of

the liver, in which different liver

functions are active depending

on the position and the

local oxygen concentration.

The microreactor is drawn

as described in [80].

b Micropatterning of

hepatocytes with supporting cell

types was shown to enhance the

liver-specific function.

A simplified procedures for the

cell patterning is shown, as

described in [87]. c Microfluidic

perfusion cell culture system,

where hepatocytes were

cultured in 3-dimensional

organization in a silicon

scaffold, as described in [90–

92]. d A simplified schematic of

a multi-compartmental

bioreactor (MCB) with

hepatocytes and human

umbilical vein endothelial cells

(HUVEC), as described in [93]

Bioprocess Biosyst Eng (2010) 33:5–19 15

123

of a lCCA, as a downscaled in vitro ‘human body’. One

thing notable in this study was that an attempt was made to

scale down the human body by using an allometric scaling

law, to conserve the ratio relationships of kinetic, meta-

bolic, volumetric parameters between cells. In this study,

the authors investigated the crosstalk between hepatocytes

and endothelia cells by culturing murine hepatocytes and

human umbilical veins endothelial cells (HUVEC) in the

device and measuring the rate of glucose consumption,

albumin and urea synthesis. An enhancement in the albu-

min and urea synthesis was observed in case when the

hepatocytes were co-cultured with HUVEC in the device,

even without a direct physical contact.

Outlook and conclusion

Cells-on-a-chip technology is a promising field, and it is

likely that in the next few years we will be seeing a

plethora of microscale devices for toxicity studies. The

concept of pharmacokinetic-based testing has been dem-

onstrated at a proof-of-concept level [68, 70–72]. In par-

ticular, combination of a mathematical modeling approach

such as a PBPK modeling and an in vitro platform such as a

lCCA can provide a valuable insight into the mechanisms

for drug toxicity [66]. A mathematical modeling has been

used to test various dosing strategies such as drug combi-

nation ratios and dosing schedule [94], and a microfluidic

system such as a lCCA can work as an in vitro platform for

verifying the mathematical simulation. However, there are

several obstacles that need to be overcome before it

becomes widely accepted outside the research community.

The first, and the most important obstacle is the lack of

realistic in vitro liver models, which has been discussed in

detail in the previous section. The second issue is the

problem of detection and analysis in microscale systems.

Miniaturization offers many advantages but also renders

detection and analysis difficult. Smaller sample size, for

example liquid medium in nanoliter scale, makes it difficult

to analyze the sample with many conventional analytical

methods. The device in microscale, especially when con-

nected with an external pump for medium perfusion, is

difficult to visually examine under a microscope in real-

time. The problem is further aggravated when multiple

microscale devices need to be analyzed for high-through-

put implementation. Consequently, there have been active

research efforts in improving or modifying current ana-

lytical techniques to adapt to the new microscale devices

[95–99]. While these systems have relied on fluorescent

signal, which is the most widely used method for non-

invasive detection, there are other approaches using elec-

trical signal [100, 101] or cellular impedance to monitor

cell viability [102], which may be advantageous for high-

throughput screening. The third challenge is the straight-

forward and stable operation of microscale systems,

especially microfluidic systems with cell cultures. The

Table 2 Practical issues in microfluidic systems and solutions

Issue Description Possible solution References

Bubble formation Microfluidic systems are susceptible to air

bubble formation, due to surface tension

in a micrometer length scale. Bubbles

can block channels, distort flow pattern,

and damage cells.

Use in-line or separate bubble traps [44, 104–107]

Place medium reservoir in elevated

position to increase pressure

Extensive rinsing with liquid with low

surface tension (ethanol or surfactant)

prior to the experiment to flush out air

completely

Bacterial contamination Bacterial contamination should be

avoided for mammalian cell culture.

Microfluidic devices require several

components from different various

materials, which may require different

sterilization methods.

Autoclaving [69, 71, 72, 104]

Washing with (acidified) ethanol

Air or oxygen plasma cleaning

Ethylene oxide sterilization

Effect of shear Shear stress from the flow can affect the

physiology and the viability of cells

Groove structure to protect cell layer from

flow

[90, 109]

Encapsulation of cells in 3-D matrix

Authenticity of cell function Cells cultured in vitro often display cell

functions different from their native

environment

Co-culture with supporting cells [87, 92]

3-D cell culture

High-throughput implementation Complex fluidic connections and flow

control required in microfluidic systems

present challenge in high-throughput

experimentation and automation

Microfluidic array [19, 27, 110]

Automated flow control

16 Bioprocess Biosyst Eng (2010) 33:5–19

123

utility of microscale systems have already been proven, but

much progress has to be made to make the systems simple,

but still versatile at the same time before they are adopted

by researchers in medicine or life sciences [21, 103]. In line

with this effort, several critical issues with microfluidic cell

culture systems have been discussed [104], and some of the

issues and their suggested solutions are summarized in

Table 2. In particular, formation of air bubbles in micro-

fluidic channels have been one of the most serious prob-

lems in microfluidic perfusion cell cultures, and several

researchers have developed microfluidic bubble traps with

different designs [105–107]. The achievements made by

many research labs in developing various microfluidic

components to enable more high-throughput, automated

multiplexing and screening will foster the development of

user-friendly microscale devices [108]. As these challenges

are overcome with novel solutions, we foresee that the

concept of PK-based drug testing will become more widely

accepted and will be able to complement the animal and

human studies, ultimately reducing the costs and time of

drug discovery process.

Acknowledgments This work was supported in part by Nanobio-

technology center (NBTC, project CM-2 (Nanotechnological

Assessment of Drug Toxicity)), NSF (National Science Foundation),

and CNF (Cornell Nanoscale Science and Technology Facility), and

by the Army Corp of Engineers (CERL) W9132T-07.

References

1. Kola I, Landis J (2004) Can the pharmaceutical industry reduce

attrition rates? Nat Rev Drug Discov 3:711–715

2. Dingemanse J, Appel-Dingemanse S (2007) Integrated phar-

macokinetics and pharmacodynamics in drug development. Clin

Pharmacokinet 46:713–737

3. Hughes B (2009) 2008 FDA drug approvals. Nat Rev Drug

Discov 8:93–96

4. Dickson M, Gagnon JP (2004) Key factors in the rising cost of

new drug discovery and development. Nat Rev Drug Discov

3:417–429

5. Andersen ME, Krewski D (2009) Toxicity testing in the 21st

century: bringing the vision to life. Toxicol Sci 107:324–330

6. Schoonen WG, Westerink WM, Horbach GJ (2009) High-

throughput screening for analysis of in vitro toxicity. EXS

99:401–452

7. Davila JC, Rodriguez RJ, Melchert RB, Acosta D Jr (1998)

Predictive value of in vitro model systems in toxicology. Annu

Rev Pharmacol Toxicol 38:63–96

8. Theil FP, Guentert TW, Haddad S, Poulin P (2003) Utility of

physiologically based pharmacokinetic models to drug devel-

opment and rational drug discovery candidate selection. Toxicol

Lett 138:29–49

9. van Kuilenburg AB (2004) Dihydropyrimidine dehydrogenase

and the efficacy and toxicity of 5-fluorouracil. Eur J Cancer

40:939–950

10. El-Ali J, Sorger PK, Jensen KF (2006) Cells on chips. Nature

442:403–411

11. Park TH, Shuler ML (2003) Integration of cell culture and

microfabrication technology. Biotechnol Prog 19:243–253

12. Khamsi R (2005) Labs on a chip: meet the stripped down rat.

Nature 435:12–13

13. Yang ST, Zhang X, Wen Y (2008) Microbioreactors for high-

throughput cytotoxicity assays. Curr Opin Drug Discov Devel

11:111–127

14. Kim BS, Lee SC, Lee SY, Chang YK, Chang HN (2004) High

cell density fed-batch cultivation of Escherichia coli using

exponential feeding combined with pH-stat. Bioprocess Biosyst

Eng 26:147–150

15. Lee PC, Lee SY, Chang HN (2008) Cell recycled culture of

succinic acid-producing Anaerobiospirillum succiniciproducensusing an internal membrane filtration system. J Microbiol Bio-

technol 18:1252–1256

16. Walker GM, Zeringue HC, Beebe DJ (2004) Microenvironment

design considerations for cellular scale studies. Lab Chip 4:91–97

17. Wang Z, Kim MC, Marquez M, Thorsen T (2007) High-density

microfluidic arrays for cell cytotoxicity analysis. Lab Chip

7:740–745

18. Wu MH, Huang SB, Cui Z, Lee GB (2008) A high throughput

perfusion-based microbioreactor platform integrated with

pneumatic micropumps for three-dimensional cell culture. Bio-

med Microdev 10:309–319

19. Sugiura S, Edahiro J, Kikuchi K, Sumaru K, Kanamori T (2008)

Pressure-driven perfusion culture microchamber array for a

parallel drug cytotoxicity assay. Biotechnol Bioeng 100:1156–

1165

20. King KR, Wang S, Irimia D, Jayaraman A, Toner M, Yarmush

ML (2007) A high-throughput microfluidic real-time gene

expression living cell array. Lab Chip 7:77–85

21. Meyvantsson I, Warrick JW, Hayes S, Skoien A, Beebe DJ

(2008) Automated cell culture in high density tubeless micro-

fluidic device arrays. Lab Chip 8:717–724

22. Warrick J, Meyvantsson I, Ju J, Beebe DJ (2007) High-

throughput microfluidics: improved sample treatment and

washing over standard wells. Lab Chip 7:316–321

23. Lee PJ, Ghorashian N, Gaige TA, Hung PJ (2007) Microfluidic

system for automated cell-based assays. JALA Charlottesv Va

12:363–367

24. Lee PJ, Gaige TA, Hung PJ (2009) Dynamic cell culture: a

microfluidic function generator for live cell microscopy. Lab

Chip 9:164–166

25. Keenan TM, Folch A (2008) Biomolecular gradients in cell

culture systems. Lab Chip 8:34–57

26. Tirella A, Marano M, Vozzi F, Ahluwalia A (2008) A micro-

fluidic gradient maker for toxicity testing of bupivacaine and

lidocaine. Toxicol In Vitro 22:1957–1964

27. Hung PJ, Lee PJ, Sabounchi P, Lin R, Lee LP (2005) Contin-

uous perfusion microfluidic cell culture array for high-

throughput cell-based assays. Biotechnol Bioeng 89:1–8

28. Lee PJ, Hung PJ, Rao VM, Lee LP (2006) Nanoliter scale

microbioreactor array for quantitative cell biology. Biotechnol

Bioeng 94:5–14

29. Walker GM, Monteiro-Riviere N, Rouse J, O’Neill AT (2007) A

linear dilution microfluidic device for cytotoxicity assays. Lab

Chip 7:226–232

30. Weuster-Botz D, Puskeiler R, Kusterer A, Kaufmann K, John

GT, Arnold M (2005) Methods and milliliter scale devices for

high-throughput bioprocess design. Bioprocess Biosyst Eng

28:109–119

31. Prokop A, Prokop Z, Schaffer D, Kozlov E, Wikswo J, Cliffel D,

Baudenbacher F (2004) NanoLiterBioReactor: long-term mam-

malian cell culture at nanofabricated scale. Biomed Microdev

6:325–339

32. Maharbiz MM, Holtz WJ, Howe RT, Keasling JD (2004) Mi-

crobioreactor arrays with parametric control for high-throughput

experimentation. Biotechnol Bioeng 86:485–490

Bioprocess Biosyst Eng (2010) 33:5–19 17

123

33. Korin N, Bransky A, Dinnar U, Levenberg S (2007) A para-

metric study of human fibroblasts culture in a microchannel

bioreactor. Lab Chip 7:611–617

34. Kim L, Vahey MD, Lee HY, Voldman J (2006) Microfluidic

arrays for logarithmically perfused embryonic stem cell culture.

Lab Chip 6:394–406

35. Abbott A (2003) Cell culture: biology’s new dimension. Nature

424:870–872

36. Cushing MC, Anseth KS (2007) Materials science. Hydrogel

cell cultures. Science 316:1133–1134

37. Park JK, Chang HN (2000) Microencapsulation of microbial

cells. Biotechnol Adv 18:303–319

38. Petersen OW, Ronnov-Jessen L, Howlett AR, Bissell MJ (1992)

Interaction with basement membrane serves to rapidly distin-

guish growth and differentiation pattern of normal and malig-

nant human breast epithelial cells. Proc Natl Acad Sci USA

89:9064–9068

39. Liu H, Roy K (2005) Biomimetic three-dimensional cultures

significantly increase hematopoietic differentiation efficacy of

embryonic stem cells. Tissue Eng 11:319–330

40. Elkayam T, Amitay-Shaprut S, Dvir-Ginzberg M, Harel T,

Cohen S (2006) Enhancing the drug metabolism activities of

C3A—a human hepatocyte cell line–by tissue engineering

within alginate scaffolds. Tissue Eng 12:1357–1368

41. Themistocleous GS, Katopodis H, Sourla A, Lembessis P,

Doillon CJ, Soucacos PN, Koutsilieris M (2004) Three-dimen-

sional type I collagen cell culture systems for the study of bone

pathophysiology. In Vivo 18:687–696

42. Smitskamp-Wilms E, Pinedo HM, Veerman G, Ruiz van

Haperen VW, Peters GJ (1998) Postconfluent multilayered cell

line cultures for selective screening of gemcitabine. Eur J

Cancer 34:921–926

43. McGuigan AP, Bruzewicz DA, Glavan A, Butte MJ, Whitesides

GM (2008) Cell encapsulation in sub-mm sized gel modules

using replica molding. PLoS ONE 3:e2258

44. Kim MS, Lee W, Kim YC, Park JK (2008) Microvalve-assisted

patterning platform for measuring cellular dynamics based on

3D cell culture. Biotechnol Bioeng 101:1005–1013

45. Kim MS, Yeon JH, Park JK (2007) A microfluidic platform

for 3-dimensional cell culture and cell-based assays. Biomed

Microdev 9:25–34

46. Koh WG, Pishko MV (2006) Fabrication of cell-containing

hydrogel microstructures inside microfluidic devices that can be

used as cell-based biosensors. Anal Bioanal Chem 385:1389–

1397

47. Torisawa YS, Shiku H, Yasukawa T, Nishizawa M, Matsue T

(2005) Multi-channel 3-D cell culture device integrated on a

silicon chip for anticancer drug sensitivity test. Biomaterials

26:2165–2172

48. Spielberg SP (1980) Acetaminophen toxicity in human lym-

phocytes in vitro. J Pharmacol Exp Ther 213:395–398

49. Tabatabaei AR, Thies RL, Farrell K, Abbott FS (1997) A rapid

in vitro assay for evaluation of metabolism-dependent cytotox-

icity of antiepileptic drugs on isolated human lymphocytes.

Fundam Appl Toxicol 37:181–189

50. Vignati L, Turlizzi E, Monaci S, Grossi P, Kanter R,

Monshouwer M (2005) An in vitro approach to detect metabolite

toxicity due to CYP3A4-dependent bioactivation of xenobiotics.

Toxicology 216:154–167

51. Guengerich FP (2001) Common and uncommon cytochrome

P450 reactions related to metabolism and chemical toxicity.

Chem Res Toxicol 14:611–650

52. Gomez-Lechon MJ, Donato MT, Castell JV, Jover R (2003)

Human hepatocytes as a tool for studying toxicity and drug

metabolism. Curr Drug Metab 4:292–312

53. Lee MY, Kumar RA, Sukumaran SM, Hogg MG, Clark DS,

Dordick JS (2008) Three-dimensional cellular microarray for

high-throughput toxicology assays. Proc Natl Acad Sci USA

105:59–63

54. Ma B, Zhang G, Qin J, Lin B (2009) Characterization of drug

metabolites and cytotoxicity assay simultaneously using an

integrated microfluidic device. Lab Chip 9:232–238

55. Kiang TK, Ensom MH, Chang TK (2005) UDP-glucuronosyl-

transferases and clinical drug–drug interactions. Pharmacol Ther

106:97–132

56. Ma B, Zhou X, Wang G, Dai Z, Qin J, Lin B (2007) A hybrid

microdevice with a thin PDMS membrane on the detection win-

dow for UV absorbance detection. Electrophoresis 28:2474–2477

57. Chang R, Nam J, Sun W (2008) Direct cell writing of 3D

microorgan for in vitro pharmacokinetic model. Tissue Eng Part

C Methods 14:157–166

58. Li AP (2008) In vitro evaluation of human xenobiotic toxicity:

scientific concepts and the novel integrated discrete multiple cell

co-culture (IdMOC) technology. ALTEX 25:43–49

59. Li AP, Bode C, Sakai Y (2004) A novel in vitro system, the

integrated discrete multiple organ cell culture (IdMOC) system,

for the evaluation of human drug toxicity: comparative cyto-

toxicity of tamoxifen towards normal human cells from five

major organs and MCF-7 adenocarcinoma breast cancer cells.

Chem Biol Interact 150:129–136

60. Koebe HG, Deglmann CJ, Metzger R, Hoerrlein S, Schildberg

FW (2000) In vitro toxicology in hepatocyte bioreactors-extra-

cellular acidification rate (EAR) in a target cell line indicates

hepato-activated transformation of substrates. Toxicology

154:31–44

61. Gerlowski LE, Jain RK (1983) Physiologically based pharma-

cokinetic modeling: principles and applications. J Pharm Sci

72:1103–1127

62. Aarons L (2005) Physiologically based pharmacokinetic mod-

elling: a sound mechanistic basis is needed. Br J Clin Pharmacol

60:581–583

63. Materi W, Wishart DS (2007) Computational systems biology in

drug discovery and development: methods and applications.

Drug Discov Today 12:295–303

64. Sweeney LM, Shuler ML, Babish JG, Ghanem A (1995) A cell

culture analogue of rodent physiology: application to naphtha-

lene toxicology. Toxicol In vitro 9:307–316

65. Ghanem A, Shuler ML (2000) Characterization of a perfusion

reactor utilizing mammalian cells on microcarrier beads. Bio-

technol Prog 16:471–479

66. Ghanem A, Shuler ML (2000) Combining cell culture analogue

reactor designs and PBPK models to probe mechanisms of

naphthalene toxicity. Biotechnol Prog 16:334–345

67. Quick DJ, Shuler ML (1999) Use of in vitro data for construc-

tion of a physiologically based pharmacokinetic model for

naphthalene in rats and mice to probe species differences. Bio-

technol Prog 15:540–555

68. Shuler ML, Ghanem A, Quick D, Wong MC, Miller P (1996) A

self-regulating cell culture analog device to mimic animal and

human toxicological responses. Biotechnol Bioeng 52:45–60

69. Sin A, Chin KC, Jamil MF, Kostov Y, Rao G, Shuler ML (2004)

The design and fabrication of three-chamber microscale cell

culture analog devices with integrated dissolved oxygen sensors.

Biotechnol Prog 20:338–345

70. Viravaidya K, Sin A, Shuler ML (2004) Development of a

microscale cell culture analog to probe naphthalene toxicity.

Biotechnol Prog 20:316–323

71. Tatosian DA, Shuler ML (2009) A novel system for evaluation

of drug mixtures for potential efficacy in treating multidrug

resistant cancers. Biotechnol Bioeng 103:187–198

18 Bioprocess Biosyst Eng (2010) 33:5–19

123

72. Sung JH, Shuler ML (2009) A micro cell culture analog

(microCCA) with 3-D hydrogel culture of multiple cell lines to

assess metabolism-dependent cytotoxicity of anti-cancer drugs.

Lab Chip 9:1385–1394

73. Bissett D, Ahmed F, McLeod H, Cassidy J (2000) Oral fluoro-

pyrimidines in the treatment of colorectal cancer. Clin Oncol (R

Coll Radiol) 12:240–245

74. Leucuta SE, Vlase L (2006) Pharmacokinetics and metabolic

drug interactions. Curr Clin Pharmacol 1:5–20

75. Brandon EF, Raap CD, Meijerman I, Beijnen JH, Schellens JH

(2003) An update on in vitro test methods in human hepatic

drug biotransformation research: pros and cons. Toxicol Appl

Pharmacol 189:233–246

76. Gebhardt R, Hengstler JG, Muller D, Glockner R, Buenning P,

Laube B, Schmelzer E, Ullrich M, Utesch D, Hewitt N, Ringel

M, Hilz BR, Bader A, Langsch A, Koose T, Burger HJ, Maas J,

Oesch F (2003) New hepatocyte in vitro systems for drug

metabolism: metabolic capacity and recommendations for

application in basic research and drug development, standard

operation procedures. Drug Metab Rev 35:145–213

77. Nahmias Y, Berthiaume F, Yarmush ML (2007) Integration of

technologies for hepatic tissue engineering. Adv Biochem Eng

Biotechnol 103:309–329

78. Mehta K, Linderman JJ (2006) Model-based analysis and design

of a microchannel reactor for tissue engineering. Biotechnol

Bioeng 94:596–609

79. Roy P, Baskaran H, Tilles AW, Yarmush ML, Toner M (2001)

Analysis of oxygen transport to hepatocytes in a flat-plate

microchannel bioreactor. Ann Biomed Eng 29:947–955

80. Allen JW, Khetani SR, Bhatia SN (2005) In vitro zonation and

toxicity in a hepatocyte bioreactor. Toxicol Sci 84:110–119

81. Camp JP, Capitano AT (2007) Induction of zone-like liver

function gradients in HepG2 cells by varying culture medium

height. Biotechnol Prog 23:1485–1491

82. Leclerc E, Sakai Y, Fujii T (2003) Cell culture in 3-dimensional

microfluidic structure of PDMS (polydimethylsiloxane). Biomed

Microdevices 5:109–114

83. Leclerc E, Sakai Y, Fujii T (2004) Microfluidic PDMS (poly-

dimethylsiloxane) bioreactor for large-scale culture of hepato-

cytes. Biotechnol Prog 20:750–755

84. Leclerc E, Sakai Y, Fujii T (2004) Perfusion culture of fetal

human hepatocytes in microfluidic environments. Biochem Eng

J 20:143–148

85. Bhatia SN, Balis UJ, Yarmush ML, Toner M (1999) Effect of

cell–cell interactions in preservation of cellular phenotype:

cocultivation of hepatocytes and nonparenchymal cells. FASEB

J 13:1883–1900

86. Nakanishi J, Takarada T, Yamaguchi K, Maeda M (2008)

Recent advances in cell micropatterning techniques for bioana-

lytical and biomedical sciences. Anal Sci 24:67–72

87. Khetani SR, Bhatia SN (2008) Microscale culture of human

liver cells for drug development. Nat Biotechnol 26:120–126

88. Mufti NA, Shuler ML (1995) Induction of cytochrome P-450IA1

activity in response to sublethal stresses in microcarrier-attached

Hep G2 cells. Biotechnol Prog 11:659–663

89. White CR, Frangos JA (2007) The shear stress of it all: the cell

membrane and mechanochemical transduction. Philos Trans R

Soc Lond B Biol Sci 362:1459–1467

90. Powers MJ, Domansky K, Kaazempur-Mofrad MR, Kalezi A,

Capitano A, Upadhyaya A, Kurzawski P, Wack KE, Stolz DB,

Kamm R, Griffith LG (2002) A microfabricated array bioreactor

for perfused 3D liver culture. Biotechnol Bioeng 78:257–269

91. Powers MJ, Janigian DM, Wack KE, Baker CS, Beer Stolz D,

Griffith LG (2002) Functional behavior of primary rat liver cells

in a three-dimensional perfused microarray bioreactor. Tissue

Eng 8:499–513

92. Sivaraman A, Leach JK, Townsend S, Iida T, Hogan BJ, Stolz

DB, Fry R, Samson LD, Tannenbaum SR, Griffith LG (2005) A

microscale in vitro physiological model of the liver: predictive

screens for drug metabolism and enzyme induction. Curr Drug

Metab 6:569–591

93. Vozzi F, Heinrich JM, Bader A, Ahluwalia AD (2009) Con-

nected culture of murine hepatocytes and HUVEC in a multi-

compartmental bioreactor. Tissue Eng Part A 15:1291–1299

94. Sung JH, Dhiman A, Shuler ML (2009) A combined pharma-

cokinetic-pharmacodynamic (PK-PD) model for tumor growth

in the rat with UFT administration. J Pharm Sci 98:1885–1904

95. Oh TI, Sung JH, Tatosian DA, Shuler ML, Kim D (2007) Real-

time fluorescence detection of multiple microscale cell culture

analog devices in situ. Cytometry A 71:857–865

96. Tatosian DA, Shuler ML, Kim D (2005) Portable in situ fluo-

rescence cytometry of microscale cell-based assays. Opt Lett

30:1689–1691

97. Novak L, Neuzil P, Pipper J, Zhang Y, Lee S (2007) An inte-

grated fluorescence detection system for lab-on-a-chip applica-

tions. Lab Chip 7:27–29

98. Yu H, Alexander CM, Beebe DJ (2007) A plate reader-

compatible microchannel array for cell biology assays. Lab Chip

7:388–391

99. Sung JH, Choi JR, Kim DH, Shuler ML (2009) Fluorescence

optical detection in situ for real-time monitoring of cytochrome

P450 enzymatic activity of liver cells in multiple microfluidic

devices. Biotechnol Bioeng. doi:10.1002/bit.22413

100. Xing JZ, Zhu L, Gabos S, Xie L (2006) Microelectronic cell

sensor assay for detection of cytotoxicity and prediction of acute

toxicity. Toxicol In Vitro 20:995–1004

101. Natarajan A, Molnar P, Sieverdes K, Jamshidi A, Hickman JJ

(2006) Microelectrode array recordings of cardiac action

potentials as a high throughput method to evaluate pesticide

toxicity. Toxicol In Vitro 20:375–381

102. Asphahani F, Zhang M (2007) Cellular impedance biosensors

for drug screening and toxin detection. Analyst 132:835–841

103. Kang L, Chung BG, Langer R, Khademhosseini A (2008)

Microfluidics for drug discovery and development: from target

selection to product lifecycle management. Drug Discov Today

13:1–13

104. Kim L, Toh YC, Voldman J, Yu H (2007) A practical guide to

microfluidic perfusion culture of adherent mammalian cells. Lab

Chip 7:681–694

105. Sung JH, Shuler ML (2009) Prevention of air bubble formation in

a microfluidic perfusion cell culture system using a microscale

bubble trap. Biomed Microdev. doi:10.1007/s10544-009-9286-8

106. Kang JH, Kim YC, Park JK (2008) Analysis of pressure-driven air

bubble elimination in a microfluidic device. Lab Chip 8:176–178

107. Skelley AM, Voldman J (2008) An active bubble trap and

debubbler for microfluidic systems. Lab Chip 8:1733–1737

108. Melin J, Quake SR (2007) Microfluidic large-scale integration:

the evolution of design rules for biological automation. Annu

Rev Biophys Biomol Struct 36:213–231

109. Horner M, Miller WM, Ottino JM, Papoutsakis ET (1998)

Transport in a grooved perfusion flat-bed bioreactor for cell

therapy applications. Biotechnol Prog 14:689–698

110. Balagadde FK, You L, Hansen CL, Arnold FH, Quake SR

(2005) Long-term monitoring of bacteria undergoing pro-

grammed population control in a microchemostat. Science

309:137–140

Bioprocess Biosyst Eng (2010) 33:5–19 19

123