Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Anal Bioanal Chem (2008) 391:1485–1498

DOI 10.1007/s00216-007-1827-5

REVIEW

The good, the bad, and the tiny: a review


of microflow cytometry
Daniel A. Ateya & Jeffrey S. Erickson &
Peter B. Howell Jr & Lisa R. Hilliard & Joel P. Golden &
Frances S. Ligler

Received: 31 October 2007 / Revised: 17 December 2007 / Accepted: 20 December 2007 / Published online: 29 January 2008
# US Government 2008

Abstract Recent developments in microflow cytometry Introduction


have concentrated on advancing technology in four main
areas: (1) focusing the particles to be analyzed in the In the last 55 years [1], flow cytometry has become a
microfluidic channel, (2) miniaturization of the fluid- standard analytical method in cell biology and medicine. As
handling components, (3) miniaturization of the optics, flow cytometers became commercially available [2], they
and (4) integration and applications development. Strate- began to increase in complexity, employing multiple lasers
gies for focusing particles in a narrow path as they pass and an expanding number of detectors to meet requirements
through the detection region include the use of focusing for multi-color fluorescence analyses. In the last decade,
fluids, nozzles, and dielectrophoresis. Strategies for optics however, advocates of point-of-care and on-site analysis
range from the use of microscope objectives to polymer have created a market for flow cytometers that are simple to
waveguides or optical fibers embedded on-chip. While use, inexpensive, and portable [3]. An intermediate class of
most investigators use off-chip fluidic control, there are a flow cytometers is currently available that are relatively
few examples of integrated valves and pumps. To date, simple to use [4, 5], but these systems are still expensive
demonstrations of applications are primarily used to and at least the size of a typical laser printer. A number of
establish that the microflow systems provide data of the investigators in the microfluidics community have focused
same quality as laboratory systems, but new capabilities— on using microfluidic technologies and incorporating small
such as automated sample staining—are beginning to optical components to create microflow cytometers with
emerge. Each of these four areas is discussed in detail in very small footprints; commercial availability is on the
terms of the progress of development, the continuing horizon.
limitations, and potential future directions for microflow The microfluidics approach to creating a flow cytometer
cytometers. has concentrated on four main areas:
1. focusing the particles to be analyzed in the microfluidic
Keywords Flow cytometry . Microfluidics . Fluid focusing .
channel,
Integrated optics . Cell sorter
2. miniaturization of the fluid-handling components,
3. miniaturization of the optics, and
4. integration and applications development.
Strategies for focusing the particles, as they pass through
Jeffrey S. Erickson, Peter B. Howell Jr and Daniel A. Ateya, provided the interrogation region in front of the laser beams, include
equivalent contributions to this review. the use of focusing fluids, nozzles, and dielectrophoresis.
D. A. Ateya : J. S. Erickson : P. B. Howell Jr : L. R. Hilliard : Strategies for optics range from the use of microscope
J. P. Golden : F. S. Ligler (*) objectives to polymer waveguides or optical fibers embedded
Center for Bio/Molecular Science and Engineering, on-chip. While most investigators use off-chip fluidic control,
Naval Research Laboratory,
Washington, DC 20375, USA there are a few examples of integrated valves and pumps. To
e-mail: frances.ligler@nrl.navy.mil date demonstrations of applications are primarily used to
1486 Anal Bioanal Chem (2008) 391:1485–1498

establish that the microflow systems provide data of the same region of space where the particle is detected. It is the union
quality as laboratory systems, but new capabilities—such as of the core stream, the volume illuminated by the excitation
automated sample staining—are beginning to emerge. Each light and the volume from which the detection optics can
of these four areas will be discussed in detail to give the detect light. Examples of specific techniques that research-
reader an understanding of the progress of development, the ers have utilized to control the interrogation region will be
continuing limitations, and potential future directions for discussed in terms of both fluidics and optics.
microflow cytometers. The convention used in this paper to describe focusing
schemes is somewhat different from that in much of the
literature. Devices that can focus the sample stream from
General theory of the microflow cytometer the sides are often referred to as two-dimensional focusers,
while those that can also focus the sample stream from the
There are several requirements for a microflow cytometer top and bottom are said to be focusing in three dimensions.
to be able to analyze particles effectively (either biological Since focusing from two opposing directions in the same
or synthetic). The first requirement is that each particle axis is localizing the sample stream in only one dimension,
must be isolated in some way so that it can be interrogated the convention used in this paper will be to refer to systems
individually within a detection region. This is most that sheath in the horizontal or vertical dimension (but not
effectively accomplished by focusing a stream of suspended both) as 1-D systems. Those that focus in both the hori-
particles down to a diameter where particles travel in single zontal and vertical dimensions are referred to here as 2-D
file through an interrogation region. By utilizing the systems. If the sample stream fills the channel completely, it
characteristics of laminar fluid flow in microfluidic devices, is referred to here as unfocused.
fluids can be used to ensheath and focus other fluids Sample interrogation is most commonly achieved using
without mixing, and thus to create confined streams within optical detection (via light scatter, fluorescence, or absorp-
a channel. Ideally, the sample stream is sufficiently tion), but electrical impedance also is sometimes used. The
confined by a sheath fluid such that it is extremely rare measurements taken from the particles can be a simple yes/
for two independent particles to pass through the detection no count, but are frequently quantitative. The signal from a
region at the same time. Figure 1 shows a schematic particle is detected as a pulse. The width and geometry of
diagram of the detection region in a common flow- the pulse is a function of the particle velocity and the length
cytometer configuration. The interrogation volume is the of the interrogation region. It is possible to simply quantify
information from the peak maximum directly, but much
higher signal-to-noise ratios are possible if the peak area is
used. This option, however, is not available if the particles
are not traveling at a uniform speed when detected.
The channel architecture and method of pumping liquid
govern the fluid velocity profile through the channel. As
such, particles traveling at different locations in the channel
will have different velocities. The great advantage of flow
focusing is that it forces all of the particles to travel in the
same location in the channel and therefore to have the same
velocity. In addition, focusing typically moves the particles
to the center of the channel, where the flow profile is
relatively flat, so small changes in position do not have a
large effect on the velocity.
However, forcing the sample into a tightly focused
stream can create another challenge. Halving the diameter
of the sample stream requires that the linear velocity be
quadrupled to maintain the same volume throughput. While
microfluidics are frequently endorsed for low volume
requirements, there are times when the lower limit in
sample size is set by the concentration of the target. If a
Fig. 1 Schematic diagram of the detection region in a common flow cytometer is to detect a cell that may only be present at a
cytometer. A particle is detected in the interrogation volume which is
defined by the union of the sample stream, the volume illuminated by
few cells mL−1, sampling only 500 nL will most likely fail
the excitation light, and the volume from which the collection optics to see that cell. One could simply increase the flow
detect light velocity, but as particles pass through the detection region
Anal Bioanal Chem (2008) 391:1485–1498 1487

faster, the sampling electronics must become faster, and the meet the essential cytometer requirements for precise non-
total number of photons available for detection decreases. A pulsatile control of flow rate. In some cases, these relatively
complete solution for these issues related to throughput has cumbersome solutions to integration are acceptable, dem-
not yet been presented. onstrating the ever-present trade-off between size and
function that is often encountered when developing
portable analytical tools.
Fluid handling There are few examples of robust fully integrated on-
chip pumping methods employed in microfluidic devices,
The development of a fully integrated microflow cytometer and fewer used in microflow cytometers due to the
relies, in large part, on precise handling of fluids for additional constraints of these systems. Several microflow
effective sample manipulation and analysis. In order to cytometers have utilized multi-layer soft lithography of
analyze a sample particle by particle, a flow cytometer must polydimethylsiloxane (PDMS) to create pneumatically
introduce a single-file stream of particles into an interroga- actuated valves and peristaltic pumps that produced flow
tion region that usually contains precisely aligned optics for velocities of up to 14 mm s−1 [18–20].
detection. Techniques such as flow focusing and dielec- A significant number of microflow cytometers have
trophoretic focusing are commonly used to orient particles turned to fluid-handling techniques that can be characterized
in this fashion before entry into the interrogation region. as non-mechanical pumping. Electrokinetic techniques [21–
Such particle-focusing techniques play an integral role in 23] utilize strong electric fields to move particles through
almost all cytometers, and this critical feature is evident in the device via electroosmosis and/or electrophoresis. From
microfluidic versions also. the standpoint of focusing, electrokinetic techniques behave
in a way very similar to pressure-driven flow. Models for
Methods for pumping both pressure-driven [24] and electrokinetic focusing [21,
22, 25] are well developed. In some cases, electroosmotic
Stable and well aligned fluid flow requires pumping flow is suppressed by modifying the channel surface charge
methods that avoid the appreciable pulsation that often [26], which means that the particles simply follow the lines
accompanies standard pumps such as peristaltic pumps and of the electric field. This offers some key advantages over
membrane driven pumps [6]. Additionally, the flow many micropumping methods. In particular, fabrication is
velocity profile, resulting from the method of pumping, simple as pumps can be avoided. Electrodes may be
channel architecture, and flow scheme, also governs such embedded in the system by standard microfabrication tech-
factors as the alignment, particle speed, stability of flow, niques, but care must be taken to avoid or control bubble
and particle detection rates. The position and speed of a formation by electrolysis. While pressure-driven flow
particle as it passes through the interrogation region are produces a nominally parabolic profile, electroosmosis
especially critical as they determine the pulse shape, width, produces flow which is much more plug-like. In some
and height of the detected signal. If these parameters are not cases, this partly relieves the increase in variance associated
well defined and reproducible for each particle, the breadth with relatively large sample streams, because all the
of information obtained from data analysis is greatly particles are traveling at the same rate, regardless of their
reduced. position. However, since the particle velocity is now a
Most microflow cytometry studies rely on external function of the surface zeta potential, variations due to
components to control fluid flows in a microchip. This is sample complexities must be accounted for. Also, many
due to the inherent obstacle faced by the field of micro- microfluidic chips are made by bonding together two
fluidics—effective on-chip miniaturization of all the com- separate layers, which may not be the same material. If this
ponents necessary for manipulation of fluids. External is the case, differences in zeta potential between the two
pressure-driven pumps often used in microflow cytometry materials will cause a non-uniform velocity field within the
applications include syringe pumps [7–13] and positively or channel [27], giving rise to the same problem.
negatively pressurized reservoirs using a compressed gas Munyan and coworkers [28] offered an interesting
source or vacuum pump [14]. Passive pumping methods alternative solution to the problem of integrating a micro-
such as gravity-driven flow have also been used [15–17]. fluidic pump for portable applications. The group demon-
Groisman et al. [17] demonstrated the use of gravity-driven strated an electrolytically actuated micropump that offers a
flow to create a high-throughput microfluidic cytometer low-power alternative for continuous flow applications.
with detection rates up to an astounding 17,000 particles This method utilized controlled electrolytic production of
s−1—a rate that approaches the limits of conventional gas in a fluidic reservoir as a pressure source to control
commercial cytometers (1,000–50,000 cells s−1, depending fluid flow rate in a microchannel. This type of pumping
on analysis and accuracy requirements). Such solutions mechanism could be a good candidate for microflow
1488 Anal Bioanal Chem (2008) 391:1485–1498

cytometry applications, as it provides a pulseless, pressure- microcytometry [9, 12, 17, 21, 22, 24, 25, 38, 41–52]. The
driven flow, and reservoirs and electrodes can be integrated sample stream is introduced into the top of the intersection
directly into a microfluidic chip. while sheath streams are introduced from the sides. The
ultimate width of the sample stream depends on the relative
Particle focusing flow rates of the sample and sheath streams and is largely
independent of the geometry of the intersection. Lancaster et
From the theoretical standpoint of individually interrogating al. [12] reported a 1-D focused flow in the vertical direction.
particles, there is actually little need for focusing. It is Using a three-layer input, they created a thin ribbon of
possible to manufacture a sufficiently narrow channel to force sample to be imaged using a microscope.
the particles into a single file. A handful of examples have In most cases that do not use microscopy and video
been reported where focusing was not used [27, 29–37]. imaging, a major difficulty with 1-D focusing is that the
While successful, these were primarily proof-of-concept illumination and collection efficiency are not uniform over
papers aimed at demonstrating an innovation in optical or a broad detection volume. The passage of the particle is
electrical [37] detection and not intended to represent fully registered as a pulse of light by the detector. Whether
functional cytometers. The use of a narrow channel raises detection is by fluorescence or scattered light, the intensity
serious dangers of clogging and surface fouling and is not of the detected pulse is proportional to the intensity of the
really effective if there is a broad range of particle sizes. illuminating light, which typically varies spatially across
The channel must be big enough to accommodate the the beam. If particles are allowed to travel through any part
largest particle or aggregate found in the sample to avoid of the beam, any attempts to obtain quantitative information
clogging, which may mean that the smallest particles from them will be confounded by variations in illumination.
(which may be targets of the study) can pass through the In the extreme case, where the sample stream is greater than
detection region in groups. There are also significant back the beam diameter, some particles may miss the beam
pressure and shear stress associated with driving fluid entirely and not be detected. A similar rule applies to the
through such small channels, which increases instrument collection optics, where the efficiency of collection from
costs and can damage cells. When the sample stream is not different regions of the sample stream may vary.
completely sheathed, particles can come into contact with This problem is not just theoretical. Pamme et al. [45]
the channel wall; fouling can alter the detectable target were able to collect light scatter of 6-μm monodisperse
concentration or interfere with optical or electrochemical latex particles at 15 and 45 degrees, but reported relative
measurements. Channel surfaces have been treated with standard deviations as high as 30%, which they attributed to
compounds such as bovine serum albumin [38, 39], the variation in excitation intensity across the interrogation
hydroxypropylmethylcellulose [16], or covalent coatings region. The broad scatter in their data inhibited their ability
such as polydimethylacrylamide [25], or trychlorohexade- to differentiate between 2 and 9 μm particles. Chung et al.
cylsilane [40] to reduce fouling. [46] demonstrated both 1-D and quasi-2-D focusing. The
quasi-2-D focusing was achieved by making the sheath
One-dimensional flow focusing channel deeper than the sample channel, so that some of the
sheath fluid went over the sample stream as well as on each
Traditional cytometers create sheathed flow by placing a side. This sheath flow then pushed the sample downward
drawn glass capillary or other small tube inside a larger against the bottom of the channel. By comparing the two
one. While highly effective at introducing a sample stream sheathing systems with all detection parameters held
into sheath flow, this kind of structure is extremely difficult constant, they were able to demonstrate reduced variance
to microfabricate. in the fluorescence intensity when the sample stream of
The earliest attempts to approximate sheathed flow used a 10 μm fluorescent beads was more effectively confined.
simple “cross” intersection. Sheath fluid is introduced on Fu et al. [47] experienced 11% variances in signal when
either side of the sample stream, focusing it laterally. The idea particles were passed through a channel focused in 1-D.
of using this style of focusing for cytometry was introduced Red blood cells showed even greater variance, 38%,
by Jacobson and Ramsey [23] using electroosmotic flow, probably due to their nonsymmetrical shape. The authors
although cytometry was not demonstrated at that time. suggested that higher dimension focusing would help solve
Cytometry capability was later demonstrated by Blankenstein the problem.
and Larsen [3], who used pressure-driven flow, and by
Schrum et al.[41], who used electroosmotic flow for the Two-dimensional flow focusing
interrogation of polystyrene beads. Later McClain et al.
demonstrated the same system with bacteria [25], and this By tightly focusing the sample stream in two dimensions,
cross has become the most widely used sheathing system for the particles can be made to travel single file along a single
Anal Bioanal Chem (2008) 391:1485–1498 1489

trajectory, further reducing variance and eliminating the risk sheath fluid into both sides of the channel and focus the
of contaminating the channel surface. Relatively few sample stream in the horizontal direction. The result was a
groups have achieved this in a microfluidic device, and roughly rectangular sample stream located in the center of
the fabrication and plumbing of the chips can be complex. the channel.
Sundararajan et al. [42] used soft lithography to create a The above 2-D designs require at least four and as many
vertical chamber made in five layers with as many as six as six inlets to fully ensheath the sample stream. The
sheath inlets. The final design was able to focus the sample numerous sheath inlets can be viewed as useful where it is
into the middle of the outlet channel. However, this desirable to steer the sample stream within the channel. In
approach is quite complex and only works if the layers most situations, however, it is more important that the
are well aligned. position of the sample stream remains constant, and the
Simonnet and Groisman [17, 49] created two closely requirement to evenly control flow into so many inlets is a
related designs that were able to fully sheath the sample. significant problem.
Their designs used a deep main channel intersected with a Wolff et al. [9, 51, 53] presented a novel design in which
series of shallow side channels with inlets near the bottom the sample stream comes up through a “chimney” to be
of the main channel. In the first design, the sheath stream injected into the middle of the channel. While better than
was introduced into the main channel, and sample stream the previous cross designs, some sample still comes into
was introduced from either side by a pair of shallow contact with the top of the channel. It is likely that a similar
channels so that it filled the lower portion of the deep structure with a higher aspect ratio could fully sheath the
channel. Additional pairs of channels introduced more flow.
sheath fluid to lift the sample stream off the bottom and Yang et al. [52] created a quite complicated structure that
to confine it laterally for complete sheathing. In the second currently comes closest to reproducing the classic annular
design, they added another set of outlets to remove fluid design. The fabrication required three masks, and four
from the region where the greatest diffusion could take exposures. Three exposures were tilted, requiring use of
place. This improved the focusing in the vertical direction. prisms and index-matching liquids. The result was a
In addition, the detection channel was very broad, so that structure with a vertical wall containing a small via at
the sample stream could be focused into a wide ribbon, up mid-depth. It is through this via that sample stream entered
to 700 μm wide, but less than 1 μm deep. A microscope the main channel, and sheath fluid was introduced on either
was used to image particles passing through the channel. side.
In a minor modification to the approach of Simmonet For a well focused stream, the greatest limitation on
and Groisman [49], Chang et al. created 2-D sheathing sensitivity may be the burden placed on data acquisition; a
using just two layers in PDMS [50]. As shown in Fig. 2, sufficient number of signals must be collected from
two separate channel levels were created with contiguous, particles as they pass through the interrogation region at
perpendicular flow channels. Sample and sheath were high speed. One possible solution to this problem was
introduced into the lower portion of the main channel from presented by Bang et al. [54], who demonstrated that after a
perpendicular channels in the lower layer to focus the sample has been focused into a stream with a width of the
sample stream in the vertical direction. Then a more order of the particle diameters, all information about the
standard cross in the upper layer was used to introduce initial distribution of the particles in the channel is lost.

Fig. 2 Schematic illustration of


the hydrodynamic focusing
device developed by Chang
et al. [50]. (a) 3-D view,
(b) top view, (c) side view,
and (d) cross-section perspective
depicting core stream centered
in channel. Reproduced with
permission from the Institute
of Physics Publishing Limited
1490 Anal Bioanal Chem (2008) 391:1485–1498

Once this happens, it becomes possible to widen the DEP to focus them vertically. This provided an excellent
channel again without spreading out the particles. This demonstration of the importance of 2-D focusing. They
slows the particles down and essentially concentrates them were able to turn the vertical focusing on and off and
in a narrow central region of the wider channel. A more observe the effect it had on the amplitude and spread of the
common solution to alleviate the burden of throughput on signal as the particles passed a pair of embedded optical
data acquisition, is integration of analog and digital fibers. The signal obtained using 2-D focusing was both
electronic components; the significance of which will be more intense and more precise for human red blood cells
discussed further in the cytometer electronics section. and for two sizes of latex beads.
Eschewing hydrodynamic focusing altogether, Holmes et
Dielectrophoretic particle focusing al. [56] created a funnel using two pairs of electrodes on the
top and bottom of the channel. In this kind of configuration,
In some circumstances, it is possible to perform focusing the particles were repelled from entering the space between
without a sheath fluid. Dielectrophoresis (DEP) provides the electrodes. They were able to fully focus a stream of latex
another mechanism for focusing particles into a tight beads at their highest potential of 20 V. The slanted surfaces
stream. When in a nonuniform alternating electric field, a of their funnel did not go all the way to the edge of the
particle experiences a force either toward (positive DEP) or channel, and at lower potentials, particles that were outside
away from (negative DEP) the points with the greatest field the sloped region were able to escape focusing. The drag
gradient. The magnitude and direction of the force depends force of the flowing solution was sufficient to overcome the
on a variety of factors, including the slope of the field repulsion of the electrodes when the particles met it head-on.
gradient, the frequency of the electric field, the particle
diameter, and the difference between the polarizabilities of
the particle and the surrounding solution. Optics
All the DEP-based focusing experiments to date have
used negative DEP. Because the strongest field gradients Once core-sheath flow has been achieved in a cytometer,
occur at the surface of the electrode, particles experiencing optical elements provide a method for interrogating,
positive DEP tend to be simply pulled toward the electro- detecting, classifying, and identifying the particles based
des, which can damage cells. Ensuring that negative DEP on both fluorescence and light-scattering criteria. Following
takes place frequently requires that the sample be diluted the trends of commercial instruments, many microflow
with solution of the appropriate ionic composition. cytometers still use traditional bulk optics and typically
As shown in Fig. 3, Lin et al. [55] used a cross-style require a confocal microscope stage for mounting commer-
fluid intersection to focus particles horizontally and then cial high-powered objectives. Lately, there has been a trend
toward the introduction of integrated optical elements
within these devices which is essential for the realization
of a portable or hand-held device [57, 58]. This section will
focus on both commercial and unique solutions to the
problems presented by this transition to integrated optical
components.

Waveguides

In many microcytometer designs, waveguides are used to


precisely deliver excitation light and collect both scattering
and fluorescent signals in a controlled manner. One common
method of incorporating integrated waveguides into micro-
cytometers is to simply add lengths of commercially
available optical fibers into pre-fabricated passively aligned
elements [34, 45, 47, 59]. One end of the fiber is easily
connected to the light source or detector, and the other end
can be inserted directly into the device. Inspired by silicon
optical bench technology in which grooves are etched into
Fig. 3 Schematic diagram of the operating principles for sample
focusing. (a) Top view and (b) side view. Cells or particles are focused
wafers for the passive alignment of parts, PDMS, plastics,
at the center of the sample stream using DEP and hydrodynamic glass, and other substrates can be machined with channels
forces. Reproduced from Lin et al. [55] with permission from IEEE and windows for the passive alignment and support of these
Anal Bioanal Chem (2008) 391:1485–1498 1491

fibers. While efficient at guiding light with little attenuation the waveguide results in 0.01 dB cm−1 attenuation, it may
(losses of the order of 0.2 dB km−1 have been reported), be necessary to shield optically sensitive areas of a
optical fibers can prove cumbersome to integrate and are not microdevice from radiation lost from the sides of rough
well suited to systems incorporating multiple excitation waveguides. Inhomogenieties in refractive index of such
sources, filters, or detectors. materials or scattering centers in the bulk of the waveguide
As an alternative to using commercial fibers, waveguides can make losses even worse. Careful fabrication techniques
can be fabricated directly into microdevices using a variety will be necessary to reduce integrated waveguide attenua-
of optically transparent or reflective materials, or the wave- tion to acceptable levels.
guides can be cast or molded into the device after other Fluidic channels have been used as waveguides in some
fabrication steps are complete. In the latter case, common instances. Waveguiding through air or aqueous solution is
materials for molding include PDMS and UV-curing optical difficult because the core of the waveguide must have a
adhesives, both of which are available with a variety of refractive index higher than the cladding in order to achieve
different refractive indices. For example, Lien and co- total internal reflection. Unfortunately, most cladding
workers demonstrate that it is possible to take advantage of materials used in device fabrication have a refractive index
these commercially available materials to create an all- higher than 1.33 (pure water); only a few materials such as
PDMS device with integrated PDMS waveguides [60]. fluoropolymers have refractive indices this low. Fluoropol-
Although the refractive index differences in commercial ymers are difficult to machine, although they can be used as
materials can be large, very small changes can be produced coatings for liquid-filled waveguide channels [65]. Alter-
for custom waveguide fabrication. Godin and co-workers natively, “leaky mode” waveguides are sometimes used in
report that it is possible to blend commercially available biosensors, in which light is allowed to leak from the core
grades of PDMS to create small refractive index changes due to the absence of total internal reflection. Despite these
[35]. Alternatively, Chang-Yen and co-workers have creat- losses, guided modes still exist, due to partial reflection
ed small refractive index changes in PDMS by curing at between the core and the cladding. In one example, light
elevated temperatures [61]. was launched into a liquid-core leaky waveguide using a
As opposed to molding, photolithography has been used prism coupling [29]. In a second case, anti-resonant Fabry–
to make well defined waveguides out of optically transpar- Perot mirror coatings were used as a replacement for the
ent photoresists such as SU-8; these waveguides are total internal reflection condition. Bernini and co-workers
integrated directly during the fabrication steps of the device deposited controlled thin layers of silicon nitride and silicon
[8, 44]. Alternatively, buried waveguides have been dioxide layers in the underlying substrate [13], creating a
fabricated directly onto silicon wafers in a variety of waveguide that essentially combined the fluidic and optical
materials including silicon dioxide [62], silicon oxynitride paths.
[57], and doped glasses [63]. Although buried waveguides Finally, non-ablative laser machining has been used to
are precisely defined by the micromachining process, they directly write waveguides and pre-pattern microfluidic
tend to be thin in the vertical direction due to the limitations channels in the bulk of monolithic glass and fused silica
of fabrication techniques and may not match up with the substrates. Using femtosecond laser pulses, the refractive
height of the fluidic channel to be interrogated. Waveguides index of glass can be changed by small amounts, of the
fabricated in this manner tend to leave a ridge on the order of 1%. As these structural changes only occur at the
surface of the device where the waveguide is buried, focused part of the beam, buried waveguides can be directly
making final device bonding difficult. In order to alleviate written into these substrates without the need to bond
this problem, further processing such as depositing a thick pieces together. One company, Translume, has commercial-
top layer of glass followed by planarization by chemical- ized this process [66]. Alternatives to use of femtosecond
mechanical polishing is required. Finally, coupling these lasers are specially doped glasses that can be written in the
waveguides to external fibers can be problematic, although absence of femtosecond pulses. In both cases, the laser-
they are well suited to cases where the detectors or sources machining process also changes the etch rates of the glass
are mounted or fabricated directly on the wafer. in HF by a factor of 20 or more, so that microfluidic
Molded and specially microfabricated waveguides have channels can be pre-patterned in the interior of a substrate
many advantages over commercially available optical with a laser, and then later fabricated by wet etching.
fibers. However, these integrated waveguides typically
have an attenuation at least five orders of magnitude greater Optical detectors
than that of optical fibers (of the order of dB cm−1),
necessitating their use in short segments. Most of this loss Depending on the source and intensity, a variety of optical
is probably due to roughness in the sides of the waveguide detectors are commonly used in microflow cytometers:
[64]. Since an average roughness of just 1% of the width of photomultiplier tubes (PMTs), avalanche photodiodes
1492 Anal Bioanal Chem (2008) 391:1485–1498

(APDs), CCD cameras and CMOS imaging arrays, and PIN data-acquisition operations. Custom-designed data-acquisi-
photodiodes. Traditionally, PMTs and APDs have been tion circuitry and signal-processing electronics will certain-
used in commercial cytometers that measure fluorescence ly help alleviate the demand on microprocessors for a
intensity due to their very high sensitivity, including the portable device using this S/N enhancement technique. As
ability to count photons. The internal gain of a detector is an alternative, successive measurements on the same
the most important signal amplification step, as it increases particle with time-of-flight analysis have been demonstrated
the signal with the smallest effect on noise. Therefore, in a PDMS cytometer with integrated PDMS waveguides of
PMTs are typically chosen for very low light applications a higher refractive index [68]. Such a device takes
because they have internal gains of many orders of advantage of the fact that the velocity of particles flowing
magnitude with low noise. However, PMTs are relatively through the core of the cytometer should be constant. An
large. APDs have even more intrinsic sensitivity than 80-dB increase in signal-to-noise ratio, as measured by a
PMTs, but only modest internal gain compared with PMTs CCD camera, was reported for 5-μm fluorescent beads. In
(typically 3–4 orders of magnitude). Photon counting is separate work, this research group integrated an 8×1 optical
possible in Geiger mode, and modest voltage levels are demultiplexing device directly into their cytometer, also
required for implementation as compared with most PMTs. with significant improvement in signal-to-noise ratio [32].
A significant advantage of APDs is their size. PMTs are CCD cameras and CMOS imaging arrays have been
bulky, because of their vacuum tube structure, whereas used for studies of a number of microflow cytometers. They
APDs are solid-state electronic devices. A significant boast reasonable sensitivities and have the advantage of
disadvantage of APDs is their sensitivity to temperature being able to image objects in the flow. However, the
changes. Compensating circuitry must be added, although largest disadvantage of this technology is the speed at
complete compensation and control electronics packages which it can run. Most cytometers using CCD cameras as
are commercially available. The disadvantage of both types detectors can operate at flow rates of less than 100 cells s−1;
of detector is their cost. When more light is available, PIN commercial instruments run significantly faster. Despite
photodiodes can be used for detection and such elements their current availability in bulky packages, it is possible to
are well suited for integration into microdevices. However, integrate these types of imagers directly into the micro-
no internal gain is available, limiting their sensitivity. While device. Nieuwenhuis and co-workers used both a two-
not ideally suited for collecting fluorescence signals, which photodiode strip sensor and an integrated linear array of
can be smaller than 1 nW, they are well suited to collecting photodiodes for imaging the projection of PVC test
light-scattering data. Additionally, PIN photodiodes can be particles as they moved through their cytometer [69].
fabricated with a hole in the center, positioned in such a Hartley and co-workers [70] describe the integration of a
way as to let the excitation beam pass through [67]. This custom-built CMOS chip directly into a flow cytometer
particular geometry makes a beamstop unnecessary in device by both PDMS encapsulation and direct insertion,
forward-scattering applications and can allow the excitation and flip-chip bonding to the rear of their transparent glass
beam to enter the system directly through the detector for device, shown in Fig. 4. Interestingly, the direct incorpo-
backscatter measurements. ration of this CMOS bare die not only eliminated the use of
Any signal from a photodetector can be externally a microscope or waveguide coupling to obtain data, but
amplified. However, the inherent noise in the signal also also included a microprocessor that performed spatial
increases when amplified in this way. Several classical filtering and eliminated the need for external data acquisi-
methods have been implemented to increase the signal-to- tion and analog signal-conditioning electronics.
noise (S/N) ratio from optical detectors in microdevices,
potentially making fluorescence measurements possible Lenses
without expensive photodetectors. One approach is lock-in
amplification, where the excitation source is modulated at a A number of microflow cytometers described in the
known frequency. Because the signals of interest (fluores- literature use bare optical fibers or waveguides without
cence and light-scattering) also cycle with this frequency, additional lensing. Light emerging from such a fiber will be
they can be isolated from the background by use of in the shape of a cone dictated by the numerical aperture
standard electronics. Significant improvements in S/N have (NA) of the fiber, causing an increase in beam size on the
been realized with lock-in amplification. As an example, excitation side and fixed collection efficiency on the
Tung and co-workers [38] developed a microflow cytom- reading side. For devices choosing to operate in such a
eter, with integrated commercially available fibers, that was way, optical fibers with a variety of both core sizes and
capable of performing two-color fluorescence measure- NAs are available, to help control the beam size and
ments utilizing standard PIN photodiodes. The disadvan- collection area. However, lensing is highly desirable,
tage of such a method is the additional burden placed on especially in cases where sensitivity is decreased due to
Anal Bioanal Chem (2008) 391:1485–1498 1493

can only focus light in a single dimension. Second,


depending on the fabrication method, it can be difficult to
control roughness down to subwavelength levels, so that
scattering from these optical elements can be problematic.
Finally, note that any lens designed to be fabricated by
casting an elastomer such as PDMS can shrink on curing,
altering its optical properties from the original design.
In many cases, lensed systems provide a creative method
of obtaining signals that are difficult to obtain directly with
fibers. For example, angle-specific light scattering data can
be difficult to collect with optical fibers, because only one
small part of the angular section can be observed with a
fixed fiber. However, Singh and co-workers demonstrate a
microfluidic device capable of obtaining light-scattering
data over a wide angular range by use of a hemispherical
lens [29]. The authors fabricate a 30-μm channel using
photoresist, and launch light directly into the waveguide
using a prism and the phase-matching condition. The liquid
in the device itself serves as a leaky waveguide through
Fig. 4 A 15 mm2 microfluidic dielectrophoretic cell sorter. The
which the light excites the beads. Scattered light is collected
device combines dual digital cytometer sensors, in-channel electroki-
netic electrodes, and hydrodynamic fluid focusing. Reproduced from through a commercially obtained hemispherical lens and is
Hartley et al. [70] with permission from the IEEE projected directly on to a CCD array. Using this configu-
ration, it is theoretically possible to obtain all light-
the small size of samples (such as for increasing intensity of scattering data from 0–180°, although the authors note that
fluorescence emission). Focusing light down to a small spot in practice such collection would be difficult due to the
can increase fluorescence emission, and the use of lenses inherent brightness of the forward scattering angles com-
can increase the NA of the collection optics. While fibers pared with other angles. The authors demonstrate that by
with lensed ends are commercially available, little variety de-centering the beads from the axis of the lens, they are
currently exists in the types of fiber that can be obtained in able to effectively collect backscattering data over the range
this way. Fiber tip etching can also be used to create lensing 140–180°.
effects, although fibers so modified may be fragile and
difficult to insert into microdevices. Other commercially Excitation sources
available solutions exist. Ball lenses (standard or GRIN)
and cylindrical GRIN lenses do exist that can be added to Light-emitting diodes (LEDs) and laser diodes can both be
microdevices to collimate or focus the beam. However, it is integrated directly into a microflow cytometer or can be
difficult to obtain such lenses on a size scale smaller than used in packaged form in the near vicinity of the fluidic
approximately 200 μm. device. Compared with lasers, LEDs suffer from the
Literature exists for the top-down fabrication, from a disadvantage of non-collimated emission. However, they
variety of materials, of lensing arrays and individual are still used in compact optical devices due to their low
elements at much smaller sizes than are commercially cost. As an example, Novak and co-workers demonstrate
available. PDMS [71], SU-8 [8, 72], fluid-filled [35], and the integration of a blue LED and collimating optics,
polymer lenses [73, 74] have been fabricated for use in photodiode, and amplifier into a compact package for
microfluidic devices. Microlenses can be fabricated for use detection of fluorescence in liquids [75] (Fig. 5).
with integrated in-plane optics, especially for coupling to Other integrated light sources are possible. Balslev and
waveguides. As an example, Camou and co-workers [71] co-workers describe the development of an integrated
describe the fabrication of a PDMS waveguide with a device for optical measurement in fluids that contains a
curved end, acting as a lens to focus light into a fluidic microfluidic optically pumped dye laser, SU-8 waveguides,
channel. Although this technique is excellent for producing and integrated photodiodes [76]. In this work, the dye
lenses with pre-defined focal lengths and fixed optical (rhodamine 6G) was introduced into a 1 mm wide fluidic
alignment, there are two significant disadvantages. The first channel and replaced at a rate of 1–10 μL h−1. The dye was
is that lenses made in this way are inherently cylindrical, pumped with a pulsed external light source. Five SU-
since the top-down machining techniques allow for no 8 waveguides collected light emitted from the laser and
continuous variations in the z-direction. Cylindrical lenses guided it to different locations in the integrated cuvette.
1494 Anal Bioanal Chem (2008) 391:1485–1498

can be accurately obtained. As microflow cytometers are


run at faster and faster speeds, sampling in this way
becomes very demanding, especially in systems that require
continuous operation or data processing in real time. As an
alternative, the integration of nested sample and hold
amplifiers allows the creation of a “peak detector”, which
enables the system to very accurately capture the maximum
value of the peak, at the expense of time resolution. These
types of electronics allow the collection of very accurate
data even when sampling just a few points per peak, greatly
easing the burden on data acquisition operations and at the
Fig. 5 Photograph of a lab-on-a-chip device with integrated micro- same time potentially reducing the coefficient of variation
fluidic dye laser, optical waveguides, microfluidic network, and
on the data collected. In addition, these types of electronics
photodiodes. The metallic contact pads for the photodiodes are seen
on the far right. The chip footprint is 15 mm by 20 mm. The are typically added to the analog side of the system,
photograph, which was taken before a lid was bonded to the reducing the number of (expensive) CPU cycles required
structures, is reproduced from Balslev et al. [75] with permission for system operation on the digital side. Simultaneous peak
from the Royal Society of Chemistry
integration, using analog electronics, can accurately obtain
peak areas with little software processing.
Finally, SU-8 waveguides guided the emitted light from the There are many other possible improvements to the
cuvette directly to integrated photodiodes, through leaky electronics of simple data acquisition systems, including
waveguide mode coupling. filtering, the introduction of thresholding circuits to limit the
number of points that pass through software, and the
Optical filters introduction of logic gates for quick comparisons between
channels. Many other improvements exist. In most cases, the
Very high quality commercial fluorescence filters are commercial availability of electronics parts in integrated
typically composite structures; because of these laminated circuit packaging, coupled with the widespread availability of
layers, they can be quite difficult to machine and are inexpensive microcontrollers and microprocessors, allow the
usually used only in manufacturer-supplied geometries. creation of compact, low-power data collection and analysis
Furthermore, they are expensive and tend not to be systems without the need to send designs to a foundry for
disposable. However, other materials have been suggested microfabrication. We cannot emphasize strongly enough the
that are easily machined or directly incorporated into the impact of a well-designed electronics and data-processing
fabrication of a device. For example, Hofmann and co- package in microcytometer development. However, a thor-
workers demonstrate the fabrication of a disposable long- ough discussion of recent innovations in cytometer electron-
pass optical filter by the addition of organic dyes to PDMS ics and data processing algorithms is beyond the scope of this
[77]. Other potential solutions include microfabrication of manuscript. A good place for an interested reader to start is
interference filters on to silicon-based devices [78, 79], the the excellent review of flow cytometer electronics that has
use of thin-film coatings [80], and the use of colored thin been published by Snow [81].
sheets of polycarbonate.

Cytometer electronics Electrical detection methods

Creative electronics design and data-processing algorithms As an alternative to standard optical methods that utilize
can have a significant effect on both the acquisition and light-scattering information in a flow cytometer, several
analysis speed of a microflow cytometer and the quality of groups have demonstrated the use of electrical detection
the data obtained. As an example, perhaps the most methods to extract a variety of useful information from
important data parameter in both fluorescence and light- these systems. The methods are amenable to portable
scattering measurements is the intensity (maximum) value cytometric applications, eliminating bulky optical compo-
of the peak, followed by the area under the peak. The nents by use of metal electrodes directly embedded in a
typical way to obtain this information in a classical data microfluidic channel. Because cell membranes act as
acquisition system is to sample each peak with many insulators at low measurement frequencies, electrical
points, with the hope that one of those samples will be near detection methods can be extremely useful for cytometry
the true peak intensity, and that there are enough points on applications. In fact, this is the basis for the Coulter counter
the peak to reproduce its shape so that the area underneath which has become a ubiquitous analytical tool in biological
Anal Bioanal Chem (2008) 391:1485–1498 1495

laboratories in the last half century. While specificity is In the first device the flow cytometer consists of a PDMS
limited with these label-free methods, electrical impedance flow channel integrated with pneumatic serpentine-shape
measurements have been used to detect the presence, size, (S-shape) micropumps, embedded optical fibers for multi-
and dielectric properties of particles as they flow through a wavelength fluorescence detection, and a microflow switch-
detection region in a microchannel. In recent years, these ing device, composed of pneumatic micro-valves capable of
methods have been utilized with increasing functionality in automated cell injection, counting, and switching. Experi-
microflow cytometric devices. Impedance spectroscopic mental results showed that the developed flow cytometer
techniques have been used on-chip to demonstrate differ- can distinguish specific cells with different dye-labeling
entiation between erythrocytes in various states, and from a mixture of oocytes (15–20 μm) and lung cancer
polystyrene and latex beads [30, 82]. By interrogating with cells (10 μm) labeled by fluorescent dyes in a single
both low and high frequencies in these studies, cells and process. In the second device the PDMS flow cytometer
particles could be differentiated by both size and electrical chip is integrated with similar microfluidic components for
properties. Chun et al. used polyelectrolytic salt bridge a cell counting/sorting system. In addition, the device
electrodes to maintain DC currents which are ideal for contains an optical detection system and a data analysis and
electrical cell detection. [83] They report detection of 10-μm control system. Fluorescently-labeled human lung cells
beads, at rates of up to 1000 beads s−1, and demonstrate the have been detected and collected (120 cells min−1) using
ability to distinguish between red and white blood cells this portable system that has dimensions of 37 cm×16 cm×
based on the signal amplitude–cell size correlation. 18 cm and weighs 3.5 kg. These two devices are capable of
sorting 120 cells min−1. A microfabricated cell sorter,
integrated with peristaltic pumps, pulse dampeners, switch
Integrated systems and applications valves, and input and output wells has been developed by
Fu et al. [18]. The device was used continuously for six
A number of microflow cytometer devices have been months with millions of actuations of each valve and pump.
described above to illustrate strides made in individual Escherichia coli cells were sorted and recovered at a rate of
areas of fluidics, optics, and detection systems. While most 44 cells s−1. Wolff et al. [9] reported a pressure-driven
reports appearing in the literature have focused on technical microfabricated fluorescence-activated cell sorter with
gains in one of these areas, a few stand out as examples of integrated chambers on the chip for holding and culturing
well-designed packages that make a serious attempt to sorted cells. Using this sorter, fluorescent latex beads were
duplicate the capabilities of benchtop instruments. These sorted from chicken red blood cells, achieving substantial
include cytometers with excellent detection efficiency over enrichments at a sample throughput of 12,000 cells s−1.
multiple channels, cytometers with very high sample Sorted yeast cells collected in the culturing chamber were
throughput, or studies targeted toward specific assays. supplied with a flow of fresh cell media and grew and
Furthermore, a number of commercial microfluidic plat- divided for several days on-chip.
forms are currently available and worthy of note, although
admittedly none is fully integrated. A small selection of Commercial systems
these papers is described below. Other excellent manu-
scripts are not described here [48, 84]. A few commercial systems that use microfluidic components
Given the potential advantages of reduction of sample are currently available. One such system, the Agilent 2100
consumption, assay time, and power consumption, sev- Bioanalyzer [88], is capable of flow cytometric analysis of
eral groups have developed sample-preparation schemes cells, and electrophoretic separation and analysis of DNA,
that may be incorporated into microflow cytometer RNA, and proteins. The Bioanalyzer was used by Chan et al.
systems; complete integration has not yet been demon- [89] for analysis of protein expression and apoptosis in
strated, however. A few recent developments include human primary cells. In the microfluidics chips, the cells are
microfluidic devices for separation and/or manipulation moved by pressure-driven flow through glass-based channels
of blood cells [85, 86]. Other examples that may be and are analyzed by two-channel fluorescence detection.
applicable to flow cytometry, such as automated fluores- Approximately 500–1000 cells can be analyzed in four
cence labeling of samples, have been highlighted in minutes, and up to six cell samples can be handled by one
another review [84]. microchip. Staining reactions can be performed on-chip and
the analysis can be done without washing steps. The reported
Integrated prototype systems results correlated with data obtained using a standard flow
cytometer. The Agilent 2100 Bioanalyzer was also used to
Chang et al. [87] and Yang et al. [19] have presented detect small amounts (10 cfu mL−1) of milk-spoiling bacteria
systems integrating two or more microfluidic components. [90] that were undetectable using the standard culture
1496 Anal Bioanal Chem (2008) 391:1485–1498

method and to analyze fluorescence in-situ hybridization in Using a cytometer based on silicon hollow-core anti-
natural marine pelagic bacterial communities [91]. resonant reflecting optical waveguides, Bernini et al. [13]
Another commercial system has been developed by differentiated between two main populations of Jurkat cells
Micronics that uses a microfluidic laboratory-on-a-card at different stages in the cell cycle, on the basis of
structure. The plastic card, made using a laminate prototyp- fluorescent labeling of DNA; the results showed the same
ing method, consists of a 30-μL sample loop, a 400-μL trend as those produced using a bench-top flow cytometer.
reagent reservoir, a sample injector, labeling loop, view port, A PMMA device has been used to count and size
and a sorting slit for removal of tagged cells or particles. As particles and particle agglomerates on the basis of laser-
discussed earlier, Lancaster et al. [12] demonstrated the light scattering [45]. Light scattering by 2 to 9-μm polymer
detection and sorting of rare cancer cells from blood using beads at two different angles, 15 and 45 degrees, was
the Micronics device. As the sample was injected, a thin detected with a throughput of 150 particles s−1. Size
ribbon monolayer of cells was produced that enabled discrimination of particles with a diameter ratio of 1:2
multiple cells to be viewed and sorted as a whole cell row was also achieved. The device was used to detect C-
or section of the ribbon at a time. The cell injector was also reactive protein (CRP) in a particle-enhanced immunoassay,
capable of antibody labeling within 20 s. Within 30–60 min, with a limit of detection of 100 ng mL−1.
target cells could be detected at sensitivity levels 1,000 to
10,000 times those of existing flow cytometers.
The way forward
Applications
Clearly, there is widespread excitement generated by the
As more flow cytometer components are miniaturized and concept of transforming flow cytometry capabilities into a
integrated with microfluidic devices, more microflow portable, microfluidic format. The ideal microflow cytom-
cytometers will be used for applications in fields including eter will be small, automated, and robust. Components are
gene diagnosis, transfusion medicine, bacteria analysis, being optimized, but systems integration efforts are rela-
clinical hematology diagnosis, DNA molecular sizing, and tively immature.
environmental microbial sensing. A few examples of As the heart of a flow cytometer is the generation of a
applications are presented below. sample stream with a diameter of the order of the particles
In a paper discussed earlier, Simonnet et al. [17], to be measured, it is no surprise that flow focusing has
describe two PDMS microfluidic devices fabricated for received the most attention to date. The “perfect” flow
two microflow cytometers. The first system analyzed up focusing system should be:
to 17,000 particles s−1 and the fluorescence detection
accuracy was comparable with that of a conventional flow 1. easily manufacturable,
cytometer. The second system design focused the sample 2. easily replaceable, and
stream to a flow layer of submicrometer thickness that 3. resistant to clogging or fouling.
enabled imaging of particles with a resolution comparable In other words, the device needs:
with that of still micrographs. To the best of our knowledge,
1. to be as simple as possible, with as few layers as
this is the highest throughput microflow cytometer reported
possible,
to date.
2. to have a minimum number of fluidic and electronic
In addition to the need for high throughput in some
interconnects, and
applications, the viability of sorted and recovered cells is
3. to allow no contact between the channel walls and the
also important. Wang et al. [92] developed a fluorescence-
sample stream wherever the diameter of that stream
activated microfluidic cell sorter that used optical forces for
approaches the diameter of the particles to be analyzed.
the rapid (2–4 ms) and active control of 20–100 cells s−1 on
a microfluidic chip. Transfected HeLa cells, detected and This goal has not yet been achieved, but the scientific
sorted using the device, were shown to be viable and community is approaching it asymptotically.
unstressed following analysis. For reliable data, the position of the sample stream within
Several bacteria studies have been performed by groups the sheath flow must be very well aligned with regard to the
using microflow cytometers. In two other examples using excitation and collection optical paths. Pumps amenable to on-
PDMS channels, the devices were used for counting bacteria chip integration are under intense development for a multitude
in potable water [93] and discrimination of live from dead of microfluidic applications and will become available in the
bacteria [10] with counts of 104–105 cells mL−1 and 1000 near future. On-chip valves are also being widely investigated
cells min−1, respectively. Cell and particle-based assays for other applications and should be available as problems
have also been analyzed in microflow cytometer systems. with the reliability of materials are solved.
Anal Bioanal Chem (2008) 391:1485–1498 1497

The minimal requirement for robust, highly sensitive 4. Dubelaar GBJ, Geerders PJF, Jonker RR (2004) J Environ Monit
6:946–952
optics will be on-chip waveguides and lenses to deliver the
5. Oh KW, Ahn CH (2006) J Micromechanics Microengineering 16:
light and collect the signal from a region of a channel R13–R39
probably less than 20 μm2. For many applications, incorpo- 6. Atencia J, Beebe DJ (2006) Lab Chip 6:567–574
ration of the excitation and collection devices on-chip will be 7. McClain MA, Culbertson CT, Jacobson SC, Allbritton NL, Sims
CE, Ramsey JM (2003) Anal Chem 75:5646–5655
important to construct a continuous monitoring system in a
8. Wang Z, El-Ali J, Engelund M, Gotsaed T, Perch-Nielsen IR,
very small footprint. The microelectronic materials commu- Mogensen KB, Snakenborg D, Kutter JP, Wolff A (2004) Lab
nity has developed the necessary tools to manufacture Chip 4:372–377
integrated optics, and the required components are becoming 9. Wolff A, Perch-Nielsen IR, Larsen UD, Friis P, Goranovic G,
Poulsen CR, Kutter JP, Telleman P (2003) Lab Chip 3:22–27
commercially available. In addition to more traditional laser 10. Inatomi KI, Izuo SI, Lee SS (2006) Lett Appl Microbiol 43:296–
diodes and APDs, polymer components such as OLEDs and 300
organic detectors could be fabricated on-chip. 11. Kruger J, Singh K, O’Neill A, Jackson C, Morrison A, O’Brien P
Applications will drive the incorporation of on-chip (2002) J Micromechanics Microengineering 12:486–494
12. Lancaster C, Kokoris A, Nabavi M, Clemmens J, Maloney P,
sample processing and sorting. For point-of-care and field
Capadanno J, Gerdes J, Battrell CF (2005) Methods 37:120–127
applications, automated sample preparation is essential. In 13. Bernini R, De Nuccio E, Brescia F, Minardo A, Zeni L, Sarro PM,
addition to requiring small pumps and valves under Palumbo R, Scarfi MR (2006) Anal Bioanal Chem 386:1267–
automated control, microfluidic mixers must be incorporated 1272
14. Stiles T, Fallon R, Vestad T, Oakey J, Marr DWM, Squier J,
into the systems—perhaps even on the same chip as the
Jimenez R (2005) Microfluidics and Nanofluidics 1:280–283
analytical device. Sorters that respond to the analytical 15. Sun Y, Yin XF (2006) J Chromatogr A 1117:228–233
information and collect targets of interest will provide 16. Yao B, Luo GA, Feng X, Wang W, Chen LX, Wang YM (2004)
capability that complements the flow cytometry analysis— Lab Chip 4:603–607
17. Simonnet C, Groisman A (2006) Anal Chem 78:5653–5663
saving viable samples for additional analysis such as 18. Fu AY, Chou HP, Spence C, Arnold FH, Quake SR (2002) Anal
confirmatory culture, genetic identification, or mass spec- Chem 74:2451–2457
troscopy. The first sorters will employ electrophoretic, 19. Yang SY, Hsiung SK, Hung YC, Chang CM, Liao TL, Lee GB
electrokinetic, or magnetic-bead-based methods. However, (2006) Meas Sci Technol 17:2001–2009
20. Wang C-H, Lee G-B (2006) J Micromechanics Microengineering
eventually sorting methods based on optical forces may
16:341–348.
prevail because they are less likely to affect fragile cells. 21. Xuan XC, Li DQ (2005) Electrophor 26:3552–3560
More attention must be paid to system integration, 22. Yang RJ, Chang CC, Huang SB, Lee GB (2005) J Micro-
including user-friendly control and data-analysis software. mechanics and Microengineering 15:2141–2148
23. Jacobson SC, Ramsey JM (1997) Anal Chem 69:3212–3217
Analytical software has largely been the purview of the
24. Lee GB, Chang CC, Huang SB, Yang RJ (2006) J Micro-
cytometry industry. As microflow cytometers move out of mechanics and Microengineering 16:1024–1032
research and into commercialization, the software issues will 25. McClain MA, Culbertson CT, Jacobson SC, Ramsey JM (2001)
be addressed. However, developers of new microflow Anal Chem 73:5334–5338
26. Krishnamoorthy S, Feng J, Henry AC, Locascio LE, Hickman JJ,
cytometers should take the time to understand the software
Sundaram S (2006) Microfluidics and Nanofluidics 2:345–355
and electronics of existing commercial systems to anticipate 27. Eyal S, Quake SR (2002) Electrophor 23:2653–2657
the need to mesh the fluidics and optics with the level of data 28. Munyan JW, Fuentes HV, Draper M, Kelly RT, Woolley AT
sophistication required by various users. Furthermore, a (2003) Lab Chip 3:217–220
29. Singh K, Su XT, Liu CG, Capjack C, Rozmus W, Backhouse CJ
design process that gives equal consideration to fluidics,
(2006) Cytometry Part A 69A:307–315
optics, biochemistry, electronics, and the intended application 30. Gawad S, Schild L, Renaud P (2001) Lab Chip 1:76–82
scenario will yield a truly effective microflow cytometer. 31. Fu AY, Spence C, Scherer A, Arnold FH, Quake SR (1999)
Nature Biotechnol 17:1109–1111
32. Lien V, Zhao K, Berdichevsky Y, Lo YH (2005) IEEE J Sel Top
Acknowledgements The preparation of this manuscript was sup-
Quantum Electron 11:827–834
ported by NRL/ONR 6.2 Work Unit 6006. DAA and LRH are
33. Stavis SM, Edel JB, Samiee KT, Craighead HG (2005) Lab Chip
National Research Council Postdoctoral Fellows. The views are those
5:337–343
of the authors and do not reflect policy or opinion of the US Navy or
34. Xiang Q, Xuan XC, Xu B, Li, DQ (2005) Instrum Sci Technol
Department of Defense.
33:597–607
35. Godin J, Lien V, Lo YH (2006) Appl Phys Lett 89
36. Perry H, Greiner C, Georgakoudi I, Cronin-Golomb M, Omenetto
FG (2007) Rev Sci Instrum 78
References
37. Wood DK, Braun GB, Fraikin JL, Swenson LJ, Reich NO,
Cleland AN (2007) Lab Chip 7:469–474
1. Crosland-Taylor PJ (1953) Nature 171:37–38 38. Tung YC, Zhang M, Lin CT, Kurabayashi K, Skerlos SJ (2004)
2. Steinkam J, Fulwyler MJ, Coulter JR, Hiebert RD, Horney JL, Sens Actuators, B, Chem 98:356–367
Mullaney PF (1973) Rev Sci Instrum 44:1301–1310 39. Dittrich PS, Schwille P (2003) Anal Chem 75:5767–5774
3. Blankenstein G, Larsen UD (1998) Biosens Bioelectron 13:427–438 40. Li PCH, Harrison DJ (1997) Anal Chem 69:1564–1568
1498 Anal Bioanal Chem (2008) 391:1485–1498

41. Schrum DP, Culbertson CT, Jacobson SC, Ramsey JM (1999) 67. Kamei T, Paegel BM, Scherer JR, Skelley AM, Street RA,
Anal Chem 71:4173–4177 Mathies RA (2003) Anal Chem 75:5300–5305
42. Sundararajan N, Pio MS, Lee LP, Berlin AA (2004) J Micro- 68. Chen CH, Tsai F, Lien V, Justis N, Lo YH (2007) IEEE Photonics
electromechanical Syst 13:559–567 Technol Lett 19:441–443
43. Blankenstein G, Scampavia LD, Ruzicka J, Christian GD (1996) 69. Nieuwenhuis JH, Bastemeijer J, Bossche A, Vellekoop MJ (2003)
Cytometry 25:200–204 IEEE Sens J 3:646–651
44. Lee GB, Lin CH, Chang GL (2003) Sens Actuators A, Phys 70. Hartley L, Kaler K, Yadid-Pecht O (2007) IEEE Trans Circuits
103:165–170 Syst I-Regular Papers 54:99–110
45. Pamme N, Koyama R, Manz A (2003) Lab Chip 3:187–192 71. Camou S, Fujita H, Fujii T (2003) Lab Chip 3:40–45
46. Chung S, Park SJ, Kim JK, Chung C, Han DC, Chang JK (2003) 72. Kuo JN, Hsieh CC, Yang SY, Lee GB (2007) J Micromechanics
Microsystem Technologies-Micro-and Nanosystems-Information and Microengineering 17:693–699
Storage and Processing Systems 9:525–533 73. Traut S, Rossi M, Herzig HP (2000) J Mod Opt 47:2391–2397
47. Fu LM, Yang RJ, Lin CH, Pan YJ, Lee GB (2004) Anal Chim 74. Nussbaum P, Philipoussis I, Husser A, Herzig HP (1998) Opt Eng
Acta 507:163–169 37:1804–1808
48. Huh D, Gu W, Kamotani Y, Grotberg JB, Takayama S (2005) 75. Novak L, Neuzil P, Pipper J, Zhang Y, Lee S (2007) Lab Chip
Physiol Meas 26:R73–R98 7:27–29
49. Simonnet C, Groisman A (2005) Appl Phys Lett 87 76. Balslev S, Jorgensen AM, Bilenberg B, Mogensen KB,
50. Chang CC, Huang ZX, Yang RJ (2007) J Micromechanics and Snakenborg D, Geschke O, Kutter JP, Kristensen A (2006)
Microengineering 17:1479–1486 Lab Chip 6:213–217
51. Klank H, Goranovic G, Kutter JP, Gjelstrup H, Michelsen J, 77. Hofmann O, Wang X, Cornwell A, Beecher S, Raja A, Bradley
Westergaard CH (2002) J Micromechanics and Microengineering DD, Demello AJ, Demello JC (2006) Lab Chip 6:981–987
12:862–869 78. Burns MA, Johnson BN, Brahmasandra SN, Handique K, Webster
52. Yang R, Feeback DL, Wang WJ (2005) Sens Actuators A, Phys JR, Krishnan M, Sammarco TS, Man PM, Jones D, Heldsinger D,
118:259–267 Mastrangelo CH, Burke DT (1998) Science 282:484–487
53. Goranovic G, Perch-Nielson IR, Larsen UD, Wolff A, Kutter 79. Adams ML, Enzelberger M, Quake S, Scherer A (2003) Sens
JP, Telleman P (2001) In Modeling and Simulation of Actuators A-Phys 104:25–31
Microsystems 80. Chediak JA, Luo ZS, Seo JG, Cheung N, Lee LP, Sands TD
54. Bang HW, Yun HY, Lee WG, Park J, Lee J, Chung S, Cho (2004) Sens Actuators A-Phys 111:1–7
K, Chung C, Han DC, Chang JK (2006) Lab Chip 6:1381– 81. Snow C (2004) Cytom Part A 57A:63–69
1383 82. Cheung K, Gawad S, Renaud P (2005) Cytom Part A 65A:124–
55. Lin CH, Lee GB, Fu LM, Hwey BH (2004) J Microelectrome- 132
chanical Systems 13:923–932 83. Chun HG, Chung TD, Kim HC (2005) Anal Chem 77:2490–2495
56. Holmes D, Morgan H, Green NG (2006) Biosensors Bioelec- 84. Sims CE, Allbritton NL (2007) Lab Chip 7:423–440
tronics 21:1621–1630 85. Chen X, Cui DF, Liu CC, Li H, Chen J (2007) Anal Chim Acta
57. Mogensen KB, Petersen NJ, Hubner J, Kutter JR (2001) Electro- 584:237–243
phor 22:3930–3938 86. Thorslund S, Klett O, Nikolajeff F, Markides K, Bergquist J
58. Verpoorte E (2003) Lab Chip 3:42N–52N (2006) Biomedical Microdevices 8:73–79
59. Wu MH, Wang JB, Taha T, Cui ZF, Urban JPG, Cui Z (2007) 87. Chang CM, Hsiung SK, Lee GB (2007) Jpn J Appl Phys 46:
Biomedical Microdevices 9:167–174 3126–3134
60. Lien V (2006) IEEE Photonics Technol Lett 16:1525–1527 88. Preckel T, Luedke G, Chan SDH, Wang BN, Dubrow R,
61. Chang-Yen DA, Eich RK, Gale BK (2005) J Lightwave Technol Buhlmann C (2002) J Assoc Lab Autom 7:85–89
23:2088 89. Chan SDH, Luedke G, Valer M, Buhlmann C, Preckel T (2003)
62. Friis P, Hoppe K, Leistiko O, Mogensen KB, Hübner J, Kutter JP Cytom Part A 55A:119–125
(2001) Appl Opt 40:6246–6251 90. Yamaguchi N, Ohba H, Nasu M (2006) Lett Appl Microbiol
63. Ruano JM, Benoit V, Aitchison JS, Cooper JM (2000) Anal Chem 43:631–636
72:1093–1097 91. Gerdts G, Luedke G (2006) J Microbiological Methods 64:232–
64. Ladouceur F (1997) J Lightwave Technol 15:1020–1025 240
65. Grewe M, Grosse A, Fouckhardt H (2000) Appl Phys B-Lasers 92. Wang MM, Tu E, Raymond DE, Yang JM, Zhang HC, Hagen N,
and Optics 70:839–847 Dees B, Mercer EM, Forster AH, Kariv I, Marchand PJ, Butler
66. Said AA, Dugan M, Bado P, Bellouard Y, Scott A, Mabesa JJR WF (2005) Nature Biotechnol 23:83–87
(2004) in Photon Processing in Microelectronics and Photonics III 93. Sakamoto C, Yamaguchi N, Yamada M, Nagase H, Seki M, Nasu
5339:194–204, SPIE, San Jose, Ca, USA M (2007) J Microbiological Methods 68:643–647

You might also like