+ All Categories
Home > Documents > 7 - Integrated Microsystems for Controlled Drug Delivery[1]

7 - Integrated Microsystems for Controlled Drug Delivery[1]

Date post: 10-Apr-2018
Category:
Upload: dovnadler
View: 214 times
Download: 0 times
Share this document with a friend

of 14

Transcript
  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    1/14

    Integrated microsystems for controlled drug delivery

    S. Zafar Razzackia,1, Prasanna K. Thwara,1, Ming Yanga,1,Victor M. Ugazb,1, Mark A. Burnsa,c,*

    aDepartment of Chemical Engineering, University of Michigan, 2300 Hayward, 3074 HH Dow Building, Ann Arbor, MI 48109-2136, USAbDepartment of Chemical Engineering, Texas A&M University, USA

    cDepartment of Biomedical Engineering, University of Michigan, 2300 Hayward, 3074 HH Dow Building, Ann Arbor, MI 48109-2136, USA

    Received 26 January 2003; accepted 20 August 2003

    Abstract

    Efficient drug delivery and administration are needed to realize the full potential of molecular therapeutics. Integrated

    microsystems that incorporate extremely fast sensory and actuation capabilities can fulfill this need for efficient drug delivery

    tools. Photolithographic technologies borrowed from the semiconductor industry enable mass production of such microsystems.

    Rapid prototyping allows for the quick development of customized devices that would accommodate for diverse therapeutic

    requirements. This paper reviews the capabilities of existing microfabrication and their applications in controlled drug delivery

    microsystems. The next generation of drug delivery systemsfully integrated and self-regulatingwould not only improve

    drug administration, but also revolutionize the health-care industry.

    D 2003 Elsevier B.V. All rights reserved.

    Keywords: Controlled drug delivery; Integrated microsystems; Microfabrication; Microfluidic; Sensing; Actuation

    Contents

    1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 186

    2. Challenges in drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 186

    3. Photolithography and microfabrication . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 187

    3.1. Silicon processing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 189

    3.2. Soft lithography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 189

    4. Microfabricated drug delivery systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190

    4.1. Biocapsules and microparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190

    4.2. Microneedles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191

    4.3. Implantable microsystems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192

    5. Smart integrated microsystems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194

    0169-409X/$ - see front matterD 2003 Elsevier B.V. All rights reserved.doi:10.1016/j.addr.2003.08.012

    * Corresponding author. Department of Chemical Engineering, University of Michigan, 2300 Hayward, 3074 HH Dow Building, Ann

    Arbor, MI 48109-2136, USA. Tel.: +1-734-763-3078; fax: +1-734-763-459.

    E-mail address: [email protected] (M.A. Burns).1 These authors contributed equally to this work.

    www.elsevier.com/locate/addr

    Advanced Drug Delivery Reviews 56 (2004) 185198

  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    2/14

    6. Commercial applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195

    7. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 196

    References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 197

    1. Introduction

    Microfabricated systems have revolutionized biol-

    ogy and medicine with numerous enabling technolo-

    gies. The plethora of biomedical microelectromechan-

    ical systems (bioMEMS) emerging today range from

    fascinating molecular motors that swim inside a cell

    utilizing the intracellular energy-rich ATP molecules

    [1] to sophisticated point-of-care diagnostic devices

    capable of foretelling ones predisposition to diseases

    by reading the information encoded in a small segment

    of DNA [2]; from devices that manipulate single

    molecules to systems that can analyze the entire

    cytosolic contents of a single cell; from extremely

    sensitive sensors that can track down even attomolar

    traces of a biochemical substance [3] to highly inte-

    grated microfluidic devices that can perform multiple

    serial processing on nanoliter volume samples in an

    automated fashion [4]. With the advent of sub-micron

    lithography techniques in the microelectronics indus-

    try, these bioMEMS devices will improve in their

    versatility, efficiency and user-friendliness. One ofthe areas where bioMEMS devices are poised to offer

    a host of benefits is therapeutics. In this paper, we

    review the current generation of bioMEMS devices

    used in drug delivery applications and offer some

    directions toward which future research might be

    steered.

    2. Challenges in drug delivery

    Modern therapeutic techniques are based onrational design and highly targeted delivery of spe-

    cific drug compounds. While this has been proven to

    be far more superior and effective than the traditional

    therapeutic methods employing non-specific drug

    cocktails, a more meticulous effort is required in

    drug design, toxicological testing and selection of

    appropriate delivery vehicles. Advances in micro-

    fabrication and bioinformatics have helped to greatly

    accelerate the drug discovery process in recent years

    by streamlining the process of identifying molecular

    targets and their agonists/antagonists. Analysis of

    genomic and mRNA data for over or under expressed

    genes can provide clues that would help to identify

    the biological molecules associated with a particular

    disease. Combinatorial approaches coupled with high

    throughput screening tools like peptide and protein

    microarrays have led to a huge surge of data to aid in

    target identification. Once a molecular target is iden-

    tified, conventional structural proteomics tools in-

    cluding capillary electrophoresis, mass spectrometer

    and NMR can be used to elucidate the complex

    secondary and tertiary structures associated with the

    target proteins. This structural information of the

    targets can then be used to browse through the

    database of known drug molecules or could be

    coupled with synthetic biochemistry methods to iden-

    tify potential therapeutic compounds. These steps,

    though not time-consuming in modern lab settings

    require huge capital investments, infrastructure and

    highly trained personnel.

    These drug discovery efforts are generally focused

    on identifying a specific drugtarget pair and rarelyinclude the toxicological information in the search

    process. In most cases, a sufficient amount of toxico-

    logical data has not yet been collected for newly

    synthesized drug compounds, making it difficult to

    select an appropriate delivery vehicle. For example,

    cytotoxic drugs like gemcitabine, rubitecan, daunoru-

    bicin, and doxorubucin, currently used to treat diseases

    like cancer and HIV are very efficient in destroying the

    invading virus and cancerous cells, but also have been

    found to induce apoptosis in adjacent normal cells.

    Even drugs with lower levels of cytotoxicity oftenmust still operate within a narrow therapeutic window

    in order to maintain bioavailability between a mini-

    mum threshold functional concentration and the criti-

    cal toxic concentration above which irreversible

    damage to functional organs could occur(Fig. 1). At

    the molecular level, drug molecules often possess non-

    specific affinity towards receptors on the cell surface

    other than the targeted proteins, which can lead to

    undesirable side effects as in the case of tranquilizers

    and histamine blockers. Moreover, the optimal con-

    S. Zafar Razzacki et al. / Advanced Drug Delivery Reviews 56 (2004) 185198186

    http:///reader/full/page3http:///reader/full/page13http:///reader/full/page13http:///reader/full/page13http:///reader/full/page13
  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    3/14

    centration necessary to induce a desired therapeuticresponse to a particular drug may vary between the

    individuals making the issue even more problematic.

    Stability and bioavailability are also important

    considerations in the design of drug delivery systems,

    especially for protein drugs. Such drugs suffer from a

    number of limitations including degradation by pro-

    teolytic enzymes, immune response and lack of flex-

    ibility for structural modifications (e.g., to tune

    absorption properties).

    Recently, there has been a great interest in gene/

    DNA therapy for treating several hereditary disorders.However, like proteins, naked DNA is extremely

    unstable and consequently must be quickly integrated

    into the host genome in order to be effective. Viral

    vectors have proven to be one of the most successful

    methods of delivering DNA to host cells, although

    many safety concerns still need to be addressed.

    Various modes of administration including oral,

    intravenous, transdermal and pulmonary pathways

    can be used to ensure sufficient bioavailability. How-

    ever, many drugs also require a more controlled and

    sustained release profile. A common example for this

    scenario is the insulin supplement given as intrave-

    nous injections to patients suffering from Type I

    diabetes. Ideally, the administered insulin supplementshould closely mimic the insulin release profile of the

    pancreatic islets. Multi-dose schemes and constant

    infusion pumps have been employed to achieve this

    goal, but have had only limited success in attaining a

    highly controlled level release. Another application

    requiring a non-linear drug release profile is tissue

    regeneration in victims of acute organ failure or partial

    organ loss, where growth factors must be precisely

    administered in order to promote growth and prolif-

    eration of the native tissue and, if necessary, facilitate

    differentiation of the cells into the appropriate cell

    type in a time-dependent fashion.

    All these constraints necessitate drug delivery

    methods with high specificity that ensure proper

    bioavailability of functional drugs at the target site

    with the desired rate and dosage of release. Recent

    technological advances are beginning to enable the

    development of devices that are powerful enough to

    deliver extremely potent drugs in a precisely con-

    trolled manner, intelligent enough to respond to the

    behavior of the surrounding physiological environ-

    ment, and compact enough to fit inside a blood vessel.

    Such devices involve complex micron-scale networksof fluidic and electronic components capable of oper-

    ating in an integrated manner. This new generation of

    novel drug delivery devices owes much of its devel-

    opment to advances in microfabrication and micro-

    machining technology for the construction of micro-

    fluidic systems.

    3. Photolithography and microfabrication

    Microfluidic devices are generally produced usingmicromachining techniques adapted from standard

    semiconductor processing technology. These techni-

    ques rely on photolithography as the principle means

    of transferring finely detailed features to the surface of

    the desired substrate. The photolithographic process

    involves applying a photosensitive coating to the

    substrate surface, exposing the coated substrate to light

    through a mask containing an image of the pattern to

    be transferred, and immersing the substrate in a devel-

    oper solution, which selectively removes photoresist

    Fig. 1. The therapeutic window for a particular drug is defined by

    the upper toxic limit and the lower threshold limit[5]. The position

    and the width of this window can vary between patients for different

    drugs and pose challenges on controlled delivery. The figure depicts

    the four cases of the therapeutic window. (A) The concentration

    profile of the drug is optimal when the fluctuations of the drug

    concentration associated with a dosage scheme are contained within

    the therapeutic window. (B) The therapeutic window can be shifted

    upwards due to high resistance in the body of patient or antagonistic

    interactions with another drug. (C) The therapeutic window can also

    be shifted downwards due to hypersensitivity of the patient or

    synergistic interactions with another drug. (D) It can also be narrow

    for a same drug in a child as compared to that of an adult. The drug

    delivery and dosage schemes have to be adjusted for these sub-

    optimal situations.

    S. Zafar Razzacki et al. / Advanced Drug Delivery Reviews 56 (2004) 185198 187

    http:///reader/full/page13
  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    4/14

    depending on whether or not it has been exposed to

    light. This process yields a high-fidelity replica of the

    mask image on the substrate surface (Fig. 2). Once a

    pattern has been transferred to the substrate, subse-quent micromachining processes are performed to

    either selectively deposit or remove material in the

    patterned regions.

    While many of these processes are borrowed

    directly from existing semiconductor fabrication tech-

    nology, microfluidic systems present a number of

    unique fabrication challenges of their own. For ex-

    ample, the size scale of structures to be machined is

    on the order of tens of microns, whereas those in

    semiconductors are typically micron scale or smaller.

    Deposition of the thick material layers necessary to

    construct these larger structures introduces difficulties

    in maintaining surface uniformity across the device

    and/or wafer, while removal of large amounts ofmaterial requires the use of etching processes that

    allow adequate control to be exerted over such

    characteristics as surface roughness and channel side-

    wall profiles. Microfabricated devices also require

    fluidic circuitry (pumps, valves, etc.) to precisely

    meter and control the motion of liquids within a

    microchannel network, as well as an effective inter-

    face with the external environment[6]. Successive

    micromachining process can also be applied to pro-

    duce devices incorporating complex multilayered

    Fig. 2. Illustration of some typical micromachining processes. (A) Photolithography is first used to transfer a pattern to the surface of a substrate

    coated with photosensitive resist. Next, the development process selectively removes photoresist depending on whether or not it has been

    exposed to light. Subsequent micromachining steps consist of either depositing or removing material in the patterned areas, after which the

    remaining photoresist is stripped away. (B) Polymer-based devices are generally produced using a casting or soft lithography process, in which

    a master incorporating the negative image of the desired structures is first produced using the micromachining techniques outlined in (A). This

    master then serves as a mold or stamp that is subsequently used (and re-used) to produce molded replicas of the initial design.

    S. Zafar Razzacki et al. / Advanced Drug Delivery Reviews 56 (2004) 185198188

    http:///reader/full/page13http:///reader/full/page4
  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    5/14

    structures. While the field of microfabrication for

    microfluidic applications is relatively new, a sufficient

    body of literature has been accumulated to stimulate

    the publication of several recent reviews and texts[712].

    3.1. Silicon processing

    Novel fabrication technologies continue to evolve

    at a rapid pace, and a comprehensive review of every

    existing application is beyond the scope of this paper.

    Instead, we focus on two predominant approaches

    currently employed, in one form or another, in the

    production of the majority of microfluidic devices.

    The first approach closely mirrors semiconductor

    fabrication by making use of conventional silicon

    and glass substrates. In these devices, fluidic channels

    are fashioned by etching directly into the substrate

    either by immersion in a chemical solution (wet

    etching) or by exposure to a chemically reactive

    plasma (dry etching). Here, the mask layer serves a

    dual purpose of defining the pattern to be etched and

    protecting the substrate from degradation in areas that

    are not to be etched. Dry etching processes are

    generally capable of producing deeper features with

    greater sidewall uniformity, however specialized

    equipment is required. Hybrid glass/glass and sili-con/glass devices, in which etched glass fluidic chan-

    nels are bonded to a flat glass or silicon substrate,

    have been widely used in a variety of microfluidic

    applications [1315]. Because arrays of electronic

    components (electrodes, heaters, temperature sensors,

    photodetectors, etc.) can be easily patterned on a

    silicon surface, glass/silicon devices are especially

    attractive in applications requiring integrated fluidic

    and electronic circuitry [4,1618].

    3.2. Soft lithography

    A second fabrication approach involves the use of

    molding or stamping techniques to construct fluidic

    channels in polymer substrates. The wide appeal of

    polymer-based devices is largely a consequence of the

    ease with which fluidic components can be con-

    structed with minimal use of specialized equipment.

    A key consideration is identifying polymer materials

    with properties suitable for biochemical applications

    such as biocompatibility, electric neutrality, and opti-

    cal transparency at desired wavelengths (see, for

    example, Ref. [19] for a recent review). While a

    number of materials have been successfully used to

    construct microfluidic devices, the silicone elastomerpoly(dimethyl siloxane) (PDMS) has emerged as one

    of the most widely used [20,21].

    Fabrication of polymer-based devices typically

    begins with the construction of a master that con-

    tains a negative image of the desired structures and

    will ultimately serve as a template for the production

    of the final microfluidic devices. Masters are usually

    constructed from silicon or thick photoresist, and are

    produced using conventional silicon-based processing

    techniques. Once the master has been constructed, it

    can then be used to produce devices by employing

    either a casting process (often referred to as soft

    lithography) whereby a mixture of silicone resin and

    crosslinker is poured over the master and allowed to

    cure, or a stamping process (hot embossing) whereby

    the master is pressed into a film of polymer that has

    been softened by heating to just below its melting

    point. Aside from fabrication of the master, this

    process does not need to be carried out in a clean-

    room environment. Polymer devices have the advan-

    tage of being simple and relatively inexpensive to

    construct since a single master can be used repeat-

    edly to produce many devices. Additionally, theinherent mechanical flexibility of PDMS structures

    can be harnessed to construct pumps and valves that

    operate by inducing deformations in the substrate

    itself[21].

    Despite their reduced cost, polymer-based devices

    offer only a limited ability to embed electronic

    circuitry. Consequently, silicon-based devices remain

    an attractive and viable alternative because of the

    ability to integrate electronic components directly

    within a microfluidic system. For example, this

    approach has been used to construct a device capableof performing DNA amplification followed by anal-

    ysis of the reaction products by gel electrophoresis

    using embedded photodetection circuitry to image

    the migrating bands [16]. Furthermore, much of the

    added cost and complexity associated with silicon-

    based devices can be offset by the economics of

    scale associated with miniaturization. Using photo-

    lithographic techniques, fabrication costs remain es-

    sentially constant regardless of the number of

    devices produced on a single wafer. Consequently,

    S. Zafar Razzacki et al. / Advanced Drug Delivery Reviews 56 (2004) 185198 189

    http:///reader/full/page13http:///reader/full/page13http:///reader/full/page13http:///reader/full/page13http:///reader/full/page13http:///reader/full/page13http:///reader/full/page13
  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    6/14

    in a fashion completely analogous to developments

    in the semiconductor industry, sufficient miniaturiza-

    tion could ultimately yield biological analysis devi-

    ces small enough to be portable, yet inexpensiveenough to be disposable [22].

    4. Microfabricated drug delivery systems

    Currently, numerous sophisticated and potent new

    drugs are being developed. However, conventional

    drug delivery methods have limitations in effectively

    delivering these drugs into the human body. For

    example, oral delivery of new protein-based or

    DNA-based drugs is generally not feasible because

    of the degradation of the drugs in the gastrointestinal

    tract and first-pass elimination in the liver. The usual

    alternative of oral delivery, intravenous or intramus-

    cular injection, can deliver these drugs in large

    quantities, but pain and tissue trauma to the patients

    often result from the use of needles. Moreover,

    conventional injection is unable to provide sustained

    release, which greatly reduces the effectiveness of the

    drugs.

    Many approaches have been proposed for effec-

    tively delivering drugs into the human body. These

    new drug delivery methods can be categorized intothree major groups: (a) biocapsules and microparticles

    for controlled and/or site-specific drug release; (b)

    Microneedles for transdermal and intravenous deliv-

    ery; and (c) implantable microsystems. Microelectri-

    cal mechanical systems (MEMS), especially Bio-

    MEMS, have found numerous applications in all

    three areas.

    4.1. Biocapsules and microparticles

    Biocapsules and microparticles have been inten-sively investigated as a new drug delivery method. By

    encapsulating drugs in a biodegradable microsphere

    or microparticle, a sustained or controlled drug release

    profile can be achieved. Site-specific delivery can be

    achieved by binding ligands to the surface of micro-

    particles in order to target specific sites. So far, much

    of the research conducted in this area has been

    focused on seeking biodegradable and biocompatible

    polymer materials to make the microspheres or micro-

    particles [2325]. Microfabrication technology has

    recently been used in fabricating nanoporous mem-

    branes for drug encapsulation, and microparticles for

    site-specific oral delivery.

    Nanoporous membranes are produced using micro-fabrication technology, such as photolithography, thin

    film depositions and selective etching, to create mem-

    branes composed of silicon with highly uniform

    nanometer-sized pores. Microfabricated membranes

    provide several advantages over polymer materials:

    smaller pore size (10100 nm range), high uniformity

    and thinner membrane thickness. Moreover, because

    microfabricated membranes are made from silicon,

    they are biologically, chemically and mechanically

    stable. Desai [26] reported microfabricated biocap-

    sules with nanoporous membranes for effective immu-

    noisolation of transplanted islet cells for treatment of

    diabetes (Fig. 3). Surface and bulk micromachining

    technologies are integrated in the biocapsule fabrica-

    tion process, resulting in a diffusion membrane with

    uniform pore size distribution as well as mechanical

    and chemical stability, surrounded by an anisotropi-

    cally etched silicon wafer that serves as the encapsu-

    lation cavity. Insulinoma cells (4500 cells/biocapsule)

    were enclosed within these microfabricated biocap-

    sules and released through the semipermeable nano-

    porous membranes. This research demonstrates the

    feasibility of using microfabricated biocapsules as analternative to conventional polymeric based biocap-

    sules for possible use as in vivo insulin secreting

    bioreactor[27].

    Microfabrication technology has also been used to

    create microparticles for site-specific delivery [26].

    Unlike conventional particulate drug delivery sys-

    tems such as polymer microspheres and liposomes,

    microfabricated microparticles are thin, disc-shaped

    structures. These particles can be designed with a

    thickness of 150 Am and diameters of 1 to 100s of

    microns. Particles can be asymmetrically designedwith single or multiple drug reservoirs, and ligands

    bound to one side can target specific sites. Micro-

    fabricated particles can combine enteric coating,

    bioadhesive agents, permeation enhancers and en-

    zyme inhibitors into a single drug delivery platform.

    The size and shape of the particles can be easily

    controlled to obtain the optimal delivery profile.

    These devices are especially effective for oral deliv-

    ery of peptide and protein-based drugs through the

    intestines.

    S. Zafar Razzacki et al. / Advanced Drug Delivery Reviews 56 (2004) 185198190

    http:///reader/full/page13http:///reader/full/page13http:///reader/full/page7http:///reader/full/page13http:///reader/full/page13http:///reader/full/page13
  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    7/14

    4.2. Microneedles

    Transdermal (across skin) delivery of drugs over

    extended periods of time is a convenient, controlled

    way to administer medication. Compared to conven-

    tional oral delivery and injection, transdermal drug

    delivery overcomes the limitation of gastrointestinaldrug degradation and eliminates the pain and incon-

    venience of intravenous and intramuscular injection.

    However, the efficiency of transdermal drug delivery

    is greatly limited by the low permeability of the

    human skin particularly the outer 1020 Am of skin

    (stratum corneum layer). A variety of approaches have

    been taken to increase the rate of transport across the

    skin using, for instance, chemical enhancers, ionto-

    phoresis, electroporation, or ultrasound [28].

    A novel approach to transdermal drug delivery

    uses microfabricated microneedle arrays that are long

    and robust enough to penetrate the stratum corneum

    but short enough not to stimulate nerves in deeper

    tissues [29]. Arrays of bulk micromachined solidsilicon microneedles were fabricated by a single-mask

    reactive ion etching process and shown to increase

    skin permeability for a variety of different molecules

    by orders of magnitude [28,30] (Fig. 4). Arrays of

    hollow silicon microneedles were also fabricated by

    using deep reactive ion etching to form the needle

    lumen, followed by a reactive ion etching process to

    Fig. 4. Microfabricated solid silicon microneedle arrays. Left: A section of a 20 20 array of microneedles. Right: Microneedle tips inserted

    across epidermis (n 1998 IEEE) [28].

    Fig. 3. A microfabricated biocapsule for in vivo insulin secretion (reprinted with permission [26]).

    S. Zafar Razzacki et al. / Advanced Drug Delivery Reviews 56 (2004) 185198 191

    http:///reader/full/page13http:///reader/full/page13http:///reader/full/page7http:///reader/full/page13http:///reader/full/page13http:///reader/full/page13
  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    8/14

    form the tapered wall of the microneedles. These

    hollow microneedles can increase the skin permeabil-

    ity even further than the solid microneedles [29].

    Microneedles can also be integrated with micro-sensors or microactuators to form sophisticated drug

    delivery systems. Zahn et al. [31] recently reported a

    portable drug delivery device with integrated micro-

    needles and an on-chip MEMS positive displacement

    micropump for continuous drug delivery applications.

    Microneedles are fabricated using a polysilicon micro-

    molding method. The generation and collapse of ther-

    mally generated bubbles with flow rectified by direc-

    tional check valves are used to achieve net pumping.

    Water flow rates of approximately 1.0 nl/s were

    obtained and continuous pumping for more than 6

    h was achieved.

    Another example is a multichannel silicon probe

    [32] to deliver very small and precise amounts of bio-

    active compounds into highly localized areas of neural

    tissue while simultaneously recording electrical signals

    from neurons and electrically stimulating neurons in

    vivo (Fig. 5). The multichannel neural probes are bulk

    silicon micromachined, using a series of oxidation,

    boron diffusion, and wet-etching steps. Integrated com-

    plementary metaloxidesemiconductor (CMOS) cir-

    cuitry and electrodes are also fabricated on the probe

    for neuron stimulation and recording. A more sophis-ticated drug delivery device containing integrated

    microchannels, fluidic cables, dielectric shutters over

    the injecting orifices, and in-line flowmeters to verify

    the intended dose, along with the electrical recording

    and stimulating circuitry, can achieve chronic in vivo

    drug delivery to the neural cells [33].

    4.3. Implantable microsystems

    Implantable devices are preferred for therapies that

    require many injections daily or weekly. They can be

    either implanted into the human body or placed under

    the skin, consequently reducing the risk of infection by

    eliminating the need for frequent injections. Most

    implantable microsystems do not cause pain or tissue

    trauma due to their small size and are often virtually

    invisible [34]. In addition to a reduction in the number

    of injections, implantable drug delivery systems have

    the advantage that the dose level can be precisely

    regulated according to the therapy and the particular

    patients requirements due to the interactive and con-

    trollable nature of these devices. MEMS systems

    combine small size, low power requirements, and the

    potential to precisely meter fluid samples, all of which

    facilitate implantation [35]. For these reasons, MEMS-

    based microsystems are attractive candidates as im-

    plantable drug delivery systems.

    One method for achieving complex drug release

    patterns involves using microfabrication technology to

    develop active implantable microfluidic devices that

    incorporate micropumps, valves and flow channels todeliver liquid solutions. One of the key components in

    those devices is the miniature fluid-dispensing system

    or micropump. Various pumping methods are avail-

    able, including electroosmotic pumping for ionic flu-

    Fig. 5. Micromachined neural tissue drug delivery probe. Left: Sketch of a probe having three delivery channels along with recording and

    stimulating electrodes. Right: SEM view of a probe with three delivery channels (n 1997 IEEE) [32].

    S. Zafar Razzacki et al. / Advanced Drug Delivery Reviews 56 (2004) 185198192

    http:///reader/full/page14http:///reader/full/page14http:///reader/full/page14http:///reader/full/page14http:///reader/full/page8http:///reader/full/page14http:///reader/full/page14http:///reader/full/page13
  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    9/14

    ids, positive displacement pumps that use piezoelectric

    components, and pneumatic, bubble, or surface-ten-

    sion pumps that rely on moving gasliquid interfaces

    to displace fluids [36].

    One example of such a micropump is a silicon

    piezoelectric pump for delivering low flow rates of

    liquid drugs with high precision [37] (Fig. 6). The

    pump was developed for insulin infusion in diabetic

    patients and is based on silicon bulk micromachining,

    silicon pyrex anodic bonding and piezoelectric actu-

    ation. The flow rate is linear with actuation frequency

    and virtually insensitive to inlet and outlet pressure,

    actuation voltage, temperature, viscosity and aging.

    More recently, the same group reported an improved

    micropump design with higher pumping linearity and

    accuracy [38]. A micromachined pressure compensat-

    ing flow regulator was also developed to provide a

    constant liquid flow rate within a pressure range of

    100600 mbar and can possibly be used to replace the

    flow restrictor in an elastomeric infusion system [39].

    Implantable drug delivery devices other than active

    microfluidic devices have also been investigated. A

    solid-state silicon microchip for controlled release of

    single or multiple chemical substances on demand

    was developed [40] (Fig. 7). The release mechanism is

    based on the electrochemical dissolution of thin anode

    gold membranes covering microreservoirs filled with

    chemicals in solid, liquid or gel form. A sequential

    microfabrication process including photolithography,

    chemical vapor deposition (CVD), electron beam

    evaporation and reactive ion etching (RIE) has been

    used to create 34 microreservoirs on the chip, each

    covered by a thin gold membrane functioning as the

    anodes. The gold membrane can be dissolved when a

    Fig. 7. A solid-state silicon microchip for controlled release [40]. (Reprinted by permission from Nature 397, 335338 (1999) Macmillan

    Magazines Ltd.)

    Fig. 6. Micromachined silicon piezoelectric micropump. Left: photograph of the micropump chip. Right: SEM view of the pumping membrane

    (n 1999 IEEE) [37].

    S. Zafar Razzacki et al. / Advanced Drug Delivery Reviews 56 (2004) 185198 193

    http:///reader/full/page14http:///reader/full/page14http:///reader/full/page9http:///reader/full/page14http:///reader/full/page14http:///reader/full/page14http:///reader/full/page9http:///reader/full/page14http:///reader/full/page14
  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    10/14

    desired voltage is applied, and the drug inside diffuses

    out into the surrounding fluid. Each reservoir can be

    addressed individually, creating a possibility for

    achieving many complex release patterns.

    5. Smart integrated microsystems

    In order to activate the delivery of drug compounds

    or solutions from the various devices previously

    described, a signal must be received by the system

    to trigger its function. In most conventional devices,

    this signal is manually initiated. However, devices that

    function without human intervention can be devel-

    oped by integrating chemical sensors into these devi-

    ces. This feature is necessary in order to make drug

    delivery devices truly robust; the systems could be

    capable of monitoring a patients physiological con-

    dition and smart enough to deliver the necessary

    combination of drugs to treat the condition at any

    given time. This means that the drug delivery system

    should be able to: (a) monitor the physiological

    conditions inside the patient body and convert it to

    electronic signals by means of physical and chemical

    transducers; (b) receive the electronic signals, analyze

    them, and make proper control regulations by means

    of microcontrollers; and (c) release the appropriateamount of drugs by means of microactuators. Thus,

    future drug delivery systems might contain not only

    the microactuator components such as micropumps,

    microvalves and flow regulators, but also physical and

    chemical microsensors, and control electronics. Elec-

    trical interconnects, wires, and packaging components

    are also needed to obtain a fully functional, automat-

    ed, self-regulating microsystem. All of these compo-

    nents must be integrated into a miniaturized device so

    that reliability is increased and cost is reduced. From

    this point of view, functional integration could be oneof the most important requirements for future drug

    delivery systems.

    Although no fully integrated, fully functional drug

    delivery device has yet been reported, the technology

    required to build such a system exists. Similar tech-

    nologies are being studied for a number of other

    bioMEMS applications. For example, a microfluidic

    system has been developed for rapid delivery of small

    sample volumes to biosensors, which can assay sam-

    ples taken from a bulk flow [41]. Other microfluidic

    immunoassay devices have also been developed with

    the ability to self-calibrate [42]. Fluidic networks have

    been developed with integrated semiconductor detec-

    tors/emitters for fluorescent spectroscopy on smallvolumes of solution [43]. Cell lysis, multiplexed

    PCR amplification and electrophoretic analysis have

    been performed sequentially on an integrated mono-

    lithic device [44]. A microfabricated electrophoretic

    bioprocessor has been developed for integrated DNA

    sample desalting, template removal, pre-concentra-

    tion, and capillary electrophoresis (CE) analysis [45].

    In addition to these simpler attempts at integration,

    more components are being combined into highly

    integrated microfluidics-based DNA analysis Micro-

    systems [4]. These microfluidic systems have found

    many potential medical applications in clinical diag-

    nostics, immunoassays, DNA and protein separation

    and analysis, cell culture and handling, and drug

    delivery. These diverse applications reflect the many

    advantages of processing fluid systems in small dimen-

    sions: precise volumes of fluid can be moved rapidly

    and efficiently; chemical analysis is especially accu-

    rate, owing to the combination of small sample vol-

    umes and sensitive detection methods; and diagnostic

    and delivery methods can often be integrated in a single

    device. The last feature is particularly important for

    constructing a fully integrated drug delivery system.An example of such an integrated chemical anal-

    ysis system is shown in Fig. 8. These types of

    microfabricated devices are often referred to as

    Lab-on-a-Chip devices since they presumably con-

    tain most of the major components of a chemical or

    biochemical lab on a single substrate. The device

    shown has regions to accurately meter and mix nano-

    liter-sized samples, thermally cycle the chemical reac-

    tants, and separate and detect the products of that

    reaction [4]. All of those components fit together onto

    a device measuring approximately 1 cm

    2

    in total area,and the devices are fabricated using conventional

    photolithography and micromachining techniques.

    The device can be interfaced and controlled by a

    personal computer, suggesting that standalone opera-

    tion is feasible. Also, since only low voltages are used

    on the chip, batteries should be sufficient to power the

    chip, a necessity for portable, handheld operation.

    The Lab-on-a-Chip device described above is

    particularly important and relevant to drug delivery

    systems because it is a fully functional example of a

    S. Zafar Razzacki et al. / Advanced Drug Delivery Reviews 56 (2004) 185198194

    http:///reader/full/page13http:///reader/full/page11http:///reader/full/page13http:///reader/full/page14http:///reader/full/page14http:///reader/full/page14http:///reader/full/page14http:///reader/full/page14
  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    11/14

    system with active sensing on-chip. As discussed

    earlier, chemical sensing is vital to the development

    of a complex drug delivery system capable of self-

    regulated, responsive operation. The chip uses temper-

    ature sensors for closed-loop control of thermal reac-

    tions such as DNA amplification polymerase chain

    reaction (PCR), which requires temperature cycling

    between denaturation, primer annealing, and extension

    temperatures. Optical fluorescence detectors are alsoincluded on the device for the sensing of size-fraction-

    ated reaction products. Diodes are implanted on the

    silicon substrate and covered with an interference

    filter. When coupled with an excitation source, they

    are capable of detecting migrating bands of DNA

    during on-chip electrophoretic separation. These are

    just two examples of the types of sensing which can be

    performed on a microfabricated device. Further inves-

    tigation has been published on other chemical sensors

    such as on-chip electrochemical detection [46]. These,

    and perhaps other more sophisticated methods ofchemical sensing, can be integrated into the drug

    delivery systems of the future.

    In addition to the importance of sensing applica-

    tions, microfluidics will help to advance the function-

    ality of drug delivery devices. By taking advantage of

    the ability to store fluids in reservoirs on chip until

    mixing is desired, it is possible to achieve complicated

    drug release profiles, delivery schemes and chemis-

    tries. Delivery of multiple solutions without mixing is

    also possible using laminar flow systems [47]. The

    components used in typical Lab-on-a-Chip and

    other similar microfluidic devices can be combined

    to design other complex integrated fluidics systems for

    applications such as drug delivery, all within an ultra-

    compact device that can be produced at low unit cost.

    6. Commercial applications

    The drug delivery devices of the future are taking

    some important cues from integrated microfluidic

    devices. This challenge of integrating active sensing

    components into a compact, low-power microfluidic

    drug delivery system is one that has appealed to

    biotechnology companies looking to develop self-

    regulating microsystems as the complete therapeutic

    solution to chronic illnesses such as diabetes or HIV.

    The Ohio-based company ChipRx is developing an

    integrated, self-regulating responsive therapeutic de-

    vice (Fig. 9). ChipRx describes its target device ashaving biosensors, electronic feedback and drug/

    countermeasure release fully integrated. The match-

    stick-sized device is designed to sense the physiolog-

    ical levels of metabolites, such as glucose. When a

    change is detected, a signal is sent from the sensor to

    the batteries, which emit an electrical charge. The

    electrical charge then triggers the opening of a re-

    sponsive material, allowing the release of the desired

    therapeutic agent (e.g., insulin) from a reservoir.

    When glucose levels return to normal, the sensor

    Fig. 8. A schematic of an integrated microfluidic device for DNA analysis (reprinted with permission [4]).

    S. Zafar Razzacki et al. / Advanced Drug Delivery Reviews 56 (2004) 185198 195

    http:///reader/full/page13http:///reader/full/page12http:///reader/full/page14http:///reader/full/page14
  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    12/14

    stops the release of electrical charge from the battery,

    closing the reservoir and preventing the release of

    more insulin [48]. This type of continuously respon-

    sive, integrated delivery system will truly revolution-

    ize individualized patient care.

    The micro-well device described earlier is being

    developed by MicroCHIPS, for use as external and

    implantable systems for the delivery of proteins,

    hormones, pain medications and other pharmaceutical

    compounds [49] (Fig. 10). One great advantage of the

    MicroCHIPS multi-well approach is the ability to

    store and release multiple drugs or chemicals from a

    single device. With increased integration, these devi-

    ces show promise of being developed into an intelli-

    gent device. By coupling the release mechanism

    (described in an earlier section) of each well to

    sensors, complex chemical release patterns can be

    achieved as needed.

    TheraFuse of San Diego is combining microneedle

    technology with microfluidics to produce a next

    generation minimally invasive wearable drug infusion

    system. At approximately the same dimensions as a

    silver dollar coin, this wearable system will consist ofa disposable polymer component containing the ther-

    apeutic compound and microneedles held to the skin

    by an adhesive. A reusable unit containing micro-

    fabricated logic, metering and communication circuits

    can then be interfaced with the disposable component

    to form a completely integrated delivery system.

    Whereas most fluidics devices rely on pumps for fluid

    delivery, the TheraFuse device incorporates a pressur-

    ized reservoir used to move the solution through a

    closed fluidic network, until injection occurs across

    the layers of the skin. In addition, a metering system isintegrated into the fluidics component to insure accu-

    rate delivery. Like the Lab-on-a-Chip devices,

    TheraFuse is also attempting to design highly inte-

    grated microfluidic systems for drug delivery.

    7. Conclusions

    The average American probably visits a local drug

    store or pharmacy once a week to refill an old

    Fig. 9. A schematic of ChipRxs self-regulating responsive therapeutic system (reprinted with permission [48]).

    Fig. 10. Above, left: On the chip, medicine is stored beneath 50-Am

    squares of gold membrane (top). Applying a small electric charge

    dissolves the gold cap, releasing the drug (bottom). Above, right:

    One prototype chip holds 100 drug-containing reservoirs (top). The

    circuitry on the reverse side (bottom) directs electric current to each

    reservoir[40,49]. (Photographs: left (2) Reprinted by permission

    from Nature 397, 335 338 (1999) Macmillan Magazines Ltd.; right

    (2) Courtesy of MicroCHIPS, Inc./Carita Stubbe.)

    S. Zafar Razzacki et al. / Advanced Drug Delivery Reviews 56 (2004) 185198196

    http:///reader/full/page14http:///reader/full/page14http:///reader/full/page12http:///reader/full/page14http:///reader/full/page14
  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    13/14

    prescription or to pick up new medications. People

    have become used to taking a pill with their meals or

    administering an insulin injection every morning.

    Microfluidic drug delivery systems may provide alter-natives to these modes of administration. Just as Lab-

    on-a-Chip technologies are scaling down entire lab-

    oratories to a single device with the capability of

    performing complex biochemical assays, it is now

    foreseeable that a compact, microfabricated device

    might soon be able to perform the same function as a

    doctor and pharmacist. An integrated, smart device

    that is able to respond continuously to physiological

    changes and deliver precise amounts of therapeutic

    compounds will make a considerable impact on the

    way people live their lives. In the near future, the

    average American may not have to worry about filling

    any prescriptions at the pharmacy. The Pharmacy-on-

    a-Chip will be delivering drugs where they are

    needed, when they are needed.

    References

    [1] C. Montemagno, G. Bachand, S. Stelick, M. Bachand, Con-

    structing biological motor powered nanomechanical devices,

    Nanotechnology 10 (1999) 225231.

    [2] C.H. Mastrangelo, M.A. Burns, D.T. Burke, Integrated micro-

    fabricated devices for genetic assays, Microproc. and Nano-tech. Conf., Yokohama, Japan 1999.

    [3] R.L. Edelstein, C.R. Tamanaha, P.E. Sheehan, M.M. Miller,

    D.R. Baselt, L.J. Whitman, R.J. Colton, The BARC biosensor

    applied to the detection of biological warfare agents, Biosens.

    Bioelectron. 14 (2000) 805813.

    [4] M.A. Burns, B.N. Johnson, S.N. Brahmasandra, K. Handique,

    J.R. Webster, M. Krishnan, T.S. Sammarco, F.P. Man, D.

    Jones, D. Heldsinger, V. Namasivayam, C.H. Mastrangelo,

    D.T. Burke, An integrated microfabricated DNA analysis

    device, Science 282 (1998) 484 487.

    [5] J.G. Hardman, L.E. Limbird, Goodman and Gilmans the

    Pharmacological Basis of Therapeutics, 9th ed., McGraw-Hill,

    Health Professions Division, New York, 1996.

    [6] D.R. Reyes, D. Iossifidis, P.A. Auroux, A. Manz, Micro totalanalysis systems 1. Introduction, theory, and technology, Anal.

    Chem. 74 (2002) 26232636.

    [7] M. Gad-el-Hak (Ed.), The MEMS Handbook, CRC Press,

    Boca Raton, 2002.

    [8] M.J. Madou, Fundamentals of Microfabrication: the Science

    of Miniaturization, 2nd ed., CRC Press, Boca Raton, 2002.

    [9] N. Maluf, An Introduction to Microelectromechanical Sys-

    tems Engineering, Artech House, Boston, 2000.

    [10] M. Koch, A. Evans, A. Brunnschweiler, Microfluidic Tech-

    nology and Applications, Research Studies Press, Baldock,

    2000.

    [11] S.D. Senturia, Microsystem Design, Kluwer Academic Pub-

    lishing, Boston, 2001.

    [12] P.A. Auroux, D. Iossifidis, D.R. Reyes, A. Manz, Micro total

    analysis systems: 2. Analytical standard operations and appli-

    cations, Anal. Chem. 74 (2002) 26372652.[13] C.T. Culbertson, S.C. Jacobson, J.M. Ramsey, Microchip de-

    vices for high-efficiency separations, Anal. Chem. 72 (2000)

    58145819.

    [14] I.L. Medintz, B.M. Paegel, R.A. Mathies, Microfabricated

    capillary array electrophoresis DNA analysis systems, J. Chro-

    matogr. A 924 (2001) 265270.

    [15] E.T. Lagally, P.C. Simpson, R.A. Mathies, Monolithic in-

    tegrated microfluidic DNA amplification and capillary elec-

    trophoresis analysis system, Sens. Actuators, B 63 (2000)

    138146.

    [16] J.R. Webster, M.A. Burns, D.T. Burke, C.H. Mastrangelo,

    Monolithic capillary electrophoresis device with integrated

    fluorescence detector, Anal. Chem. 73 (2001) 16221626.

    [17] M. Krishnan, S.N. Brahmasandra, D.T. Burke, C.H. Burns,

    M.A. Burns, A novel strategy for the design of multiple re-

    action systems for genetic analysis, Sens. Actuators, A 95

    (2002) 250 258.

    [18] V.M. Ugaz, S.N. Brahmasandra, D.T. Burke, M.A. Burns,

    Cross-linked polyacrylamide gel electrophoresis of single-

    stranded DNA for microfabricated genomic analysis systems,

    Electrophoresis 23 (2002) 14501459.

    [19] H. Becker, L.E. Locascio, Polymer microfluidic devices, Ta-

    lanta 56 (2002) 267 287.

    [20] J.M.K. Ng, I. Gitlin, A.D. Stroock, G.M. Whitesides, Compo-

    nents for integrated poly(dimethylsiloxane) microfluidic sys-

    tems, Electrophoresis 23 (2002) 3461 3473.

    [21] T. Thorsen, S.J. Maerkl, S.R. Quake, Microfluidic large-scaleintegration, Science 298 (2002) 580584.

    [22] M.A. Burns, Everyones a (future) chemist, Science 296

    (2002) 18181819.

    [23] R. Gref, Y. Minamitake, M. Peracchia, V. Trubetskoy, V.

    Torchilin, R. Langer, Biodegradable long-circulating poly-

    meric nanospheres, Science 263 (1994) 16001603.

    [24] D. Luo, K. Woodrow-Mumford, N. Belcheva, W. Saltzman,

    Controlled DNA delivery systems, Pharm. Res. 16 (1999)

    13001308.

    [25] H. Cohen, R.J. Levy, J. Gao, I. Fishbein, V. Kousaev, S.

    Sosnowski, S. Slomkowski, G. Golomb, Sustained delivery

    and expression of DNA encapsulated in polymeric nanopar-

    ticles, Gene Ther. 7 (2000) 18961905.

    [26] T.A. Desai, Micromachined therapeutic delivery systems:from concept to clinic, Proc. SPIE 4265 2001, pp. 2635.

    [27] T.A. Desai, W.H. Chu, G. Rasi, P.S. Vallebona, E. Guarino, M.

    Ferrari, Microfabricated biocapsules provide short-term im-

    munoisolation of insulinoma xenografts, J. Biomed. Micro-

    dev. 1 (2) (1999) 131 138.

    [28] S. Henry, D. McAllister, M. Allen, M. Prausnitz, Microma-

    chined needles for the transdermal delivery of drugs, Proc.

    IEEE MEMS 98, 1998, pp. 494498.

    [29] D. McAllister, M. Allen, M. Prausnitz, Microfabricated micro-

    needles for gene and drug delivery, Annu. Rev. Biomed. Eng.

    2 (2000) 289313.

    S. Zafar Razzacki et al. / Advanced Drug Delivery Reviews 56 (2004) 185198 197

  • 8/8/2019 7 - Integrated Microsystems for Controlled Drug Delivery[1]

    14/14

    [30] S. Henry, D. McAllister, M. Allen, M. Prausnitz, Microfabri-

    cated microneedles: a novel approach to transdermal drug

    delivery, J. Pharm. Sci. 87 (1998) 922925.

    [31] J. Zahn, A. Deshmukh, A. Pisano, D. Liepmann, Continuous

    on-chip micropumping through a microneedle, Proc. IEEEMEMS 01, 2001, pp. 503506.

    [32] J. Chen, K. Wise, A multichannel neural probe for selective

    chemical delivery at the cellular level, IEEE trans. Biomed.

    Eng. 44 (8) (1997) 760769.

    [33] D. Papageorgiou, S.C. Bledsoe, M. Gulari, J.F. Hetke, D.J.

    Anderson, K.D. Wise, A shuttered probe with in-line flow-

    meters for chronic in-vivo drug delivery, Proc. IEEE MEMS

    01, 2001, pp. 212215.

    [34] P.P. Dario, M. Carrozza, Interfacing Microsystems and bio-

    logical systems, IEEE Proc. Int. Symp. Mic. Mach. and

    Hum. Sci. 95, 1995, pp. 5766.

    [35] D.L. Polla, A.G. Erdman, W.P. Robbins, D.T. Markus, J.

    Diaz-Diaz, R. Rizq, Y. Nam, H.T. Brickner, Microdevices

    in medicine, Annu. Rev. Biomed. Eng. 2 (2000) 551576.

    [36] W. Saltzman, W. Olbricht, Building drug delivery into tissue

    engineering, Nat. Rev., Drug Discov. 1 (2002) 177 186.

    [37] D. Maillefer, H. van Lintel, G. Rey-Mermet, R. Hirschi,

    A high-performance silicon micropump for an implant-

    able drug delivery system, Proc. IEEE MEMS 99, 1999,

    pp. 541546.

    [38] D. Maillefer, S. Gamper, B. Frehner, P. Balmer, A high-per-

    formance silicon micropump for disposable drug delivery sys-

    tems, Proc. IEEE MEMS 01, 2001, pp. 413417.

    [39] P. Cousseau, R. Hirschi, B. Frehner, S. Gamper, D. Maillefer,

    Improved micro-flow regulator for drug delivery systems,

    Proc. IEEE MEMS 01, 2001, pp. 527530.

    [40] J. Santini, M. Cima, R. Langer, A controlled-release micro-

    chip, Nature 397 (1999) 335338.

    [41] O. Hoffman, G. Voirin, P. Niedermann, A. Manz, Three-di-

    mensional microfluidic confinement for efficient sample de-

    livery to biosensor surfaces. Application to immunoassays onplanar optical waveguides, Anal. Chem. 74 (20) (2002)

    42505243.

    [42] C.X. Qiu, D.J. Harrison, Integrated self-calibration via electro-

    kinetic solvent proportioning for microfluidic immunoassays,

    Electrophoresis 22 (18) (2001) 39493958.

    [43] M.L. Adams, G.A. DeRose, S.R. Quake, A. Scherer, Funda-

    mental approach for optoelectronic and microfluidic integra-

    tion for miniaturizing spectroscopic devices, Func. Integ. Opt-

    elec-mech. Dev. and Sys. II Proc. Soc. Photo-opt. Instr. Engrs.

    SPIE 4647 2002, pp. 16.

    [44] L.C. Walters, S.C. Jacobson, N. Kroutchinina, J. Khandurina,

    R.S. Foote, J.M. Ramsey, Microchip device for cell lysis,

    multiplex PCR amplification and electrophoretic sizing, Anal.

    Chem. 70 (1998) 158 162.

    [45] B.M. Paegel, S.H.I. Yeung, R.A. Mathies, Microchip biopro-

    cessor for integrated nanovolume sample purification and

    DNA sequencing, Anal. Chem. 74 2002, pp. 50925098.

    [46] P. Selvaganapathy, M.A. Burns, D.T. Burke, C.H. Mastrange-

    lo, Inline Electrochemical detection for capillary electropho-

    resis, IEEE MEMS Conf. 01.

    [47] S. Takayama, E. Ostuni, P. LeDuc, K. Naruse, D.E. Ingber,

    G.M. Whitesides, Laminar flows: subcellular positioning of

    small molecules, Nature 411 (2001) 1016.

    [48] http://www.chiprx.com/products.html (01/21/03).

    [49] http://www.mchips.com/tech.html (01/21/03). (Courtesy of

    MicroCHIPS/Carita Stubbe).

    S. Zafar Razzacki et al. / Advanced Drug Delivery Reviews 56 (2004) 185198198

    http://www.mchips.com/tech.htmlhttp://www.chiprx.com/products.html

Recommended