Sie sind auf Seite 1von 17

CRITICAL REVIEW www.rsc.

org/loc | Lab on a Chip

Lab-on-a-chip devices for global health: Past studies and future


opportunities
Curtis D. Chin,a Vincent Linderb and Samuel K. Sia*a
Received 8th August 2006, Accepted 10th October 2006
First published as an Advance Article on the web 27th October 2006
DOI: 10.1039/b611455e

A rapidly emerging field in lab-on-a-chip (LOC) research is the development of devices to


improve the health of people in developing countries. In this review, we identify diseases
that are most in need of new health technologies, discuss special design criteria for LOC
devices to be deployed in a variety of resource-poor settings, and review past research into
LOC devices for global health. We focus mainly on diagnostics, the nearest-term application
in this field.

1. Introduction profound issue of global health, where the challenges in device


designs are arguably the most demanding, and the need for
Lab-on-a-chip (LOC) technologies have a tremendous but new health technologies the greatest.
unproven potential to improve the health of people in There is an urgent need in developing countries for new
developing countries. Ever since the modern inception of health-related technologies, and specifically, new technologies
LOC and microfluidic technologies around 1990, use in remote for health diagnostics. For example, in one survey of
settings has been perceived as potentially one of the most international scientists familiar with the public health pro-
powerful applications of the technology by taking advantage grams of developing countries, Singer and colleagues found
of its small size, low volume requirement for samples, and rapid that the top-ranking overall priority was ‘‘modified molecular
analysis. Indeed, portable LOC devices are now beginning to technologies for affordable, simple diagnosis of infectious
be used in remote settings, as a result of developments in diseases’’.1 Similarly, in a study by the Bill and Melinda Gates
integrating fluid actuation, sample pre-treatment, sample Foundation and the NIH to identify ‘Grand Challenges for
separation, signal amplification, and signal detection into Global Health’, two of the 14 priorities involved diagnosis
a single device. As they stand, these devices are not yet and measurement of patients’ health statuses (i.e. ‘‘develop
appropriate for use in the extreme resource-poor settings of technologies that allow assessment of individuals for multiple
developing countries; nevertheless, these advances place the conditions or pathogens at point-of-care’’, and ‘‘develop
field of LOC research in a prime position to tackle the technologies that permit quantitative assessment of population
health status’’).2 LOC research holds substantial potential for
a fulfilling these priorities by automating complex diagnostic
Department of Biomedical Engineering, Columbia University, 351
Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, procedures that are normally performed in a centralized
USA. E-mail: ss2735@columbia.edu laboratory into a hand-held microfluidic chip; this capability
b
SAMLAB, Institute of Microtechnology, University of Neuchâtel, Rue could empower health-care workers and patients with impor-
Jaquet-Droz 1, P. O. Box 526, CH-2002 Neuchâtel, Switzerland
tant health-related information in even the most remote
settings. To this effect, funding by philanthropic foundations
(such as those from Doris Duke, Soros, and Gates) are leading
Curtis Chin is a PhD student in the Department of Biomedical the development of microfluidics technologies for diagnostics
Engineering at Columbia University. He obtained his BS in
in developing countries. The broad aim of these scientific
Chemical Engineering from the Massachusetts Institute of
Technology. initiatives is to combine new diagnostic and prevention
methods with treatment to improve public health,3 which is
Vincent Linder, PhD, is a research scientist at the University of in turn linked closely to the macroeconomic health of a
Neuchatel and Claros Diagnostics. He holds a MSc in nation.4
Chemistry and a PhD in Sciences from the University of In this review, we aim to aid interested scientists and
Neuchâtel (Switzerland), where he worked on microfluidic engineers by systematically reviewing the fledgling field of
technology for immunoassays. He completed his postdoctoral LOC devices for global health. We will focus mainly on
work in the Department of Chemistry at Harvard University. diagnostics, the nearest-term application of LOC devices,
although we will also discuss other applications. We will first
Samuel Sia, PhD, is an Assistant Professor of Biomedical
Engineering at Columbia University. He holds a BS in identify the most critical health conditions and diseases that
Biochemistry from the University of Alberta (Canada), and a are in need of new diagnostic methods, with an emphasis on
PhD in Biophysics from Harvard University. He completed his those in need of new LOC diagnostic devices (Section 2). In
postdoctoral work in the Department of Chemistry at Harvard subsequent sections, we will discuss the special design criteria
University. that are needed for LOC devices to be deployed in developing

This journal is ß The Royal Society of Chemistry 2007 Lab Chip, 2007, 7, 41–57 | 41
countries (Section 3), and provide a review of past and current childhood-cluster diseases (such as diphtheria, measles,
studies on LOC devices for global health, as well as examples pertussis, and tetanus). Another important category of infec-
of LOC devices that have not been—but could be—applied to tious diseases is neglected tropical diseases (which includes
these settings (Section 4). We will conclude with a summary lymphatic filiariasis, dengue, Chagas disease, leishmaniasis,
and future directions (Section 5). Throughout the review, we onchocerciasis, schistosomiasis, trypanosomiasis, trachoma,
will identify examples of past research, current promising and guinea worm), which cause 500 000 deaths annually.
technologies, and future challenges and opportunities. Although they do not contribute as significantly as some other
infectious diseases by the measure of DALYs in the Global
2. Current need for diagnostic devices in developing Burden of Disease report, the real burden of disease is likely to
be higher than these estimates, with up to 90% of the burden
countries
concentrated in sub-Saharan Africa (for example, there are
In developed and developing countries alike, early and 200 million cases of hookworm infections in Africa alone).11
accurate diagnosis is important for the health of individual Current methods for diagnosing neglected diseases are
patients as well as that of the general public: it permits prompt cumbersome, invasive, and largely inadequate (e.g. for human
and proper treatment of patients, limits the spread of disease in African typanosomiasis and visceral leishmaniasis, see ref. 12),
the population, and minimizes the waste of public resources on a consequence of the low priority given to neglected diseases
ineffective treatments.1 In developing countries, the value of for research funding (as pointed out poignantly by studies such
diagnosis for certain diseases is sometimes mitigated by the as the 10/90 Report on Health Research, www.globalforum-
lack of available treatment (for example, in cases of certain health.org, and ref. 13). In one analysis to define priorities for
neglected tropical diseases). On the whole, however, the value diagnostics development, Mabey and colleagues charted the
of diagnosis is very high in developing countries: early need versus feasibility for selected diseases (including many
diagnosis, although not without logistical hurdles, can often neglected diseases), with the conclusion that African trypano-
lead to some kind of treatment (either directly against the somiasis, visceral leishmaniasis, and TB are three of the tests
condition or at worst palliative care), and investments in most in need of development.14 Other important diseases
diagnostics and prevention can be more cost-effective than include sexually-transmitted infections other than HIV/AIDS
treatment.5 Moreover, point-of-care devices can improve the (such as hepatitis B and C, chlamydia, gonorrhea, and
epidemiological surveillance of diseases,6 which is an especially syphilis), some of which (most notably, hepatitis B and C,
challenging problem in developing countries. and HIV) are bloodborne pathogens that can also be
For scientists and engineers who aim to design new transmitted by contaminated needles as well as contaminated
diagnostic technologies, a crucial question for achieving real- blood supply for transfusions.15
world impact is which health conditions in developing Like infectious diseases, the burden of non-communicable
countries are most in need of diagnostic devices. In a study diseases is significant (at 43.5% DALY, it even exceeds that of
led by Murray and Lopez, the World Health Organization infectious diseases by a large margin) (Table 1); unlike
conducted an unprecedented and comprehensive initiative infectious diseases, the burden of non-communicable diseases
to compile statistics for comparing the relative burden of in developing countries is often underappreciated.16 The
diseases, conditions, injuries, and risk factors on a global specific list of important non-communicable diseases is
scale.7–9 In Table 1, we list the most common diseases by familiar to readers from Western countries: cardiovascular
disability-adjusted life years (DALYs) in developing countries, disease (such as ischaemic heart disease and stroke), cancer,
a metric that accounts for years of life lost due to premature neuropsychiatric conditions (such as unipolar depressive
mortality as well as disability (in order to properly account for disorder), and respiratory diseases (such as chronic obstructive
the impact of conditions that cause significant ill health but pulmonary disorder and asthma). As the standard of living in
few direct deaths, such as neuropsychiatric conditions9,10). developing countries improves and average life span increases,
As expected,10 infectious diseases constitute a large burden the burden of disease will gradually shift to the non-
of disease in developing countries (32.1%; by comparison, they communicable diseases; this shift is exacerbated by changes
represent only 3.7% of total DALYs in developed countries). in diet (towards saturated fats and sugars) and high tobacco
The trifecta of HIV/AIDS, malaria, and tuberculosis (TB), use.16 Already, obesity and diabetes are increasingly prevalent
which has merited a dedicated focus from the international in developing countries.17 Even for children in developing
community (most notably the Global Fund, which has thus far countries, asthma, epilepsy, dental caries, diabetes, rheumatic
committed $5.5 billion, www.theglobalfund.org), constitutes heart disease, and injuries are becoming increasingly promi-
an important 12% of DALYs in developing countries. The nent contributors to morbidity.18 As these trends develop,
social impact of these diseases stretches beyond the DALY accessibility of the corresponding diagnostic technologies in
statistics, however, since HIV/AIDS (along with common developing countries cannot be assumed from their likely
coinfections of TB) targets healthy adults, thereby leaving availability in Western countries, due to the special constraints
behind villages of orphans which destroy the underlying of resource-poor settings (see Section 3).
fabric of entire communities. Other infectious diseases are Maternal, perinatal and nutritional diseases contribute a
also important. Most significantly, lower respiratory infections significant fraction of DALYs (11.8%) in developing
and diarrheal diseases (such as rotavirus and cholera) countries (Table 1). Two important risk factors for material
impose large burdens; these diseases are also the biggest diseases include anemia and vitamin A deficiency;10 although
killers of children,10 even more so than vaccine-treatable treatment is the most important consideration for these two

42 | Lab Chip, 2007, 7, 41–57 This journal is ß The Royal Society of Chemistry 2007
micronutrient deficiencies, diagnosis can lead to improved upper-arm circumference in order to diagnose the stage of
epidemiological surveillance. Overall, malnutrition is the single malnourishment; biochemical measurements may be useful for
most important cause of loss in global health, with the greatest more specific diagnoses (e.g. serum albumin levels for protein-
effect felt in sub-Saharan Africa.10 To combat malnutrition in energy malnutrition).
children under five years of age, a simple bracelet made by Finally, intentional and unintentional injuries (including
Medicins Sans Frontieres can be used to measure the mid war) constitute a significant DALY fraction (12.5%) that rivals

Table 1 Important diseases in developing countries, burden of disease, diagnostic assays, and corresponding potential LOC devices

% %
Diseasea DALYb Type of assayc Device Disease DALY Type of assay Device
Communicable diseases 32.1 Non-communicable diseases 43.5
Respiratory infect. 6.8 IA Sec. 4.2 Neuropsychiatric conditions 11.7 Sx/Hx/PE N/A
(lower, upper, Slide agg. w/antisera Sec. 4.2 (unipol & bipol deprs, Hormone levels Sec. 4.5
ottis media) GS and cul. of CSF Sec. 4.4 others)
HIV/AIDS 6.1 IA for a-HIV Ab Sec. 4.2 Cardiovascular diseases (isc., 9.5 ELISA of CRP, BNP Sec. 4.2
(¡ vs quant) hyp., rheum., inflamm.) Cholesterol test Sec. 4.5
RT-PCR for HIV RNA Sec. 4.3 Sense order diseases (cataracts, 4.6 Sx/Hx/PE N/A
CD4+ counts Sec. 4.4 hearing loss, glaucoma)
Diarrheal diseases 4.5 EIA Sec. 4.2 Cancer 4.2 IA of biomarkers Sec. 4.2
(rotavirus, Latex agg. assay of stool Sec. 4.2 (e.g. PSA)
cholera) RT-PCR Sec. 4.3 Gene expression Sec. 4.3
Malaria 3.4 Mic. of blood smears Sec. 4.4 Respiratory diseases (COPD, 3.5 Spirometry N/A
IC test for HRP-2, Sec. 4.2 asthma, others) Sx/Hx/PE N/A
LDH, PS Digestive diseases (liver 3.0 Complete blood count Sec. 4.4
PCR for plasmodium Sec. 4.3 cirrhosis, peptic ulcer Electrolytes, creatinine Sec. 4.5
Tuberculosis 2.5 Tuberculin skin test N/A disease) Elevated liver enzymes Sec. 4.2
Mic. and sputum culture Sec. 4.4 Congenital abnormalities 1.9 Karyotype analysis N/A
Release of IFN-c Sec. 4.2 (heart dis., DS) trisomy 21
from blood RBC count Sec. 4.5
Measles 1.6 Virus isolation Sec. 4.3 Sx/Hx/PE N/A
Monitor specific IgG titers Sec. 4.2 Musculoskeletal dis. 1.8 Differential WBC Sec. 4.4
IA for virus (MV) IgM Sec. 4.2 (osteoarthritis, ELISA rheumatoid Sec. 4.2
Pertussis 0.9 PCR of nasal secretions Sec. 4.3 rheumatoid arthritis) factor
Culture Sec. 4.4 Genitourinary dis. (neph., 1.0 Sx/Hx/PE (physician) N/A
Tetanus 0.5 Culture Sec. 4.4 bphyp.)
IA Sec. 4.2 Diabetes mellitus 1.0 Plasma/glucose test Sec. 4.5
Meningitis 0.4 CSF glucose Sec. 4.5 Insulin Sec. 4.2
CSF cell count Sec. 4.4 Endocrine disorders 0.5 Hormone levels Sec. 4.5
GS & cul. of CS Sec. 4.4 X-rays, radiological N/A
Lymphatic filariasis 0.4 ELISA Sec. 4.2 exams
IC test of W. bancrofti Sec. 4.2 Oral conditions (dental caries, 0.5 Sx/Hx/PE (dentist) N/A
Mic. blood samples Sec. 4.4 edentulism, others) X-rays, radiological N/A
midnight exams
Hepatitis B & 0.3 IA HBsAg, anti-HBs, Sec. 4.2 Skin diseases 0.3 Sx/Hx/PE (physician) N/A
hepatitis C antiHBc
IA liver enzyme, AFP Sec. 4.2 Maternal, perinatal, and 11.8
HCV detection & Sec. 4.3 nutritional conditions
genotying Perinatal cond. (LW., BA., 7.0 Sx/Hx/PE (physician) N/A
Syphilis 0.3 VDRL Sec. 4.2 trauma)
RPR Sec. 4.2 Nutritional deficiencies 2.4 IA albumin Sec. 4.2
FTA-ABS of TPHA Sec. 4.2 (protein-energy, Fe-anaemia) Cell count anemia Sec. 4.4
Chlamydia 0.3 PCR from urine dipstick Sec. 4.3 Maternal cond. (hem., 2.4 Haemotology Sec. 4.4–5
ELISA of C. trachomatis Sec. 4.2 seps., hypertens.)
antigens
NAAT, hybridization test Sec. 4.3 Injuries 12.5
Gonorrhea 0.2 Mic & cul. urethral cervical Sec. 4.4 Unintentional injuries (RA, 9.2 Analytical toxicology Sec. 4.2,5
Trachoma 0.2 Culture Sec. 4.4 falls, fires, dr.)
Antigen detection Sec. 4.2 Intentional injuries (violence, 3.3 Culture (eg. C. tetani) Sec. 4.4
IC test Sec. 4.2 SII, war) IA Sec. 4.2
Leishmaniasis 0.2 Mic. & cul. spleen, bone Sec. 4.4
marrow
Trypanosomiasis & 0.2 Agg. test for IgM, PCR Sec. 4.2–3
schistosomiasis parasite
Mic. & cul. spleen, bone Sec. 4.4
marrow
Intestinal nematode 0.2 Mic. & cul. anal swab Sec. 4.4
infect.
Japanese 0.1 IA of blood and Sec. 4.2
encephalitis spinal fluid

This journal is ß The Royal Society of Chemistry 2007 Lab Chip, 2007, 7, 41–57 | 43
Table 1 Important diseases in developing countries, burden of disease, diagnostic assays, and corresponding potential LOC devices (Continued )
a
Disease categories as grouped by Murray and Lopez,8 except maternal, perinatal, and nutritional conditions are shown as a separate
category. b The burden of disease as measured by the percentage contribution to total disability-adjusted life years (DALYs) in middle- and
low-income countries. Percentages were derived using data from WHO Global Burden of Disease 2002 revised estimates by World Bank income
groups (high, upper middle, lower middle, and low income countries)9 http://www3.who.int/whosis/
menu.cfm?path=whosis,burden,burden_estimates,burden_estimates_2002N,burden_estimates_2002N_2002Rev&language=english c Data taken
from ref. 5, 7–9, 18. Abbreviations used: immunochromatographic (IC), immunoassay (IA), enzyme-linked immunosorbent assay (ELISA),
microscopy (mic.), venereal disease research laboratory (VDRL), rapid plasma regin (RPR), fluorescent treponemal antibody-adsorption (FTA-
ABS), T. pallidum hemagglutination assay (TPHA), agglutination (agg.), culture (cul.), iodine (iod.), vitamin (vit.), gram stain (GS),
cerebrospinal (CSF), hypertension (hypertens.), iron (Fe), infections (infect.), Down syndrome (DS), disorder (dis.), ischaemic cerebrovascular
disease (isc.), rheumatory heart disease (rheum.), inflammatory heart disease (inflamm.), depression (deprs.), hepatitis B surface antigen
(HBsAg), antibody to hepatitis B surface antigen (anti-HBs), antibody to hepatitis B core antigen (anti-HBc), alpha-fetoprotein (AFP),
hepatitis C virus (HCV), C- reactive protein (CRP), prostate-specific antigen (PSA), brain naturietic peptide (BNP), hypertensive heart disease
(hyp.)., nephritis and nephrosis (neph.), benign prostatic hypertrophy (bphyp.), low weight (LW.), birth asphyxia (BA), hemorrhage (hem.),
sepsis (seps.), conditions (cond.), road accidents (RA), drownings (dr.), self-inflected injuries (SII), white blood cell (WBC), Sx/Hx/PE
(symptoms/medical history/physical exam), red blood cell (RBC).

that of other categories, and which is higher than that in developing countries. For example, in developing countries,
Western countries (9.1%) (Table 1). A number of behavioral different design criteria apply to centralized testing in a
changes can be undertaken for preventing injuries in develop- national laboratory, in a rural health clinic, and in a remote
ing countries.19 From the perspective of diagnostic devices, this setting with no infrastructure (Table 2). Similarly, there exist
burden may call for devices for detecting poisons, diagnosing subtle but important distinctions in the constraints. For ‘low
neuropsychiatric conditions (such as epilepsy) and substance cost’, the economics of centralized testing may allow for the
of abuse to ensure prompt treatment, and diagnosing tetanus purchase of a moderately priced or even expensive fixed
infections to strengthen epidemiological surveillance. instrument (tens of thousands of dollars), if the cost of
To diagnose this wide array of diseases and conditions, disposables is kept sufficiently low. By contrast, remote point-
assays with a variety of methodologies will be needed. The of-care testing requires low cost in both the fixed instrument
types of assays that are currently used to diagnose them are and the disposable (pennies). Since these considerations hold
listed in Table 1; some assays are in great need of new direct pertinence to the design of the diagnostic technology, it
diagnostic methods, and some are not. For each diagnostic is beneficial to be aware of the final targeted use of the device
assay, potential corresponding LOC devices are also listed. at the earliest design stages. For example, in the extreme points
Analysis of this table, which cross-lists diseases and techno- of the landscape of resource availability in developing
logies, reveals a couple of points. (1) Similar classes of analytes countries, devices targeted for national centralized laboratories
(e.g. proteins, nucleic acids) serve as useful markers for very may include currently available technologies for Western
different diseases and conditions; hence, similar designs of countries (e.g. 96 well plate assays using an expensive and
diagnostic technologies will be applicable for disparate classes bulky fluid-handling machine and detector), and devices for
of diseases. (For example, yes/no protein markers are useful point-of-care testing in rural settings will need to be designed
for diagnosis of HIV/AIDS as well as indicators of coronary with all of the constraints in mind.
heart disease). In Section 4, we will review potential LOC In the centralized laboratories of Western countries (run by
technologies as grouped by analytes. (2) Multiple classes of companies such as Quest Diagnostics as well as in-house
assay technologies are needed to produce complete diagnostic centers in hospitals) with skilled personnel, established
information for groups of related diseases, and often even for infrastructure, and high financial resources, sophisticated tests
a single disease (for confirmatory testing, identification of such as microscopy, ELISAs, and nucleic acid amplification
resistant subtypes, and/or staging of a disease). (For example, tests are routinely performed (Table 2).14 Although it would be
yes/no testing for antibodies, analysis of RNA levels, and desirable for diagnostic tests in this setting to meet criteria such
counting of CD4+ lymphocytes are all crucial information for as rapid analysis to improve efficiency of operation, there exist
diagnosing and staging HIV/AIDS.) This observation calls for few constraints (compared to other settings for diagnostics)
carefully considering the integration of multiple modular that result from low capital or poor infrastructure.
technologies at the earliest design stages of LOC diagnostic In developing countries, there exist a small number of
devices for developing countries. privately funded centralized testing centers that have essen-
tially the same infrastructure and resources as the testing
3. Third-world design constraints centers in Western countries (Table 2). By contrast, in most
centralized testing centers of developing countries, cost (due to
Like no other setting, the use of LOC devices in developing limited budgets via public financing) and availability of skilled
countries poses a set of extremely challenging design criteria. workers (due to attrition of the best workers) are still limited
For maximum range of use, a LOC device would have to compared to the centers of Western countries. Thus, the cost
perform reliably under the well-documented constraints of low of the microfluidic device (which includes both the material
cost, absence of trained workers, lack of electricity, poorly and the manufacturing process) must be kept low in most
equipped laboratories, and transportation and storage in settings in developing countries. Moreover, for remote point-
unrefrigerated conditions with rough handling.14 In practice, of-care testing in developing countries, the fixed instrument
however, not all of these constraints apply to all settings in must be portable and cheap, and the disposable must be

44 | Lab Chip, 2007, 7, 41–57 This journal is ß The Royal Society of Chemistry 2007
Table 2 LOC design constraints in different settingsa

Low- &
Low- & middle- Low- &
High- middle- High- income middle- Low- &
income income income countries income middle-
countries countries countries national/ countries income
national centralized Extra- point-of- regional rural countries
testing lab terrestrial care testing health remote
Constraint on design criteria centerb (private)c Military sensors (POC) centerd clinice POCf

Absence of ground electricity U U I I U U S I

This journal is ß The Royal Society of Chemistry 2007


Low cost of fixed instrument U U U U U S I I
Low cost of disposables U U U U S I I I
Absence of trained end-users U U S N/A I S S I
Absence of equipped end-use U U I I I S I I
facilities
Rapid Analysis U U I S I U I I
High sensitivity & specificity I I I I I I I I
Large fluctuations in U S I I I S I I
temperature
Portability U U I I I S I I
Rough handling U S I I S S I I
a
I = important, S = somewhat important, U = unimportant. b In a typical in-house hospital laboratory in Western countries, for example, costs of reagents are $5 for immunoassays, and $5–$30
for nucleic acid tests.138 Reimbursement rates are typically $20–$50 per test for immunoassays, and $50–$300 for nucleic acid tests. Fixed instruments (such as liquid handling robots and detectors)
can cost hundreds of thousands of dollars, although they can be leased or borrowed using reagent-rental agreements. Typically, turnaround times for most tests of between 0.5 and 2 days (from
the time it is ordered to the time the result is provided to the physician). Excluding sample transportation to the lab, turnaround time for emergency testing is about 1 h. Most reagents are stored
in the fridge until time of use. c This heading describes centralized testing centers that are funded by private organizations (e.g. philanthropies, NGO). These testing centers are typically located in
urban locations, and can be as well-equipped as testing centers in Western countries. An example is the MTCT-Plus Initiative that funded two clinical laboratories in Maputo and Nampula to
provide diagnostics services for supporting HIV programs in Mozambique. d Centralized testing centers in national or regional health centers are typically funded by the government, and are
responsible for many of the tests in the country. The quality of these health centers vary substantially, depending on the investment of the government into public health infrastructure.4 For
example, a cell-counting instrument (about $20 000) would be among the most expensive instruments, with typical equipment including a spectrophotometer, a microscope and a centrifuge costing
$1000 to $2000 each. e Rural towns often have a local health clinic or a hospital, with a wide range of conditions. Often, their clinical laboratories are run by poorly trained workers, are poorly
equipped, and have intermittent or no ground electricity. One such laboratory would be equipped, for instance with a centrifuge (manual or electric), a microscope (for counts of cells like platelets
or total WBC, the diagnosis of TB using colorations and the direct observation of stool), semi-quantitative strip tests for clinical chemistry applications (i.e., pH and sugar in urines), agglutination
tests for HIV, and the reagents for basic tests like blood type compatibility on glass slides. f These tests can be administered anywhere in developing countries. Currently used non-LOC tests in this
category include yes/no immunochromatographic tests for malaria (i.e., a Paracheck Pf, which at a price of $1 can be limited to an emergency case for ruling out infection of Plasmodium
falciparum) or semi-quantitative strip tests for clinical chemistry. An external instrument must be light, portable, rugged, and cheap (tens of dollars or cheaper, for widespread use), with very cheap
disposable microfluidic chips (pennies). For example, optical microscopes in such settings can be only equipped with a mirror as a light source to facilitate maintenance.

Lab Chip, 2007, 7, 41–57 | 45


Fig. 1 Pictures of current LOC devices for use in remote settings. (A) A handheld device, iSTAT, for measuring electrolyte levels. Taken from
www.istat.com with permission of i-STAT Corporation. (B) A portable device and a fixed instrument for analyzing DNA from the company
Cepheid. Taken from www.cepheid.com with permission of Cepheid Inc. (C) Mars Organic Analyzer, an extraterrestrial LOC device. Reprinted
from ref. 24. Copyright 2005 by the National Academy of Sciences of the United States of America, all rights reserved.

extremely cheap. All components of the device (including appropriate LOC technologies for use in developing countries.
the instrument and disposable) must be robust and rugged In general, the differences in appropriate LOC technologies are
under a variety of environmental conditions. Overall, remote pronounced between centralized testing versus point of care,
point-of-care testing in developing countries imposes perhaps with cost being the distinguishing feature between technologies
the severest constraints on the design of LOC devices, with for use in high-income versus low-income countries.
extremely low cost a distinguishing feature from most
applications in the Western world. Material and manufacturing
Although interest in designing LOC devices for developing
countries is only beginning, it will be beneficial to exploit the To minimize the cost of the microfluidic device, it is important
commonalities in the settings of developing countries with to reduce the footprint of expensive components such as glass
other resource-poor settings in Western countries (Table 2). ($500–4000 m22),26 quartz, and silicon (a number of challen-
For example, one can leverage the large body of existing ging issues exist in the scale-up of silicon micromachining for
research on LOC designs for point-of-care testing for biological devices27). An alternative is to use plastics, which
physicians’ and home use,20,21 devices for military applica- are inexpensive (the manufacturing cost of an injection-
tions22 and first responders, and extraterrestrial sensors23–25 in moulded device is generally less than $0.3028), available in an
designing LOC devices for developing countries (Fig. 1). In all abundant choice of materials, and appropriate for single-use
these settings, integration, portability, low power consump- disposal to avoid cross-contamination. Challenges in plastic
tion, automation, and ruggedness are important qualities. include minimization of batch-to-batch variation, improve-
These general design constraints suggest some LOC com- ment in chemical resistance, improvement in control over
ponents and procedures to be more appropriate than others surface chemistry, and compatibility with fluorescence.26,29,30
in resource-poor settings. In Fig. 2, we outline a sample of Additional steps for processing the microfluidic chip (such as

Fig. 2 A range of appropriate LOC procedures for different settings.

46 | Lab Chip, 2007, 7, 41–57 This journal is ß The Royal Society of Chemistry 2007
pre-treatment of surfaces with capture reagents) should be geometry and topography of the microchannels, can also be
simple and scalable to minimize the cost of the manufacturing used to mix and dilute samples.49 In general, however,
process. heterogeneous assays (which include many immunoassays)
do not require mixing since the analyte is captured on the
Storage and transportation surface.
Unlike controlled research environments, the LOC device
Signal detection
will be subjected to a variety of environmental conditions.
Reagents, including those stored inside the microfluidic chip, An intrinsic challenge in microfluidics is detection of a signal
must be stable to fluctuations in temperature as well as emanating from a small physical region; in developing
physical shocks. Methods for stabilizing dry reagents (such as countries, this detection must be inexpensive, and ideally, use
trehalose,31 a chemical that has been used to stabilizing dried compact instrumentation that consumes little power.
proteins in conventional 96 well ELISA assays) and wet Fluorescence is a sensitive and popular detection method,
reagents32,33 will be needed. but typically requires expensive and complicated optics and
consumes significant power, whereas absorbance can be low-
Sample pre-treatment cost.44,50 With simple electronic modulation, the background
in optical detection can be shielded from ambient light.44
A number of sources of physiological fluids are available. Electrical measurements such as conductance are also poten-
Although whole blood (from venipuncture or finger prick) tially appropriate low-cost and portable detection methods for
and its derivatives (plasma and serum) are most common, use in developing countries.51
the use of non-invasive samples such as saliva34 and urine35 are
gaining prominence. For real-world samples, sample pre- Disposal
treatment before the analysis step is needed. Pre-treatment
steps include sampling, extraction, filtration, pre-concentra- In point-of-care settings in both Western and developing
tion, and dilution;36 a centrifugation or filtration step is countries, it is ideal to contain the chemical reagents and
necessary, for example, to obtain plasma or serum from whole blood samples in the LOC device for disposal. Also, because
blood. In a centralized laboratory, these steps can be incineration is often not accessible, environmentally friendly
performed by a technician or a liquid handling robot before chemicals are preferred.
injection into the microfluidic chip. In remote settings,
however, automation and integration of these steps in the Overall strategy for development of LOC devices
LOC device is ideal.37–43 To minimize cost and power More broadly, in developing a LOC device for a new setting,
consumption, passive methods may be most appropriate. scientists and engineers can take two different approaches:
adapt existing methods or design new technologies. Both
Fluid actuation approaches have been undertaken in the past for developing
An ideal LOC device for developing countries should be countries. Adaptation of Western-world technologies for use
capable of actuating the flow of fluids with reliable flow rates in developing countries has a number of precedents in
using inexpensive and compact instrumentation. Electrokinetic technology-transfer projects; to be successful, these projects
actuation of fluids is popular in research laboratories, but it must carefully take into account the local needs, culture, and
requires a charged surface for electroosmotic flow (which constraints.52 It is arguably more challenging (but perhaps
limits the type of material that can be used) and a high voltage more scientifically rewarding) to develop new technologies
supply. Pneumatic actuation may be most practical for with consideration of local factors from the earliest stages of
portable applications, with battery- or hand-powered vacuum design. Collaboration with local partners could produce a
sources.44 Capillary force is a simple method for pumping device that best suits the health needs of the local people
fluids in portable LOC devices.45 (rather than a device driven by the convenience of available
technology); with the right initial testing group, this route can
Fluid control succeed extremely well as a disruptive technology.53

For complex assays, a series of different reagents need to be


4. Review of current work on LOC devices for global
delivered into the microfluidic chip. In centralized testing
health
facilities, these procedures can be performed manually by a
technician, an external liquid handling robot, or on-chip valves 4.1 Overview
that are controlled by an external instrument. For portable
Examination of diagnostic tests that are currently used in the
automated devices, passive delivery of a series of reagents is an
field in developing countries can bring insights for designing
attractive option.32,45–47
LOC devices. Most of the current diagnostic tests used in
developing countries are simple to use and provide rapid
Mixing
results.54 The most prevalent example is immunochromato-
Some assays will require mixing of samples with different graphic tests (also known as dipstick or lateral-flow tests),
reagents. In such cases, active micromixers can be used if a which provide yes/no results in minutes in the form of a visible
power supply is available.48 Passive mixers, which rely on the band (these tests typically use gold colloids or latex beads

This journal is ß The Royal Society of Chemistry 2007 Lab Chip, 2007, 7, 41–57 | 47
conjugated to antibodies).55–57 Moreover, immunochromato- (1) Fluid actuation. Capillary force is a simple method for
graphic strips are cheap to produce. These strip tests, however, pumping fluids in a LOC device, because the liquid flows
are not quantitative, and are not sufficiently sensitive for the spontaneously with no external power or moving parts.
detection of all important markers. As such, development of Delamarche and co-workers demonstrated autonomous
strip tests for diseases in developing countries (such as capillary flow in an array of microchannels to carry out an
chlamydia and trachoma from Lee’s group)58 is ongoing. immunoassay for C-reactive protein.45 The sensitivity was
LOC devices bring exciting capabilities of high analytical increased by integrating Pelletier elements underneath the
performance, but they must be made cost-effective, among LOC device to tune the rate of evaporation and hence capillary
other important requirements (Table 2). Also, LOC devices flow.47 As an alternative to capillary force, pressure or vacuum
can also potentially be used for multiplexed and parallel can be generated by applying a finger on the surface of a LOC
analysis of many relevant markers at once; this capability, device, as illustrated by an assay for botulinum toxin
however, is challenging since different analytes typically call developed by Beebe and co-workers,33 or the commercially
for the designs of different LOC methods. Below, we review available ABO Card from the company Micronics. A hand-
LOC studies that have been applied (or have the potential to operated vacuum pump can also be used to actuate fluid flow
be applied) for use in developing countries, as grouped by the in the field.32,44
class of analytes.
(2) Mixing and fluid control. Besides pumping, fluid mixing
4.2 Proteins is often required in protein assays. In resource poor-setting,
passive mixers offer the advantages of intrinsic automation
A wide range of diseases is characterized by changes in protein (no user intervention) and the absence of power requirements.
concentrations in a patient’s physiological fluids. These Planar mixers based on chaotic mixing with staggered
diseases span viral infections (e.g. anti-HIV antibodies as a herringbones,61 creation of bubbles,62 in situ photopoly-
marker for HIV/AIDS), bacterial infections (e.g. enterotoxin B merized porous material,42 three-dimensional microchannels,41
as a marker for Staphylococcus aureus), parasitic infections design of curved channel geometries,63 and other methods48
(e.g. histidine-rich protein 2 as a marker for malaria), and have demonstrated efficient mixing capabilities. Whitesides
non-communicable diseases (e.g. PSA as a marker for prostate and colleagues created a device based on splitting and re-
cancer) (Table 1). Immunoassays are routinely used, with mixing to dilute a sample over a range of 210 (or y1000-fold),
high sensitivity and specificity, to detect and quantitate thus circumventing off-chip pipetting steps.49 Also,
protein markers. The most commonly used samples from Delamarche and colleagues developed a micromosaic immuno-
the patient are whole blood, serum, and plasma, with less assay that significantly reduced the number of pipetting steps
common samples being saliva, urine, feces, sperm, tears and required for a multiplexed assay;64 this method was used to
sweat. In developing countries, the use of fluids that can be develop a panel test for cardiac markers (C-reactive protein,
sampled non-invasively can encourage adoption of tests and myoglobin, and cardiac troponin I).65
decrease the incidence of infection due to contaminated We and Whitesides reported a simple and reliable technique
needles. For example, saliva and urine offer a simple and safe for storing and delivering a sequence of reagents to a
alternative to blood sampling, but they typically contain microfluidic device to carry out automated immunoassays32
lower concentrations of protein makers than in blood; (Fig. 3A). In this method, cartridges made of commercially
successful detection will necessitate an improved sensitivity in available tubing were filled by sequentially injecting plugs
the LOC device. As an example, a LOC immunoassay of reagents separated by air spacers (which prevented the
developed by McDevitt and colleagues to measure levels of reagents from mixing with each other); in this form, the
C-reactive protein in saliva required a 1000-fold improvement reagents were stable to shocks, and antibody solutions could
in sensitivity over ELISA in microtiter plates (by using porous be stored for months without loss in activity. By applying
beads to increase the surface density of capture probe on the negative pressure at the outlet, all reagents were dispensed
solid phase).59 Microfluidic chips to detect enteric antigens sequentially, and a solid-phase immunoassay could be
from human stool, a complex sample matrix, are being completed in 2 minutes with low nM sensitivity. Another
developed.60 attractive method for passive fluid control and delivery is
Enzyme immunoassays, which comprise most protein tests, the autonomous capillary system,45,47 although this
typically require the established infrastructure of centralized method currently requires a multi-step fabrication process.
testing facilities to accomplish complex reagent handling and In contrast to passive delivery, active valves can also be
optical detection. LOC devices have the potential to transpose used to deliver a complex series of pre-stored reagents.
antigen–antibody assays into assay formats that are much less The groups of Beebe33 and Whitesides66 have developed
demanding in infrastructure. At least two important hurdles inexpensive, hand-operated valving systems for point-of-care
exist in the processes of miniaturization and automation: immunoassays. For long-term storage at high temperatures,
storage of multiple reagents and fluid handling capability to storage of the reagents in a dry form may be more stable than
carry out the complex protein assay, and detection of the as wet liquids. Yager and colleagues demonstrated that
signal in the microfluidic system. Below, we review approaches enzymes could be stored dry in LOC devices in mixtures of
that have addressed these challenges using inexpensive and dextran and trehalose.67
portable solutions that are, or have the potential to be, All common fluid handling steps required for protein assays,
compatible with use in resource-poor settings. including actuation and control of fluid flow, have been

48 | Lab Chip, 2007, 7, 41–57 This journal is ß The Royal Society of Chemistry 2007
Fig. 3 Simple and low-cost LOC methods for detecting proteins in developing countries. (A) Optical detection of proteins and reagent storage and
delivery. (i) Schematic representation of the POCKET immunoassay powered by a 9 V battery. (ii) Actual device. (iii) Apparent silver absorbance
values of anti-HIV-1 antibodies from HIV-positive patients and control patients. (iv) Schematic representation of reagent-loaded cartridges. (v)
Overlay of fluorescence and brightfield images of the immunoreaction area, with fluorescent signal corresponding to presence of labeled detection
antibodies on antigen stripes. The concentrations indicated above the picture refer to the concentration of sample tested in each microchannel.
Reprinted from ref. 32 with permission from ACS Publications. (B) Immunomagnetic separation and detection of proteins with CMOS Hall
sensors. (i) Schematic representation with inset showing actual chip. (ii) Comparison of the outputs of CMOS chip and ELISA. Reprinted from
ref. 78 with permission from Elsevier.

demonstrated on centrifugal microfluidic devices.68,69 For products that would be generated in a continuous-flow format
example, the Bioaffy chip from the company Gyros can for ELISA (Fig. 3A). Importantly for use in developing
quantify clinical markers such as a-fetoprotein, interleukin-6 countries, since the silver signal developed over an area of
and carcinoembryonic antigen down to low pM concentra- 2 6 2 mm2, an inexpensive custom-built detector (for less than
tions.70 Also, Bernard and colleagues used a common $45 in components, including a laser diode and a photo-
compact-disc player as a digital detector of silver-based signals detector) can be used to detect the signal, thereby circumvent-
of C-reactive protein in immunoassays;71 the simplicity of this ing the need for expensive optical instrumentation for
overall format makes it attractive for use in resource-poor detecting signals confined to small microchannels.
settings.72 For high-sensitivity assays, Mirkin and co-workers have
reported an assay based on DNA-coated nanoparticles,
(3) Signal detection. Silver dots produced by autometallo- magnetic separation, and PCR to detect protein markers at
graphic precipitation are also directly detectable by optical up to 6 orders of magnitude in improvement in the limit of
density measurements, as illustrated by the microfluidic detection compared to conventional assays.73 This format
POCKET assay as developed by ourselves and Whitesides can in principle be adapted to a low-cost LOC device. High-
and co-workers,44 as well as by Mirkin and colleagues using a sensitivity and quantitative assays can greatly benefit certain
scanometric reader.50 Since amplification resulted in a signal applications, including the measurement of levels of HIV-1
that was immobilized on a surface, the POCKET assay p24, a viral protein antigen which has been advocated as a
avoided washing away optically active and freely diffusible potential simpler alternative to viral load or CD4 counting for

This journal is ß The Royal Society of Chemistry 2007 Lab Chip, 2007, 7, 41–57 | 49
Fig. 4 LOC methods for detecting nucleic acids that can be adapted for use in developing countries. (A) Integrated nanolitre DNA analysis
device. (i) Schematic representation with two liquid samples and electrophoresis gel present. (ii) Optical micrograph of device. Reprinted with
permission from ref. 90. Copyright 1998 AAAS. (B) Schematic representation of oligonucleotide-conjugated nanoparticles for probing DNA
sequence arrays. Reprinted with permission from ref. 50. Copyright 2000 AAAS.

monitoring AIDS progression in HIV patients and diagnosing 4.3 Nucleic acids
HIV/AIDS in newborns.74
Analysis of nucleic acids offers powerful diagnostic informa-
With appropriate amplification schemes, surface plasmon
tion that complement protein analysis of antigens and
resonance (SPR), which detects at the surface minute changes
antibodies. For example, by analyzing conserved DNA or
in the index of refraction induced by the binding of molecule,
viral RNA sequences, PCR and RT-PCR can be used to
can approach the sensitivity of ELISA.75 The company Texas
specifically detect infectious diseases important in developing
Instruments Sensors & Controls (now renamed Sensata countries (such as HIV/AIDS, hepatitis B and C, and TB).79,80
Technologies) has developed a portable SPR sensor (named For HIV/AIDS, quantitative measurements of RNA levels
Spreeta) for heterogeneous antibody-antigen binding and (based on amplification of the 59-long terminal repeat) provide
solid-phase DNA hybridization; this disposable device is information on the stage of diseases; as such, low-cost methods
designed to be manufacturable in very large quantities.76 The for PCR have been studied for use in developing countries.81–83
device can be integrated with a temperature-controlled As a technology, nucleic acid detection can be very sensitive
instrument that runs on a 12 V battery to detect enterotoxin due to amplification, and specific due to the intrinsic com-
B in urine, milk, and sea water, with a sensitivity in the fM plementarity of the base-pairing interactions. Nevertheless, the
range.77 Currently, a Spreeta evaluation module is available building of an integrated LOC device for detecting nucleic
at $200 per sensor (http://aigproducts.com/surface_plasmon_ acids is typically more challenging than for proteins. Overall,
resonance/spr_evaluation_module.htm). Since the gold-coated there are at least three LOC design issues for nucleic acid
chip could be regenerated up to 80 times before disposal, each detection.
assay can potentially be priced below $1 in the future.77
Boser, Harris, and colleagues developed on a 2.5 6 2.5 mm2 (1) Sample pre-treatment. A general challenge in nucleic acid
CMOS chip an array of Hall sensors to quantify the number of detection is the requirement of processes (such as cell sorting,
magnetic beads associated to immunocomplexes at the surface concentration, and lysis, as well as DNA extraction) to isolate
of the sensor (Fig. 3B).78 The use of magnetic beads facilitated the DNA or RNA of interest from desired cells,84–86 in
removal of unbound antibodies conjugated to magnetic beads, contrast to protein detection in which the analyte is typically
and produced signals at the surface that were recorded by the free-floating in the blood, saliva, or urine sample. These steps
Hall sensors. The average reading from 120 sensors was of sample pre-treatment can take place off the chip, but for use
sufficient to quantify dengue fever antibodies in clinical serum in remote settings with untrained users, it is ideal to integrate
samples with a good correlation compared to ELISA assays. these steps seamlessly with the rest of the microfluidic
When combined with proper fluidic control, this method can processing steps.87 The challenge in developing countries is
potentially be developed into an integrated simple and low- even greater since these procedures must be performed at
cost LOC device for immunoassays. low cost.

50 | Lab Chip, 2007, 7, 41–57 This journal is ß The Royal Society of Chemistry 2007
A number of groups have successfully integrated sample cavity to yield comparable performance to conventional
pre-treatment with analysis.84,87 For example, Quake’s group PCR;98 because this system requires only a single heating
used valves to automate cell isolation, cell lysis, nucleic acid element held at a fixed temperature, it can potentially be made
purification, and analysis on the same microchip.86 Integration low cost, although design challenges exist in adapting the
of sample pre-treatment with analysis is important to achieve system to a LOC device.84
ease of use, as well as to improve sensitivity by reducing
sample losses in between steps. For example, integration of Isothermal. The need for temperature cycling in PCR has
cell capture, cell lysis, mRNA purification, cDNA synthesis, made it challenging to build low-cost and simple devices
and cDNA purification has been demonstrated for a RT-PCR suitable for point-of-care testing. An exciting development that
microfluidic chip.88 Grodzinski and co-workers have bypasses thermocycling is isothermal DNA amplification,
developed a self-contained device in plastic that integrates which includes techniques such as single-strand displacement
sample preparation, cell capture, cell preconcentration amplification, rolling circle amplification, and ligase chain
and purification, cell lysis, PCR, DNA hybridization, and reaction.84 Harrison and co-workers have integrated isother-
electrochemical detection to analyze DNA from pathogenic mal amplification in an electrokinetic LOC device that used
bacteria.89 In addition, metering samples is important to cycling probe technology to amplify a DNA sequence from
automate sample preparation,90 an important consideration in S. aureus.99 More recently, Zhang and co-workers demon-
remote settings. strated loop-mediated isothermal amplification (LAMP) in a
cross-shaped microfluidic system in PMMA.100 Also,
(2) Method of signal amplification. PCR/RT-PCR. Gulliksen and colleagues used an isothermal amplification
Miniaturizing PCR on LOC devices has the potential to method in a microfluidic device made of cyclic olefin
reduce the cost of reagents, speed up analysis, and automate copolymer for multiplexed detection of human papilloma
the procedure for use in remote settings by integrating multiple virus at-the-point-of-care application.101 In the future, other
functionalities such as cell concentration and lysis, DNA schemes (such as helicase-dependent DNA amplification102)
extraction, removal of PCR inhibitors, amplification of DNA, may be integrated into LOC devices.
and separation and detection of the amplified products of
interest.84 In the most straightforward adaptation of conven- (3) Detection. The most popular method for analyzing DNA
tional PCR into LOC devices, a microchamber or microwell after amplification is gel electrophoresis of fluorescently
can be created in which the sample and PCR reaction mixture labeled DNA. In LOC devices, DNA analysis by capillary
are thermally cycled (Fig. 4A). In one of the first studies by electrophoresis may be suitable for centralized testing centers
Burns and colleagues (which featured microfluidic channels, in which power supplies and external fluorescence detectors
mixers, heaters, temperature sensors, and fluorescence detec- are available. For remote testing, molecular beacons, which
tors90), the low voltages and power suggested that hand-held are single-stranded oligonucleotides whose fluorescence is
battery operation is feasible; this technology is now being dequenched upon hybridization to a target probe, are a
commercialized by the company HandyLab. Although promising technology for use in developing countries due to
directed more for biodefense than global health, the company their high sensitivity and specificity.103,104 For example, they
Cepheid has developed a miniature analytical thermal cycling have been used to detect pathogenic retroviruses,79 and in
instrument (MATCI) that consists of silicon-micromachined LOC devices to screen for the breast cancer gene
reaction chambers with integrated heaters, optical windows, (BRCA1).105,106 The use of molecular beacons, however, still
and diode-based fluorescence detection.22,91 Although the requires an external fluorescence detector.
current size of these instruments may be too large for use in A potentially low-cost and sensitive detection system for
remote testing, they may be appropriate for centralized testing nucleic acids that may be appropriate for resource-poor
centers in developing countries. Similarly, since most LOC settings is oligonucleotide-conjugated nanoparticle probes, as
devices that use well-based PCR require bulky instruments as demonstrated by Mirkin and others (and now commercialized
well as expensive and complex manufacturing, they may be
by the company Nanosphere) (Fig. 4B). Coupled with silver
most appropriate for use in centralized testing centers.92–94 The
reduction amplification, they can be quantitated by an
design of disposable micro-PCR devices on polycarbonate
inexpensive scanometric reader,50 and potentially by a low-
plastic95 may ultimately be suitable for use in resource-poor
cost and portable reader.44 These nanoparticles, when reduced
settings, although it currently lacks extensive integration.
to form a micron-sized metallic bridge across an electrode gap,
In contrast to well-based LOC PCR, continuous-flow
can also result in quantifiable changes in conductivity,73 which
PCR systems operate by passing a sample continuously over
can in principle be measured by a low-cost and portable
regions of different temperatures. Continuous-flow systems
conductivity meter. Other electrochemistry-based methods for
offer flexible design geometry (for changing the number of
detecting DNA have been reported.107 Simple detection of
amplification cycles), and fast transition times for heating and
amplified nucleic acid products with a dipstick method for
cooling (which depend on the flow rate and kinetics for
resource-poor settings has been demonstrated.60,108
reaching thermal equilibrium).84,96 Another design that is
potentially simple to manufacture and simple to use include
4.4 Cells
passive reactors in closed-loop designs that operate without
valves.97 An interesting novel scheme for PCR amplification Analysis and counting of cells are important for diseases such
takes advantage of convective flow inside a Rayleigh–Bénard as anaemia and hematology (via erythrocyte and complete

This journal is ß The Royal Society of Chemistry 2007 Lab Chip, 2007, 7, 41–57 | 51
blood counts), as well as for monitoring the progression of subsequently be used to sort the cells.111 These devices,
AIDS. Flow cytometry, the current standard for cell analysis however, have yet to be made portable and inexpensive.
and counting, can measure up to 10 or more cell properties A different approach to count cells takes advantage of the
and separate and isolate cells at rates up to 10 000 cells per relatively large size of cells (compared to the scale of
second without loss of viability.109 Since conventional flow microfabrication techniques) by capturing them for analysis.
cytometers are bulky, expensive, and mechanically complex, For example, in an initial study based on the capture of
they are currently limited to well-financed centralized testing microbeads, McDevitt and colleagues micromachined arrays
centers. of pyramidal cavities on silicon wafers; these cavities housed
Due mainly to the importance of counting CD4+ lympho- microspheres that produced optical changes in the presence of
cytes for monitoring the progression of AIDS, a number of analytes.112 Subsequently, Rodriguez, Walker, and colleagues
initiatives have started to support the development of an adapted this device (by adding a polycarbonate, track-etch
inexpensive and compact device for cell counting for global filter that selectively trapped lymphocytes and not red blood
health. In one non-LOC method, Mwaba and colleagues used cells) to capture and measure the levels of CD4+ lymphocytes
filter papers to store dried blood samples, which were from blood samples in Botswana (Fig. 5).113 The results were
transported to a centralized facility for ELISA testing using in good agreement with those obtained with a conventional
anti-CD4 antibodies.110 The results of this simple technology flow cytometer. This device (now being commercialized by the
were encouraging but exhibited limitations in accuracy. With company LabNow) is potentially cheaper than other available
support from the Gates Foundation, Imperial College London systems for single-purpose flow cytometry114 and microbead
is supporting the development of a simple, low-cost, and semi- separation; because of the cost, bulkiness, and power
quantitative CD4+ lymphocyte-counting device (http:// requirements of an epifluorescence microscope and a camera,
www1.imperial.ac.uk/medicine/about/divisions/medicine/infec- however, the current system is likely most suitable for
tious_diseases/cd4_initiative/) that exhibits cut-offs at 200, 350, centralized testing centers rather than remote point-of-care
500 cells mm23 with 10% coefficient of variation. Perhaps testing. Other inexpensive flow cytometric methods have been
more so than simple membrane-based tests, LOC devices have developed, including capabilities for multiplexing.115
the potential to meet these targets due to their increased A third approach for capturing and counting cells is
versatility in design and enhanced analytical performance. immunomagnetic separation, which typically uses antibody-
conjugated superparamagnetic beads to isolate the cells of
Fluid control for design of mechanism of cell sorting. There interest. Tibbe and co-workers used antibody-labeled ferro-
are different methods for designing a LOC device for cell magnetic nanoparticles to perform a differential white blood
counting, all of which require different methods for fluid cell count (with analysis of neutrophils, lymphocytes, mono-
control. For example, conventional flow cytometers use a cytes, and eosinophils).116 In a magnetic field, immuno-
sheath fluid to enclose a stream of cells such that the cells pass magnetic cells are aligned in a capillary along the magnetic
by the detector one by one in a single file for analysis. In a field lines for fluorescent analysis. This design functions with
LOC device, microfluidic mechanisms such as hydrodynamic potentially simple fluidic control, but still requires a conven-
flow switching, electrokinetic flow switching, dielectro- tional fluorescence imaging system.
phoresis, and electrowetting-assisted flow switching can be Cell counting has important applications beyond the
used to focus the cells into a stream.109 Valves can monitoring of HIV/AIDS progression. For example,

Fig. 5 A low-cost LOC device for counting CD4+ lymphocytes. (A) Schematic representation of pyramidal wells in Si. (B) Scanning electron
micrograph of single well with microbead. Reprinted from ref. 112 with permission from ACS publications. (C) Schematic representation of the
device system. (D) (Left) Transmission image of membrane flow cell showing selective capture of lymphocytes. Holes are 3 mm in diameter. (Right)
Fluorescent antibody staining of lymphocytes. (E) Results of cell counting from microchip versus flow cytometry. Reprinted from ref. 113.

52 | Lab Chip, 2007, 7, 41–57 This journal is ß The Royal Society of Chemistry 2007
Fig. 6 Integrated LOC devices with potential for use in developing countries. (A) Schematic representation of LOC for detecting enteric diseases.
Reprinted from ref. 60 with permission from the International Society for Optical Engineering. (B) Picture of a plastic LOC device for point-of-care
clinical diagnostics. Reprinted from ref. 139 with permission from the Proceedings of the IEEE.

McDevitt and colleagues adapted their microbead system to area of research in this field is potentiometric sensing using
build a multifunctional LOC device for performing leukocyte ion-selective field-effect transistors (ISFET); ISFETs, however
counts and measuring C-reactive protein levels, two important often require a large reference electrode.27 Nevertheless,
indicators of coronary heart disease.117 Like the CD4+- integration of electrochemical detection and semiconductor
lymphocyte counting device, this device may be useful in technologies have resulted in commercial products, such as the
centralized testing centers in developing countries (in this case, iSTAT from Abbott Diagnostics.20 This device is a portable
to address the increasing incidences of cardiovascular disease, blood analyzer that uses microfabricated thin-film electrodes
which constitutes 9.5% of total DALYs in developing to measure levels of electrolytes (Na+, K+, Cl2, Ca2+), general
countries). In a different application, Chiu and colleagues chemistries (pH, urea, glucose), blood gases (pCO2, pO2),
used a microfluidic device to show decreased deformability in and hematology (hematocrit). The electrochemical detection
erythrocytes when infected with P. falciparum.118 Although system includes amperometry, voltammetry, and conductance,
this study did not focus on a diagnostic application, it suggests depending on the analyte.
a potential route for diagnosing and monitoring malaria Despite this important achievement, there are limitations of
infection using a LOC device. For example, in a study by MEMS devices that feature electrochemical detection. The
Gascoyne and colleagues in Thailand using dielectrophoresis lack of suitable manufacturing facilities makes it expensive
and field-flow fractionation, parasitized erythrocytes eluted and, for some devices, impractical to scale up the manufactur-
more quickly than normal erythrocytes.119 ing of the sensor. Although it is in principle possible to
Another promising cell-based application of LOC devices leverage existing microelectronics fabrication facilities for the
for global health is the miniaturization of microbiological manufacturing of biological and chemical sensors, there exist a
culture assays. Currently, conventional microbiological number of challenging issues, such as differences in dimensions
techniques are used to identify drug-resistant bacterial strains; (below 1 mm for microelectronics and above 5 mm for sensors),
this identification is critical for administering efficacious in thickness and type of gate insulator, in materials (e.g. for
therapy for TB patients. Techniques in culturing bacteria conductors, Al, polysilicon, and Cu for electronics, versus
in microfluidic chips120–122 can potentially automate this Au and Pt in sensors), and in the passivation layer (e.g. high-
laborious process (and other microbiological assays involving density silicon nitride for sensors that are exposed to
differential cultures) even in remote settings. Devices that solutions).27,125,126 Although their current design still relied
culture and analyze pathogens and cells in microfluidic devices on a bulky instrument for fluid actuation and detection,
could moreover be adapted to perform diagnostic procedures Madou, Bachas and colleagues developed a LOC device that
that are conventionally microscope-based (such as blood measured electrolyte levels optically using optodes;127 in the
smears for diagnosis of malaria). Low-cost microscopy future, it may be possible to design a low-cost, integrated,
using optofluidics, if it can be made robust, may be an optical device for electrolyte measurements.128
attractive technique in resource-poor settings.123 The power of
LOC technologies can potentially be augmented with genetic 4.6 Non-diagnostics applications of LOC devices in developing
and molecular biological approaches, such as Jacobs’ clever countries
luminometric technology for low-cost TB diagnosis using
Although health diagnostics is the nearest-term application of
luciferase-reporter phage124 (now commercialized by the
LOC devices in developing countries, other important areas of
company Sequella).
global health can benefit in the future from this technology.
For example, LOC diagnostic devices can be used for
4.5 Clinical chemistry
environmental sensing and monitoring.60 Sandia Labs, for
Currently, the most popular LOC technologies for analyzing example, is developing a portable device to monitor water
electrolytes are based on electrochemical detection. An active quality by detecting pathogenic bacteria and toxins.129

This journal is ß The Royal Society of Chemistry 2007 Lab Chip, 2007, 7, 41–57 | 53
Table 3 Sample list of current commercial ventures that have the potential to develop LOC devices for developing countries
Company Website Focus Funding source

Cepheid http://www.cepheid.com/ DNA (TB) FIND, US gov.


Claros Diagnostics http://www.clarosdx.com/ Proteins Private
HandyLab http://www.handylab.com/ DNA, proteins NIST, private
iStat http://www.istat.com/ Clinical chemistry markers Private
LabNow http://www.labnow.com/ CD4 for HIV/AIDS George Soros, private
Micronics http://www.micronics.net/ Enteric disease pathogens PATH, University of Washington,
NIH, Gates Foundation
Nanogen http://www.nanogen.com/ Cardiac biomarkers, DNA/RNA Private
Nanosphere http://www.nanosphere-inc.com/ DNA, proteins NIAID, NIH, private
Sensata http://www.sensata.com/ Proteins, viruses, bacteria Private
Sequella http://www.sequella.com/ Proteins (TB) Private

In addition to diagnostics and sensors, LOC technologies moderate resources (such as point-of-care health testing,
can be used to promote the development of therapeutic military sensors, and extraterrestrial devices), and through
compounds. The use of LOC technologies (through high- advances in the young but growing field of LOC devices for
throughput drug screening, genomics, and proteomics) for developing countries.
drug discovery has been reviewed in detail elsewhere;130 as What will be the roadmap in the near future for designing
research into drug discovery for neglected diseases increases and deploying LOC devices in developing countries? First and
(in universities and industrial companies such as the Novartis foremost, new devices will be needed (Fig. 6); the design
Institute for Tropical Diseases), LOC technologies can play criteria of these devices are vast, demanding, and context-
an increasingly important role. More broadly, Singer and dependent, and they will need to be considered carefully
colleagues have investigated the potential contribution of from the early stages of development. Beyond the scientific
biotechnology, genomics, and nanotechnology to address challenges, successful deployment of the device in developing
issues in global health and development;1,131,132 LOC techno- countries will involve a complex interplay of political and
logy has a clear potential to address these issues through its socioeconomic considerations.136 Scientists will therefore need
close association with all these research fields. to work closely with members from NGOs and local
Even when drugs (and vaccines) are available, simple governments as early as possible (and well before a field-
needle-free delivery into the patient is an important challenge testable device is ready) to ensure proper distribution channels
in developing countries.2 Current approaches for improving (Table 4 and Table 5). Since much of the work in engineering
drug delivery in developing countries include inhalation and and development will best take place in the industrial rather
oral delivery, and are a focus of a non-profit organization than academic setting, the private sector has a significant role
(Medicine in Need) based on the work of Edwards et al.133 to play as well (Table 3). As an example of the importance of
Langer and colleagues have developed LOC devices for such multidisciplinary efforts, the Bill and Melinda Gates
programmed time-release of drugs.134,135 In the long term, Foundation is supporting a consortium of academic
these technologies can conceivably be combined with other researchers (Yager and co-workers), industry (Micronics,
miniaturized medical devices (such as wireless capsule endo- Nanogen, and Invetech), and an NGO (PATH) to develop a
scope) for use in health centers without large infrastructure. multifunctional LOC device for infectious diseases137 (Fig. 6A),
and another consortium of academic researchers (Kelso and
co-workers) and industry (Abbott and Inverness Medical
5 Conclusion
Innovations) to develop a low-cost diagnostic device.
There is a pressing need for new health technologies for Finally, potential obstacles can be anticipated from studies
diagnosing and treating communicable and non-communic- on the introduction and dissemination of LOC devices in
able diseases in developing countries. The LOC field is well- developed countries; these obstacles include cost per test
positioned to contribute to this challenge by leveraging recent and reliability (see the market study FlowMap, available at
advances in integrated devices for use in settings with low or http://www.microfluidics-roadmap.com/).

Table 4 Sample list of NGOs focusing on the development of new diagnostics for global health
NGOs Website Focus Year Founded

FIND http://www.finddiagnostics.org/ TB 2003


PATH http://www.path.org/ Diarrheal diseases, malaria, STIs, 1977
AIDS, and cervical cancer
Tuberculosis Diagnostics Initiative http://www.who.int/tdr/diseases/tb/tbdi.htm TB 1996
WHO Malaria Rapid Diagnostics Tests http://www.wpro.who.int/sites/rdt Malaria 2005
WHO Sexually Transmitted Diseases http://www.who.int/std_diagnostics/ Chlamydia, gonorrhea, syphilis 2001
Diagnostic Initiative
WHO/TDR http://www.who.int/tdr/ TB, AIDS, malaria, STIsa 1975
a
Abbreviation: sexually transmitted infections (STIs).

54 | Lab Chip, 2007, 7, 41–57 This journal is ß The Royal Society of Chemistry 2007
Table 5 Sample list of organizations that sell or provide low-cost diagnostic instruments to developing countries
Organization Website Focus

Binax, Inc. http://www.binax.com/ ICTsa for malaria, influenza, filariasis, pneumonia, strep A
Gede Foundation http://www.gedefoundation.org/ AIDS-related diagnostics (flow cytometry, real-time PCR, hematology
analyzer, genotyping services)
Human GmbH http://www.human.de/ Various (clinical chemistry and hematology analyzer, microscopy, ELISA)
Partec http://www.partec.de/ Flow cytometry
Sustainable Sciences http://www.ssilink.org/ Various (low-cost PCR, antibody-detection)
Initiative
a
Abbreviation: immunochromatographic tests (ICTs).

In the long term, miniaturization of medical technologies 18 J. L. Deen, T. Vos, S. R. Huttly and J. Tulloch, Bull. W. H. O.,
1999, 77, 518–524.
has the potential to improve public health, and perhaps even
19 S. N. Forjuoh and G. Li, Soc. Sci. Med, 1996, 43, 1551–1560.
change the basic methods by which patients are diagnosed and 20 I. R. Lauks, Acc. Chem. Res., 1998, 31, 317–324.
treated, in developing countries. This pathway has a well- 21 A. J. Tudos, G. A. J. Besselink and R. B. M. Schasfoort, Lab
known precedent in information and communication tech- Chip, 2001, 1, 83–95.
22 P. Belgrader, S. Young, B. Yuan, M. Primeau, L. A. Christel,
nologies. Over the last 10 years in developing countries,
F. Pourahmadi and M. A. Northrup, Anal. Chem., 2001, 73,
adoption of cell phones and wireless internet has resulted in 391–391.
‘leapfrogging’ over conventional communication technologies 23 C. T. Culbertson, Y. Tugnawat, A. R. Meyer, G. T. Roman,
(such as landlines) that require significant infrastructure.52 In J. M. Ramsey and S. R. Gonda, Anal. Chem., 2005, 77,
7933–7940.
the next 10 years, will LOC methods follow a similar path, by
24 A. M. Skelley, J. R. Scherer, A. D. Aubrey, W. H. Grover,
promoting in developing countries a leapfrog over conven- R. H. C. Ivester, P. Ehrenfreund, F. J. Grunthaner, J. L. Bada
tional medical technologies (such as radiology, microbiological and R. A. Mathies, Proc. Natl. Acad. Sci. U. S. A., 2005, 102,
culture, and centralized testing centers)? 1041–1046.
25 T. Akiyama, S. Gautsch, N. F. de Rooij, U. Staufer,
P. Niedermann, L. Howald, D. Muller, A. Tonin, H. R. Hidber,
Acknowledgements W. T. Pike and M. H. Hecht, Sens. Actuators, A, 2001, 91,
321–325.
We acknowledge Roberto Delatour from Medecins Sans 26 H. Becker and C. Gartner, Electrophoresis, 2000, 21, 12–26.
Frontieres and George Whitesides for helpful discussions, 27 J. Janata, Proc. IEEE, 2003, 91, 864–869.
28 A. J. Ricco, T. D. Boone, Z. H. Fan, I. Gibbons, T. Matray,
Benjamin Wang for help on the figures, and Andreas Martinez S. Singh, H. Tan, T. Tian and S. J. Williams, Biochem. Soc.
for help in compiling Table 1. This work was supported by an Trans., 2002, 30, 73–78.
Early Career Award from the Wallace H. Coulter Foundation. 29 A. de Mello, Lab Chip, 2002, 2, 31N–36N.
30 K. R. Hawkins and P. Yager, Lab Chip, 2003, 3, 248–252.
31 E. Garcia, J. R. Kirkham, A. V. Hatch, K. R. Hawkins and
References P. Yager, Lab Chip, 2004, 4, 78–82.
32 V. Linder, S. K. Sia and G. M. Whitesides, Anal. Chem., 2005, 77,
1 A. S. Daar, H. Thorsteinsdottir, D. K. Martin, A. C. Smith,
64–71.
S. Nast and P. A. Singer, Nat. Genet., 2002, 32, 229–232.
33 J. Moorthy, G. A. Mensing, D. Kim, S Mohanty, D. T. Eddington,
2 H. Varmus, R. Klausner, E. Zerhouni, T. Acharya, A. S. Daar
W. H. Tepp, E. A. Johnson and D. J. Beebe, Electrophoresis,
and P. A. Singer, Science, 2003, 302, 398–399.
2004, 25, 1705–1713.
3 A. Alwan and B. Modell, Nat. Rev. Genet., 2003, 4, 61–68.
4 J. Sachs, Macroeconomics and Health: Investing in Health for 34 Y. Li, P. Denny, C. M. Ho, C. Montemagno, W. Shi, F. Qi,
Economic Development, World Health Organization, Geneva, B. Wu, L. Wolinsky and D. T. Wong, Adv. Dent. Res., 2005, 18,
Switzerland, 2001. 3–5.
5 R. Laxminarayan, A. J. Mills, J. G. Breman, A. R. Measham, 35 V. Srinivasan, V. K. Pamula and R. B. Fair, Lab Chip, 2004, 4,
G. Alleyne, M. Claeson, P. Jha, P. Musgrove, J. Chow, S. Shahid- 310–315.
Salles and D. T. Jamison, Lancet, 2006, 367, 1193–1208. 36 A. J. de Mello and N. Beard, Lab Chip, 2003, 3, 11N–19N.
6 B. H. Robertson and J. K. A. Nicholson, Annu. Rev. Public 37 S. S. Shevkoplyas, T. Yoshida, L. L. Munn and M. W. Bitensky,
Health, 2005, 26, 281–302. Anal. Chem., 2005, 77, 933–937.
7 C. J. L. Murray and A. D. Lopez, Lancet, 1997, 349, 1498–1504. 38 J. P. Brody and P. Yager, Sens. Actuators, A, 1997, 58, 13–18.
8 C. J. L. Murray and A. D. Lopez, Lancet, 1997, 349, 1436–1442. 39 S. Song, A. K. Singh, T. J. Shepodd and B. J. Kirby, Anal. Chem.,
9 World Bank World Development Report 1993: Investing in Health, 2004, 76, 2367–2373.
Oxford University Press, New York, NY, 1993. 40 S. Song and A. K. Singh, Anal. Bioanal. Chem., 2006, 384, 41–43.
10 A. Lopez, C. Mathers, M. Ezzati, D. Jamison and C. Murray, 41 R. H. Liu, M. A. Stremler, K. V. Sharp, M. G. Olsen,
Global Burden of Disease and Risk Factors, Oxford University J. G. Santiago, R. J. Adrian, H. Aref and D. J. Beebe,
Press and the World Bank, New York, NY, 2006. J. Microelectromech. Syst., 2000, 9, 190–197.
11 D. H. Molyneux, P. J. Hotez and A. Fenwick, PLoS Med., 2005, 42 T. Rohr, C. Yu, M. H. Davey, F. Svec and J. M. J. Frechet,
2, 1064–1070. Electrophoresis, 2001, 22, 3959–3967.
12 E. Torreele, C. Royce, R. Don, A. M. Sevcsik and S. Croft, PLoS 43 P. Sethu, L. L. Moldawer, M. N. Mindrinos, P. O. Scumpia,
Med., 2006, 3, e282. C. L. Tannahill, J. Wilhelmy, P. A. Efron, B. H. Brownstein,
13 P. J. Hotez, D. H. Molyneux, A. Fenwick, E. Ottesen, R. G. Tompkins and M. Toner, Anal. Chem., 2006, 78,
S. Ehrlich Sachs and J. D. Sachs, PLoS Med., 2006, 3, e102. 5453–5461.
14 D. Mabey, R. W. Peeling, A. Ustianowski and M. D. Perkins, 44 S. K. Sia, V. Linder, B. A. Parviz, A. Siegel and G. M. Whitesides,
Nat. Rev. Microbiol., 2004, 2, 231–240. Angew. Chem., Int. Ed., 2004, 43, 498–502.
15 H. H. Lee and J. P. Allain, Vox Sang., 2004, 87, 176–179. 45 D. Juncker, H. Schmid, U. Drechsler, H. Wolf, M. Wolf,
16 R. Beaglehole and D. Yach, Lancet, 2003, 362, 903–908. B. Michel, N. de Rooij and E. Delamarche, Anal. Chem., 2002,
17 J. C. Seidell, Br. J. Nutr., 2000, 83, S5–S8. 74, 6139–6144.

This journal is ß The Royal Society of Chemistry 2007 Lab Chip, 2007, 7, 41–57 | 55
46 D. L. L. Chen and R. F. Ismagilov, Curr. Opin. Chem. Biol., 2006, 78 T. Aytur, J. Foley, M. Anwar, B. Boser, E. Harris and R. Beatty,
10, 226–231. J. Immunol. Methods, 2006, 314(1–2), 21–29.
47 S. Cesaro-Tadic, G. Dernick, D. Juncker, G. Buurman, 79 J. A. M. Vet, A. R. Majithia, S. A. E. Marras, S. Tyagi, S. Dube,
H. Kropshofer, B. Michel, C. Fattinger and E. Delamarche, B. J. Poiesz and F. R. Kramer, Proc. Natl. Acad. Sci. U. S. A.,
Lab Chip, 2004, 4, 563–569. 1999, 96, 6394–6399.
48 S. K. Sia and G. M. Whitesides, Electrophoresis, 2003, 24, 80 D. Candotti, J. Temple, S. Owusu-Ofori and J. P. Allain, J. Virol.
3563–3576. Methods, 2004, 118, 39–47.
49 X. Jiang, J. M. Ng, A. D. Stroock, S. K. Dertinger and 81 C. Drosten, M. Panning, J. F. Drexler, F. Hansel, C. Pedroso,
G. M. Whitesides, J. Am. Chem. Soc., 2003, 125, 5294–5295. J. Yeats, L. K. de Souza Luna, M. Samuel, B. Liedigk, U. Lippert,
50 T. A. Taton, C. A. Mirkin and R. L. Letsinger, Science, 2000, 289, M. Sturmer, H. W. Doerr, C. Brites and W. Preiser, Clin. Chem.,
1757–1760. 2006, 52, 1258–1266.
51 S. J. Park, T. A. Taton and C. A. Mirkin, Science, 2002, 295, 82 F. Rouet, D. K. Ekouevi, M. L. Chaix, M. Burgard, A. Inwoley,
1503–1506. T. D. Tony, C. Danel, X. Anglaret, V. Leroy, P. Msellati, F. Dabis
52 United Nations Development Programme, Human Development and C. Rouzioux, J. Clin. Microbiol., 2005, 43, 2709–2717.
Report 2001: Making New Technologies Work for Human 83 J. Coloma and E. Harris, Br. Med. J., 2004, 329, 1160–1162.
Development, Oxford University Press, New York, NY, 2001. 84 P. A. Auroux, Y. Koc, A. deMello, A. Manz and P. J. R. Day,
53 S. L. Hart and C. M. Christensen, MIT Sloan Manage. Rev., Lab Chip, 2004, 4, 534–546.
2002, 44, 51–56. 85 L. C. Waters, S. C. Jacobson, N. Kroutchinina, J. Khandurina,
54 M. Usdin, M. Guillerm and P. Chirac, Nature, 2006, 441, R. S. Foote and J. M. Ramsey, Anal. Chem., 1998, 70, 158–162.
283–284. 86 J. W. Hong, V. Studer, G. Hang, W. F. Anderson and S. R. Quake,
55 M. Cheesbrough, District Laboratory Practice in Tropical Nat. Biotechnol., 2004, 22, 435–439.
Countries, Part 1, Cambridge University Press, Cambridge, UK, 87 Y. Huang, E. L. Mather, J. L. Bell and M. Madou, Anal. Bioanal.
2000. Chem., 2002, 372, 49–65.
56 M. Cheesbrough, District Laboratory Practice in Tropical 88 J. S. Marcus, W. F. Anderson and S. R. Quake, Anal. Chem.,
Countries, Part 2, Cambridge University Press, Cambridge, UK, 2006, 78, 3084–3089.
2000. 89 R. H. Liu, J. N. Yang, R. Lenigk, J. Bonanno and P. Grodzinski,
57 D. D. Cunningham, Anal. Chim. Acta, 2001, 429, 1–18. Anal. Chem., 2004, 76, 1824–1831.
58 C. E. Michel, A. W. Solomon, J. P. Magbanua, P. A. Massae, 90 M. A. Burns, B. N. Johnson, S. N. Brahmasandra, K. Handique,
L. Huang, J. Mosha, S. K. West, E. C. Nadala, R. Bailey, J. R. Webster, M. Krishnan, T. S. Sammarco, P. M. Man,
C. Wisniewski, D. C. Mabey and H. H. Lee, Lancet, 2006, 367, D. Jones, D. Heldsinger, C. H. Mastrangelo and D. T. Burke,
1585–1590. Science, 1998, 282, 484–487.
59 N. Christodoulides, S. Mohanty, C. S. Miller, M. C. Langub, 91 M. A. Northrup, B. Benett, D. Hadley, P. Landre, S. Lehew,
P. N. Floriano, P. Dharshan, M. F. Ali, B. Bernard, J. Richards and P. Stratton, Anal. Chem., 1998, 70, 918–922.
D. Romanovicz, E. Anslyn, P. C. Fox and J. T. McDevitt, Lab 92 J. Y. Lee, J. J. Kim and T. H. Park, Biotechnol. Bioprocess Eng.,
Chip, 2005, 5, 261–269. 2003, 8, 213–220.
60 B. H. Weigl, J. Gerdes, P. Tarr, P. Yager, L. Dillman, R. Peck, 93 J. S. Marcus, W. F. Anderson and S. R. Quake, Anal. Chem.,
S. Ramachandran, M. Lemba, M. Kokoris, M. Nabavi, 2006, 78, 956–958.
F. Battrell, D. Hoekstra, E. J. Klein and D. M. Denno, Proc. 94 E. T. Lagally, J. R. Scherer, R. G. Blazej, N. M. Toriello,
SPIE—Int. Soc. Opt. Eng., 2006, 6112, 1–11. B. A. Diep, M. Ramchandani, G. F. Sensabaugh, L. W. Riley and
61 A. D. Stroock, S. K. W. Dertinger, A. Ajdari, I. Mezic, R. A. Mathies, Anal. Chem., 2004, 76, 3162–3170.
H. A. Stone and G. M. Whitesides, Science, 2002, 295, 647–651. 95 J. N. Yang, Y. J. Liu, C. B. Rauch, R. L. Stevens, R. H. Liu,
62 P. Garstecki, J. F. M, M. A. Fischbach, S. K. Sia and R. Lenigk and P. Grodzinski, Lab Chip, 2002, 2, 179–187.
G. M. Whitesides, Lab Chip, 2006, 6, 207–212. 96 M. U. Kopp, A. J. Mello and A. Manz, Science, 1998, 280,
63 A. P. Sudarsan and V. M. Ugaz, Proc. Natl. Acad. Sci. U. S. A., 1046–1048.
2006, 103, 7228–7233. 97 Z. Chen, S. Qian, W. R. Abrams, D. Malamud and H. H. Bau,
64 A. Bernard, B. Michel and E. Delamarche, Anal. Chem., 2001, 73, Anal. Chem., 2004, 76, 3707–3715.
8–12. 98 M. Krishnan, V. M. Ugaz and M. A. Burns, Science, 2002, 298,
65 M. Wolf, D. Juncker, B. Michel, P. Hunziker and E. Delamarche, 793–793.
Biosens. Bioelectron., 2004, 19, 1193–1202. 99 T. Tang, M. Y. Badal, G. Ocvirk, W. E. Lee, D. E. Bader,
66 D. B. Weibel, M. Kruithof, S. Potenta, S. K. Sia, A. Lee and F. Bekkaoui and D. J. Harrison, Anal. Chem., 2002, 74,
G. M. Whitesides, Anal. Chem., 2005, 77, 4726–4733. 725–733.
67 E. Garcia, J. R. Kirkham, A. V. Hatch, K. R. Hawkins and 100 Y. Hataoka, L. Zhang, Y. Mori, N. Tomita, T. Notomi and
P. Yager, Lab Chip, 2003, 4, 78–82. Y. Baba, Anal. Chem., 2004, 76, 3689–3693.
68 J. V. Zoval and M. J. Madou, Proc. IEEE, 2004, 92, 140–153. 101 A. Gulliksen, L. A. Solli, K. S. Drese, O. Sorensen, F. Karlsen,
69 M. J. Pugia, G. Blankenstein, R. P. Peters, J. A. Profitt, K. Kadel, H. Rogne, E. Hovig and R. Sirevag, Lab Chip, 2005, 5, 416–420.
T. Willms, R. Sommer, H. H. Kuo and L. S. Schulman, Clin. 102 M. Vincent, Y. Xu and H. M. Kong, EMBO Rep., 2004, 5,
Chem., 2005, 51, 1923–1932. 795–800.
70 N. Honda, U. Lindberg, P. Andersson, S. Hoffman and H. Takei, 103 S. Tyagi and F. R. Kramer, Nat. Biotechnol., 1996, 14, 303–308.
Clin. Chem., 2005, 51, 1955–1961. 104 S. Tyagi, S. A. E. Marras and F. R. Kramer, Nat. Biotechnol.,
71 S. A. Lange, G. Roth, S. Wittemann, T. Lacoste, A. Vetter, 2000, 18, 1191–1196.
J. Grassle, S. Kopta, M. Kolleck, B. Breitinger, M. Wick, 105 M. Culha, D. L. Stokes, G. D. Griffin and T. Vo-Dinh, Biosens.
J. K. Horber, S. Dubel and A. Bernard, Angew. Chem., Int. Ed., Bioelectron., 2004, 19, 1007–1012.
2005, 45, 270–273. 106 M. Culha, D. L. Stokes, G. D. Griffin and T. Vo-Dinh, J. Biomed.
72 F. S. Ligler and J. S. Erickson, Nature, 2006, 440, 159–160. Opt., 2004, 9, 439–443.
73 J. M. Nam, C. S. Thaxton and C. A. Mirkin, Science, 2003, 301, 107 T. G. Drummond, M. G. Hill and J. K. Barton, Nat. Biotechnol.,
1884–1886. 2003, 21, 1192–1199.
74 J. Schupbach, AIDS Rev., 2002, 4, 83–92. 108 M. A. Dineva, D. Candotti, F. Fletcher-Brown, J. P. Allain and
75 J. Homola, S. S. Yee and G. Gauglitz, Sens. Actuators, B, 1999, H. Lee, J. Clin. Microbiol., 2005, 43, 4015–4021.
54, 3–15. 109 D. Huh, W. Gu, Y. Kamotani, J. B. Grotberg and S. Takayama,
76 T. M. Chinowsky, J. G. Quinn, D. U. Bartholomew, R. Kaiser Physiol. Meas., 2005, 26, R73–98.
and J. L. Elkind, Sens. Actuators, B, 2003, 91, 266–274. 110 P. Mwaba, S. Cassol, R. Pilon, C. Chintu, M. Janes, A. Nunn and
77 A. N. Naimushin, S. D. Soelberg, D. K. Nguyen, L. Dunlap, A. Zumla, Lancet, 2003, 362, 1459–1460.
D. Bartholomew, J. Elkind, J. Melendez and C. E. Furlong, 111 A. Y. Fu, H. P. Chou, C. Spence, F. H. Arnold and S. R. Quake,
Biosens. Bioelectron., 2002, 17, 573–584. Anal. Chem., 2002, 74, 2451–2457.

56 | Lab Chip, 2007, 7, 41–57 This journal is ß The Royal Society of Chemistry 2007
112 A. Goodey, J. J. Lavigne, S. M. Savoy, M. D. Rodriguez, 124 M. H. Hazbon, N. Guarin, B. E. Ferro, A. L. Rodriguez,
T. Curey, A. Tsao, G. Simmons, J. Wright, S. J. Yoo, Y. Sohn, L. A. Labrada, R. Tovar, P. F. Riska and W. R. Jacobs, J. Clin.
E. V. Anslyn, J. B. Shear, D. P. Neikirk and J. T. McDevitt, Microbiol., 2003, 41, 4865–4869.
J. Am. Chem. Soc., 2001, 123, 2559–2570. 125 G. T. A. Kovacs, Micromachined Transducers Sourcebook,
113 W. R. Rodriguez, N. Christodoulides, P. N. Floriano, S. Graham, McGraw-Hill, New York, 1998.
S. Mohanty, M. Dixon, M. Hsiang, T. Peter, S. Zavahir, I. Thior, 126 M. Madou, Fundamentals of Microfabrication, CRC Press, Boca
D. Romanovicz, B. Bernard, A. P. Goodey, B. D. Walker and Raton, FL, 1998.
J. T. McDevitt, PLoS Med., 2005, 2, e182. 127 I. H. A. Badr, R. D. Johnson, M. J. Madou and L. G. Bachas,
114 M. Fryland, P. Chaillet, R. Zachariah, A. Barnaba, L. Bonte, Anal. Chem., 2002, 74, 5569–5575.
R. Andereassen, S. Charrondière, R. Teck and O. Didakus, 128 H. Hisamoto, M. Yasuoka and S. Terabe, Anal. Chim. Acta, 2006,
Trans. R. Soc. Trop. Med. Hyg., 2006, 100, 980–985. 556, 164–170.
115 I. V. Jani, G. Janossy, D. W. Brown and F. Mandy, Lancet Infect. 129 B. H. Lapizco-Encinas, B. A. Simmons, E. B. Cummings and
Dis., 2002, 2, 243–250. Y. Fintschenko, Anal. Chem., 2004, 76, 1571–1579.
116 A. G. J. Tibbe, B. G. de Grooth, J. Greve, P. A. Liberti, 130 P. S. Dittrich and A. Manz, Nat. Rev. Drug Discovery, 2006, 5,
G. J. Dolan and L. W. M. M. Terstappen, Cytometry, 2001, 43, 210–218.
31–37. 131 P. A. Singer and A. S. Daar, Science, 2001, 294, 87–89.
117 N. Christodoulides, P. N. Floriano, S. A. Acosta, K. L. M. Ballard, 132 F. Salamanca-Buentello, D. L. Persad, E. B. Court, D. K. Martin,
S. E. Weigum, S. Mohanty, P. Dharshan, D. Romanovicz and A. S. Daar and P. A. Singer, PLoS Med., 2005, 2, 383–386.
J. T. McDevitt, Clin. Chem., 2005, 51, 2391–2395. 133 N. Tsapis, D. Bennett, B. Jackson, D. A. Weitz and D. A.
118 J. P. Shelby, J. White, K. Ganesan, P. K. Rathod and D. T. Chiu, Edwards, Proc. Natl. Acad. Sci. U. S. A., 2002, 99, 12001–12005.
Proc. Natl. Acad. Sci. U. S. A., 2003, 100, 14618–14622. 134 D. A. LaVan, T. McGuire and R. Langer, Nat. Biotechnol., 2003,
119 P. Gascoyne, J. Satayavivad and M. Ruchirawat, Acta Trop., 21, 1184–1191.
2004, 89, 357–369. 135 J. Z. Hilt and N. A. Peppas, Int. J. Pharm., 2005, 306, 15–23.
120 F. K. Balagadde, L. You, C. L. Hansen, F. H. Arnold and 136 E. Rodgers, Diffusion of Innovations, the Free Press, New York,
S. R. Quake, Science, 2005, 309, 137–140. 4th edn, 1995.
121 N. Futai, W. Gu, J. W. Song and S. Takayama, Lab Chip, 2006, 6, 137 P. Yager, T. Edwards, E. Fu, K. Helton, K. Nelson, M. R. Tam
149–154. and B. H. Weigl, Nature, 2006, 442, 412–418.
122 S. Petronis, M. Stangegaard, C. B. Christensen and M. Dufva, 138 Frost & Sullivan Report: United States In Vitro Cancer Diagnostics
Biotechniques, 2006, 40, 368–376. Market, Frost & Sullivan, March 1, 1999.
123 X. Heng, D. Erickson, L. R. Baugh, Z. Yaqoob, P. W. Sternberg, 139 C. H. Ahn, J. W. Choi, G. Beaucage, J. H. Nevin, J. B. Lee,
D. Psaltis and C. Yang, Lab Chip, 2006, 6, 1274. A. Puntambekar and J. Y. Lee, Proc. IEEE, 2004, 92, 154–173.

This journal is ß The Royal Society of Chemistry 2007 Lab Chip, 2007, 7, 41–57 | 57

Das könnte Ihnen auch gefallen