Mobile Health Technologies: Methods and Protocols
Mobile Health Technologies: Methods and Protocols
Avraham Rasooly
Keith E. Herold Editors
Mobile Health
Technologies
Methods and Protocols
METHODS IN M O L E C U L A R B I O LO G Y
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
Avraham Rasooly
National Cancer Institute, Rockville, Maryland, USA
Keith E. Herold
Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
Editors
Avraham Rasooly Keith E. Herold
National Cancer Institute Fischell Department of Bioengineering
Rockville, MD, USA University of Maryland
College Park, MD, USA
v
vi Preface
and pathological tremors and Parkinson’s disease, EEG and sleep disorders, and a mobile
device application for surgery support.
The section on mHealth imaging technologies includes chapters on cervical cancer and
skin cancer analysis and the imaging technologies of microendoscopy and skin lesion
imaging.
We would like to express our thanks and gratitude to the authors whose hard work and
excellent contributions are helping make mHealth technologies more accessible to clini-
cians, engineers, and researchers. We appreciate the authors’ time and especially their
patience during a long and arduous review process.
Our hope is that mHealth will play an instrumental role in improving access to medical
procedures including early detection, diagnostics, and treatment through the development
of new portable and accessible devices, and that this will lead to improved health
technologies.
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 497
Contributors
xi
xii Contributors
Abstract
Here we describe a low-cost mobile device that combines cell-phone and satellite communication
technologies with fluid miniaturization techniques for performing all essential functions of enzyme-linked
immunosorbent assay (ELISA). Disease-specific antigens are immobilized on the microfluidic surface, and
disease specific antibodies are captured on the surface and visualized with silver–gold amplification. The
diagnostic result is automatically determined by the device by measuring the absorbance through the
silver–gold amplification in the microchannel. The results are displayed for the user and are synchronized
to a remote patient record. The overall system aims to be portable, robust, low-power, and fully utilize the
ability of mobile devices for bringing better health care to resource poor areas.
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_1, © Springer Science+Business Media New York 2015
3
4 Tiffany W. Guo et al.
2 Materials
2.3 Results 20. Compact satellite modem (Iridium 9601 SBD Transceiver).
Communication 21. GSM/GPRS module (LinkSprite SM5100B).
3 Methods
3.1 Microfluidic 1. Plastic cassettes (Fig. 1c (Right)) are sonicated for 10 min in
Immunoassay deionized water mixed with detergent, rinsed off with deion-
ized water and dried using N2 gas to remove any dust or par-
3.1.1 Surface
ticulates, which may block the microfluidic channels.
Modification of COC
Cassette 2. Next, 20 μL of each antigen is spotted on top of their corre-
sponding meander zones for physisorption (Fig. 1e). Working
concentration of antigens and positive control are prepared in
carbonate–bicarbonate buffer.
3. Test format for zones: negative control–HIV 1/2–positive
control (see Note 1).
4. Physisorption of antigens and antibodies is carried out in
humid chamber at room temperature for 2 h.
5. After physisorption, droplets are aspirated and each meander
zone is washed two times with 1× PBS to remove any unbound
antigen and dried with N2 gas.
6. The cassettes are sealed by application of a clear adhesive tape
over the entire cassette surface.
7. Channels are filled by flowing through the blocking agent
using negative pressure. Blocking agent is incubated in chan-
nels for 1 h at room temperature. This step is important to
prevent any nonspecific protein adsorption.
8. The channels are cleared and the cassettes are either run imme-
diately or stored in dry chamber at 4 °C.
3.1.2 Reagent Loading 1. Working solution (75× dilution) of patient sample (Sera/
Plasma) is prepared in sample diluent (see Note 2).
2. Working solution of gold conjugated anti hIgG is prepared in
gold diluent.
3. A series of reagents is loaded manually using 1 mL syringe to
draw reagents into PE tubing. Air spacers of ~1.6 μL (0.3 cm)
are used to separate the reagents.
4. For sera/plasma, the reagent sequence (Fig. 1d) was a plug of
diluted sample ~27 μL, four plugs of wash buffer ~1.6 μL each,
8 Tiffany W. Guo et al.
3.1.3 Assay Operation To operate the test, user draws less than one microliter of whole
blood (or 5 cm of diluted sera/plasma) in a capillary tube, con-
nects the tube into a microfluidic cassette, and inserts the cassette
into device; the tube forms a fluidic connection between the pre-
loaded reagents and the detection zones. Sample and reagents are
drawn through the cassette by vacuum generated by a micropump.
The liquid handling module includes a diaphragm micropump
(Fig. 1c) to generate negative pressure, a vacuum regulator to reg-
ulate the vacuum pressure applied to the microfluidic chip, and
two pressure sensors to monitor pressure at the outlet of the pump
and the inlet of the chip in real-time. With the use of pressure sen-
sors, we are able to precisely control the pressure drop and flow
rate in the fluidic chip. The ability to vary flow rate over a wide
range via vacuum pressure affords flexibility in adjusting assay time.
Operating within the range of vacuum pressure in the mChip
device, we found it possible to achieve flow rates of 2.5–20 μL/min
using a hand pump as a substitute. At the optimal pressure, the
running times of the plugs are ~4.5 min for sera/plasma samples,
~2.5 min for the gold anti-human IgG antibody, and ~25 s per
wash. After the final water wash, silver solution A and silver solu-
tion B (composed of silver nitrate and reducing agent, respectively)
is mixed through the mixing point on the cassette before passing
through the meanders. Silver reagents are run continuously for
4 min (see Note 3). The overall assay time should be 20 min or less
(see Note 4).
3.2 Data Acquisition We programmed the device to run silver enhancement for 4 min
and Analysis and measure optical transmittance through each analysis zone at
the beginning and end of the duration of silver enhancement.
At the end of the assay, a silver film develops at the detection zones
according to the amount of analytes captured. The signal detection
module of the mChip device contains four pairs of low-cost, super-
bright red light-emitting diodes (LEDs) and photodetectors and a
microcontroller. Each of the four detection zones on the fluidic
chip is sandwiched between a pair of LED and photodetector. The
silver development determines the amount of red light (660 nm)
that can be transmitted. The intensity of transmitted light mea-
sured by the photodetector is converted from analog signals to
Mobile Device for Resource-Limited Settings 9
æ I ö
OD = - log10 ç ÷
è Io ø
Email:
Diagnostic results:
Perform assay: From: [email protected]
Subject: SBD Msg From Unit: 300034013408780 Name Value
Date: May 26, 2010 4:28:44 PM EDT
satellite To:
Reply-to:
Sam Sia
[email protected]
UNIT_ID 1
1 Attachment, 13 bytes Save Quick Look TEST_ID 1
MOMSN: 0 TEST_DATE 11/18/09
MTMSN: 0
PATIENT_ID 1001
Time of Session (UTC): Wed May 26 20:28:43 2010
Session Status: 00 – Transfer OK decode AGE 36
Message Size (bytes): 13
cellphone 01001da60fd249051e5821cb00
Fig. 2 Representative responses obtained during the data communication step. At the end of each assay, the
optical density of each detection zone was displayed on the LCD screen. Transmission of the diagnostic results
is initiated via the satellite or cell phone network. The former resulted in the receipt of an e-mail to a predes-
ignated e-mail address, with the diagnostic data as attachments. Note that the testing location was also
reported in this e-mail. If the results were transmitted via the cell phone network, 13 characters (as hexadeci-
mals) representing the encoded diagnostic results were received as an SMS message at a predesignated cell
phone number. Finally, the results were decoded into readable values. In this case, observed optical densities
are 0.02, 0.485, 0.52, and 0.459 at the four detection windows respectively [17]
10 Tiffany W. Guo et al.
3.3.2 Messaging Protocol In order to minimize the size of data to be transmitted (and
hence reduce the cost of data transmission), all the data is format-
ted, including date/time of test, patient’s information, and test
results, into a 15-byte binary string. As opposed to sending data
on a character-by-character basis, the messaging format treats
each type of data (mobile device identifier number, date and time
of test, patient identifier number, age, gender, pregnancy status,
absorbance values to three decimal places for each analysis zone,
Mobile Device for Resource-Limited Settings 11
Bit position Bit 119-112 Bit 111-95 Bit 94-83 Bit 82-73 Bit 72-66 Bit 65
Name UNIT_ID TEST_DATE TEST_TIME PATIENT_ID AGE GENDER
Bit 64 Bit 63-54 Bit 53-44 Bit 43-34 Bit 33-24 Bit 23-16 Bit 15-0
PREGNANT RESULT_0 RESULT_1 RESULT_2 RESULT_3 RESERVE CHECK
Fig. 4 Messaging protocol for formatting diagnostic data into a 15-byte binary string to reduce the size of data
to be transmitted. In this study, private fields related to patient’s information (e.g., age, gender, and pregnancy
status) were not used. Reprinted with copyright permission from Clinical Chemistry [18]
3.3.3 Data Confidentiality The data transmission must be secure. As one example, because
in Transmission data is first encoded as a 15-byte binary string before it is transmit-
ted through the satellite network, decoding the information on the
receiver end would require a password-protected lookup table.
Within the data messages, even if they are intercepted and decoded,
patients are identified only by numerical ID’s known only to health
care providers. In the future, if even higher level of data security is
required, Iridium SBD transceivers can transmit data using the
Enhanced Mobile Satellite Services (EMSS). EMSS is a Department
12 Tiffany W. Guo et al.
4 Notes
References
1. Morris K (2009) Mobile phones connecting 8. Willyard C (2010) Focus on electronic health
efforts to tackle infectious disease. Lancet records. Electronic records pose dilemma in
Infect Dis 9:274 developing countries. Nat Med 16:249
2. Lester R, Karanja S (2008) Mobile phones: 9. Fraser HS, Biondich P, Moodley D et al (2005)
exceptional tools for HIV/AIDS, health, and cri- Implementing electronic medical record sys-
sis management. Lancet Infect Dis 8:738–739 tems in developing countries. Inform Prim
3. Zurovac D, Sudoi RK, Akhwale WS et al Care 13:83–95
(2011) The effect of mobile phone text- 10. Nsanzimana S, Ruton H, Lowrance DW et al
message reminders on Kenyan health workers’ (2012) Cell phone-based and internet-based
adherence to malaria treatment guidelines: a monitoring and evaluation of the national
cluster randomised trial. Lancet 378:795–803 antiretroviral treatment program during
4. Pop-Eleches C, Thirumurthy H, Habyarimana rapid scale-up in Rwanda: Tracnet, 2004–
JP et al (2011) Mobile phone technologies 2010. J Acquir Immune Defic Syndr
improve adherence to antiretroviral treatment 59:e17–e23
in a resource-limited setting: a randomized 11. NIH. A summit on mobile technologies as a
controlled trial of text message reminders. platform for health research and healthcare
AIDS 25:825–834 delivery. http://www.fic.nih.gov/news/
5. Lester RT, Ritvo P, Mills EJ et al (2010) events/mhealthsummit.htm. Accessed Nov
Effects of a mobile phone short message ser- 2012
vice on antiretroviral treatment adherence in 12. Schreiner S (2009) Inaugural mhealth summit
Kenya (WelTel Kenya1): a randomised trial. successfully brings together health, technology
Lancet 376:1838–1845 and policy communities to advance techno-
6. Bellina L, Missoni E (2009) Mobile cell- logical innovation in global healthcare.
phones (m-phones) in telemicroscopy: increas- Business Wire. October 30, 2009
ing connectivity of isolated laboratories. Diagn 13. Kost GJ (2002) Part III: Point-of-care testing
Pathol 4:19 in health system, community and field. In:
7. Blaya JA, Cohen T, Rodriguez P et al (2009) Kost GJ (ed) Principles & practice of point-of-
Personal digital assistants to collect tuberculo- care testing. Lippincott Williams & Wilkins,
sis bacteriology data in Peru reduce delays, Baltimore, p 386
errors, and workload, and are acceptable to 14. CDC. Flu activity & surveillance. http://www.
users: cluster randomized controlled trial. Int J cdc.gov/flu/weekly/fluactivitysur v.htm.
Infect Dis 13:410–418 Accessed Nov 2012
14 Tiffany W. Guo et al.
15. Dzenowagis J, Kernen G (2005) Connecting 17. Cheung YK (2010) Fabrication of multi-
for health: global vision, local insight. World component hydrogel microstructures and micro-
Health Organization, Geneva. Accessed Dec devices. Dissertation, Columbia University.
2012 Publication Number: 3447863
16. Chin CD, Laksanasopin T, Cheung YK et al 18. Chin CD, Cheung YK, Laksanasopin T et al
(2011) Microfluidics-based diagnostics of (2013) Mobile device for disease diagnosis and
infectious diseases in the developing world. data tracking in resource-limited settings. Clin
Nat Med 17:1015–1019 Chem. doi:10.1373/clinchem.2012.199596
Chapter 2
Abstract
Molecular (nucleic acid)-based diagnostics tests have many advantages over immunoassays, particularly
with regard to sensitivity and specificity. Most on-site diagnostic tests, however, are immunoassay-based
because conventional nucleic acid-based tests (NATs) require extensive sample processing, trained opera-
tors, and specialized equipment. To make NATs more convenient, especially for point-of-care diagnostics
and on-site testing, a simple plastic microfluidic cassette (“chip”) has been developed for nucleic acid-
based testing of blood, other clinical specimens, food, water, and environmental samples. The chip com-
bines nucleic acid isolation by solid-phase extraction; isothermal enzymatic amplification such as LAMP
(Loop-mediated AMPlification), NASBA (Nucleic Acid Sequence Based Amplification), and RPA
(Recombinase Polymerase Amplification); and real-time optical detection of DNA or RNA analytes. The
microfluidic cassette incorporates an embedded nucleic acid binding membrane in the amplification reac-
tion chamber. Target nucleic acids extracted from a lysate are captured on the membrane and amplified at
a constant incubation temperature. The amplification product, labeled with a fluorophore reporter, is
excited with a LED light source and monitored in situ in real time with a photodiode or a CCD detector
(such as available in a smartphone). For blood analysis, a companion filtration device that separates plasma
from whole blood to provide cell-free samples for virus and bacterial lysis and nucleic acid testing in the
microfluidic chip has also been developed. For HIV virus detection in blood, the microfluidic NAT chip
achieves a sensitivity and specificity that are nearly comparable to conventional benchtop protocols using
spin columns and thermal cyclers.
Key words Point-of-care, On-site, Molecular diagnostics, DNA, RNA, Infectious diseases, Pathogen,
Microfluidics, Enzymatic, Amplification, LAMP, Isothermal
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_2, © Springer Science+Business Media New York 2015
15
16 Michael G. Mauk et al.
Fig. 1 (Left ) Photograph of a chip containing an array of three multifunctional amplification reactors. (Right ) A
cross section of a multifunctional amplification reactor featuring the flow-through membrane for the capture
and immobilization of nucleic acids
2 Materials
3 Methods
3.1 Chip Design The microfluidic chips (Fig. 1) are fabricated in clear (transparent)
and Fabrication plastic materials as bonded laminates. We have tested three differ-
ent thermoplastic polymers: polycarbonate (PC), acrylic (PMMA),
and cyclic olefin copolymer (COC) (Table 1). The chips have been
rapidly prototyped using computer-aided design (CAD) and
computer-aided manufacturing software (SolidWorks™,
MasterCAM™, or AutoCAD™). The 0.1–1-mm thick plastic
sheets are cut with a computer-controlled (CNC) milling machine
(e.g., Haas Office Mill OM-1, Haas Automation, Oxnard, CA) or
with a CO2 laser (e.g., Universal Laser Systems, Scottsdale, AZ,
30–50 W power) to define the microfluidic circuit(s) and other
features (Fig. 2). Chip fabrication and bonding methods for these
materials are discussed in more details in references [55–62]. The
smallest feature size is the channel width, which is ~800 μm. For
acrylic-based chips, the component layers are bonded with a weak
solvent such as acetonitrile. These polymer materials exhibit low
surface adsorption and usually do not require any special surface
treatments; however, dynamic surface passivation with BSA (bovine
serum albumin, 1–2 %), PVP-40 (polyvinylpyrrolidone 40), or
22 Michael G. Mauk et al.
Table 1
Thermoplastic polymer stock for chipsa
Optical
Trade CO2 Laser transparency Autofluore-
Material names cutting (nm) scence Bonding Cost
“Acrylic” Plexiglass®, Yes 300–1,200 Moderate Thermal/ Low
Polymethyl- Lucite®, to high, pressure, solvent
methacrylate Perspex® depending (acetonitrile),
(PMMA) on particular ultrasonic
PMMA welding
Polycarbonate Lexan® No 250–1,200 Moderate to Thermal/pressure, Low
(PC) relatively high solvent (acetone)
Cyclic olefin TOPAS® Yes 200–1,200 Low Thermal/pressure, Moderate
copolymer COC solvent (hexane)
(COC)
a
These materials are available in sheets of thicknesses ranging from 0.1 to 3 mm. One layer of the chip is made from a
PCR sealing film (PCR Sealers™ Microseal B, Bio-Rad, Hercules, CA). This is a PCR-compatible adhesive tape with
good transparency and low autofluorescence
Fig. 2 Microcluidic NAT chip CAD drawings (SolidWorks™) showing assembled, bonded structure and compo-
nent layers. (a) Assembled chip. (b) Top view showing the inlets and outlets and the PMMA tape covering the
reaction chambers. (c) Underside view showing the PCR tape covering the membrane isolation chamber and
conduits. (d) Exploded view. (e) Top view of the main slab showing the uncovered amplification chambers.
(f) Underside view of the main slab (without the PCR sealing tape) showing the isolation chambers with
embedded membranes and the conduits
Microfluidic Devices for Nucleic Acid (NA) Isolation, Isothermal NA… 23
Fig. 3 Experimental Setup for operating microfluidic cassette showing cassette mounted on base supporting
heater and blue LED. Detector or camera is positioned over cassette. Optical filter blocks blue excitation light
to allow detection of green fluorescence from LAMP reaction(s) on cassette. A power/control box provides
power for heater and LED, and auxilliary functions such as data logging and communication with smartphone.
Inset shows photo of real-time LAMP reaction on cassette
3.2 Use of Nucleic The chip features solid-phase extraction to isolate nucleic acids
Acid Binding Phases from samples. The chip described here is based on the method of
for Isolation of Nucleic Boom et al. [68–70] using chaotropic salts to promote selective
Acids binding of NAs to a (silica-based) solid phase, and as adapted by
others [71–77] for microfluidic implementation. Several types of
porous membranes for the nucleic acid (NA) binding phase have
been employed, including “silica” (Whatman™ GF/F borosilicate
glass fiber filter paper, 0.42 mm thick, 0.7 μm pore size) and
cellulose-based membranes (Whatman FTA™ paper: 3MM
Whatman cellulose filter paper, 0.5 mm thick, with dried Tris–HCl,
EDTA, and SDS, which can be washed off the FTA paper before
mounting in the chip). Both these membranes can be readily cut to
the desired shape and size (e.g., 1.5 mm diameter) using a hole
punch (Harris Unicore™, Ted Pella, Inc., Redding, CA), and then
inserted into the chip during its assembly. Other membranes such
as nanoporous alumina (Whatman Nanopore™) can also function
as nucleic acid binding phases, but are difficult to cut to size and
mount in the chip due to their brittleness [52, 53] (see Note 2).
Microfluidic Devices for Nucleic Acid (NA) Isolation, Isothermal NA… 25
Table 2
Reagents and buffers for on-chip isolation of DNA targets (i.e., B. cereus)
Table 3
Reagents and buffers for on-chip isolation of viral RNA targets (i.e., HIV virus)
Qiagen QIAamp™
Reagent step viral RNA kit Roche High Pure™ Viral RNA kit Generic reagents
Lysis/binding AVL buffer (contains Lysis/binding buffer 6 M Guanidine HCl
buffer guanidine thiocyanate) Inhibitor removal buffer (4.5 M or 6 M guanidine
guanidine-HCl, 50 mM Tris– thiocyanate
HCl, 30 % Triton® X-100)
Wash buffer 1 AW1 buffer (contains Inhibitor removal buffer (5 M 50 % ethanol; 50 %
guanidine HCl), guanidine-HCl, 20 mM water
~50 % ethanol Tris–HCl, 40 % (v/v)
ethanol, pH 6.6)
Wash buffer 2 AW2 buffer, ~50 % Wash Buffer (approx. 50 % (v/v) 50 % ethanol; 50 %
ethanol ethanol, 20 mM NaCl, 2 mM water
Tris–HCl, pH 7.5)
Table 4
Isothermal amplification specifications
Incubation
Amplification Reaction mix (25 μl, temperature
method Supplier total volume) (°C) Primers
LAMP (Loop- Eiken Chemical 20 mM Tris–HCl (pH 8.8), 60–65 6 primers:
mediated (Tochigi, Japan) 10 mM KCl, 10 mM (NH2) FIP 1.6 μM
amplification) SO4, 8 mM MgSO4, 0.1 % BIP
Tween 20, 0.8 M betaine, 8 U 1.6 μM
Bst DNA polymerase, 1.4 mM Loop-F
dNTPs, and 4.0 μM SYTO® 9 0.8 μM
Green intercalating dye, 8 U Loop-B
RNase inhibitor 0.8 μM
F3 0.2 μM
B3 0.2 μM
RPA (recombinase TwistDX Rehydration buffer 30 μl 37–40 2 primers:
polymerase (Cambridge, UK) Freeze-dried reaction mix primer A
amplification) Template + H2O to 47.5 μl 10 μM
total volume, 4.0 μM SYTO® primer B
9 Green intercalating dye; 10 μM
Add 2.5 μl of 280 mM MgAc
to initiate reaction, 8 U RNase
inhibitor
NASBA (nucleic Biomérieux 5 μl enzyme mix (RNase H, T7- 55–60 2 primers
acid sequence (Durham, NC) RNA polymerase, reverse primer F
based transcriptase), 10 μl NASBA 0.2 μM
amplification) buffer + 5 μl sample, 4.0 μM primer R
SYTO® 9 Green intercalating 0.2 μM
dye, 8 U RNase inhibitor
Positive Controls and their primers are included in these kits
Reverse transcriptase is added to reaction mix for RNA targets, e.g., 0.63 U AMV reverse transcriptase (Invitrogen,
Carlsbad, CA)
Table 5
A few fluorescent reporters for real-time amplification
Excitation Emission
Dye Mechanism wavelength (nm) wavelength (nm)
Ethidium bromide Intercalation of ds-DNA 300/360 590
Loop-Amp Amplification phosphate by product 240–370 515
Fluorescent binds Mg and induces fluorescence 340 (peak)
(calcein)
SYBR Green I Intercalation of ds-DNA 497 520
SYTO-9 Intercalation of ds-CNA 485 498
EvaGreen™ Hybridization/intercalation 490 530
light into the amplification chamber(s); (3) filtering out the excita-
tion spectra to prevent its detection; and (4) the detection of the
(longer wavelength) light emitted from the reaction chamber with
a detector having appropriate spectral responsivity. Various
approaches to fluorescence detection on chips are feasible [80–85].
To detect the fluorescence signal on the chip, we have used either
(1) a commercial compact fluorometer such as the Qiagen ESE
Fluo-Sens reader (Model ESE Model ESML 10-MB-3007 with
520 nm excitation and silicon photodiode detector); (2) a portable
(palm-sized) fluorescence microscope (DinoLite™ Model
AM4113T-GFBW) with seven built-in blue LEDs for illumination,
an emission filter with a 510-nm wavelength cutoff, a CCD camera
detection, and a USB connection; and (3) a cell phone camera in
conjunction with LED excitation and an appropriate filter. Both the
Qiagen ESE reader and the portable fluorescence microscope have
their own excitation sources and optical filters and are suitable for
detection with SYTO Green, SYBR Green, and EVA Green dyes.
The Qiagen ESE reader can view only one reactor at a time and
must be mounted on a scanner when the chip houses an array of
reactors. In contrast, the USB-based microscope and the CCD
camera can monitor multiple reactors simultaneously and do not
require scanning. Both the USB-based microscope and the smart-
phone camera require imaging software (e.g., ImageJ® or
MATLAB®) to quantify and process the detected signal. Given the
ubiquity of cell phones even in Third World countries, the use of a
cell phone is an attractive option. By taking advantage of mobile
devices, we can reduce the cost of the instrumentation needed for
the diagnostic devices while at the same time providing the ability
of transferring test results wirelessly to a centralized depository for
archiving, analysis, and public health monitoring.
For smart cell phone-based detection, one can illuminate the
edge of the chip with blue light LED or laser. The light is guided
into the reaction chambers through the transparent chip material.
Lensing and waveguide structures can also be formed in the chip
during the microfluidic circuit machining process to attain more
uniform excitation of the reactor array [86]. LEDs with emission
wavelengths of 450–500 nm are needed to excite many of the
common dyes (Table 5). Unfortunately, many nominally “blue”
LEDs have a significant green component that overlaps with the
emission of the fluorophore. To minimize interference from the
excitation source, suitable bandpass and longpass colored glass or
interference coating optical filters (Thor Labs, Newton, NJ or
Edmund Scientific, Barrington, NJ) must be used to block excita-
tion light from the detector. For example, when using blue emis-
sion (450–500 nm), we employed a Thor Labs LED465E filter
(2.5-mm diameter, nominal emission peak at 465 nm with spectral
FWHM, full-width half maximum of 50 nm). Note that the plastic
chip materials are not very transmissive for UV (for dyes such as
Microfluidic Devices for Nucleic Acid (NA) Isolation, Isothermal NA… 29
Fig. 4 Plasma separation device: (a) whole blood inserted into the separator
chamber, (b) sedimentation of blood components, (c) extraction of plasma.
(d) A photograph of a standalone version of the plasma separation device
3.6 Plasma For the analysis of blood samples, plasma is first separated from
Extraction from Whole whole blood. Traditionally, plasma is separated from blood by
Blood Using centrifugation. Centrifugation is, however, inappropriate for on-site
a Filtration Module applications. As an alternative means, we developed a passive, low-
cost, pump-free separator (Fig. 4) that utilizes both sedimentation
and filtration to extract plasma from whole blood [89]. The sedi-
mentation process reduces the concentration of the cells in the
blood that is then filtered through an asymmetric plasma separation
membrane (polysulfone, Vivid™ GR, Pall Corp., Port Washington,
NY). Red cells, white cells, and platelets are trapped without lysis in the
large pores on the upstream side of the membrane. The plasma out-
flows through the smaller pores on the membrane’s downstream side.
The separated plasma is collected with a pipette. The plasma separation
module was tested by extracting about 275 μl of plasma from 1.8 ml of
whole blood with known titer of HIV, in less than 7 min, using only
30 Michael G. Mauk et al.
a pipette to load and retrieve the sample. The plasma was then
subjected to lysis to solubilize target nucleic acids from the virus (or
bacteria) of interest, and processed in our microfluidic chip as
described in the next section.
3.7 Chip Operation We focus on the operation of a microfluidic module that integrates
nucleic acid isolation, amplification, and detection using the multi-
functional amplification reactor described above. For clarity and
brevity, only manual operation of the chip is described. The sample
is lysed off-chip by mixing it with a chaotropic salt (guanidinium
chloride) and other optional lysing agents (e.g., detergents, pro-
teinase K, lysozyme). The lysis deactivates any infectious agents in
the sample as well as nucleases that might degrade target NAs. The
lysate is then pipetted through the membrane and the reaction
chamber (see Note 3). The chaotropic salt promotes binding of the
nucleic acids to the membrane. The membrane-bound nucleic
acids are then washed with buffer to remove debris and inhibitors.
In the normal mode of solid-phase extraction (i.e., spin column),
the captured nucleic acids are eluted from the membrane into the
amplification chamber with a low-salt, pH-neutral buffer. In con-
trast, in our device, we simplify the process by forgoing a separate
elution step. The membrane is sited at the inlet of the amplification
chamber and is in contact with the amplification reaction mixture.
The membrane-bound nucleic acids serve directly as templates for
amplification. An important advantage of this configuration is that
the sample volume (the amount of lysate filtered through the
membrane) is largely decoupled from the volume of the amplifica-
tion reaction. Nevertheless, there is an optimum membrane size
for a given sample volume and amplification chamber size. The
membrane must be large enough to capture a high fraction of the
NA from the perfusing lysate, but not too large as to impede
desorption of captured NA during amplification. Also, the silica-
based membrane material partially inhibits enzymatic amplifica-
tion. The cellulose-based FTA membrane shows a better
compatibility with enzymatic amplification, i.e., less inhibition of
amplification. We speculate that inhibition of amplification by the
membrane material results from irreversible adsorption of tem-
plate, primers, and/or polymerase to the membrane since experi-
ence teaches that inhibition effects are reduced when the
concentrations of these three components are increased in the
reaction mix.
As an example of the chip operation, we describe the detection
of HIV virus in blood. The various processing steps are depicted
schematically in Fig. 5. Plasma separated from whole blood was
subjected to a chemical lysis step using a combination of chaotropic
salts, detergents, and enzymes. Nucleic acids were isolated from the
lysate by solid-phase extraction, and the purified, concentrated,
immobilized nucleic acids were subjected to sequence-specific
Microfluidic Devices for Nucleic Acid (NA) Isolation, Isothermal NA… 31
3.8 Steps for a Blood 1. A blood sample laden with the suspected pathogen (e.g., HIV
Sample Molecular virus), ranging in volume from 100 μl to 1 ml (depending on
Analysis on a Chip the required limit of detection), is inserted into the plasma
separator. The plasma separator utilizes a combination of filtra-
tion and sedimentation to separate the plasma from the blood.
2. Add 4 volumes of lysis/binding buffer to 1 volume of plasma
(e.g., add 400 μl lysing buffer to 100 μl plasma) and incubate
for 10 min at room temperature.
3. Optionally, add carrier RNA suspension (20 ng per sample) to
the lysis buffer.
4. Pipette a volume of lysate (50–500 μl) into the chip, perfusing
the membrane.
5. Pipette 100 μl ethanol-based wash solution through the mem-
brane, washing the membrane.
32 Michael G. Mauk et al.
3.10 Conclusion The microfluidic module and protocol for nucleic acid isolation
and Discussion and amplification described here are a convenient alternative to
benchtop molecular assays. The microfluidic device combines
nucleic acid isolation typically done using commercial spin column
kits with enzymatic amplification and detection as conventionally
done in Eppendorf tubes in a benchtop thermal cycler instrument.
A sample can be processed in the microfluidic chip in less than an
hour. In several applications, such as analysis of an RNA virus in
blood plasma, limits of detection comparable to benchtop assays
(<1,000 virus particles per ml sample) have been realized. This
“lab-on-a-chip” approach is attractive for processing of a small
number of samples. More importantly, the microfluidic module
described here can serve as a component of an autonomous, por-
table, on-site molecular diagnostic system. Such next-generation
devices may incorporate programmed fluidic actuation and flow
control, as well as smartphone detection of amplicons’ fluorescence
emission. Chips, preloaded with lyophilized reagents, primers for a
specified target, and buffers, could be hermetically packaged,
bar-coded, and stored (without refrigeration) with a shelf-life of a
year or more. Chips, customized for various targets, could operate
with the same portable processor. A simple filtration separation
device that extracts plasma samples from whole blood for microflu-
idic nucleic acid testing could be combined with the NAT chip into
34 Michael G. Mauk et al.
4 Notes
Acknowledgments
References
1. Olasagasasti F, Ruiz de Gordoa J (2012) HIV-1 and coinfections. J Clin Microbiol
Miniaturized technology for protein and 49(10):3584–3590
nucleic acid point-of-care testing. Transl Res 15. Lin C-C, Wang J-H, Wu H-W, Lee G-B
160(5):332–345 (2010) Microfluidic immunoassays. J Lab
2. Weigl B, Domingo G, LaBarre P, Gerlach J Autom 15:253–274
(2008) Towards non- and minimally instru- 16. Ngom B, Guo Y, Wang X, Bi D (2010)
mented, microfluidics-based diagnostics Development of lateral flow strip technology
devices. Lab Chip 81:1999–2014 for detection of infectious agents and chemical
3. Wang S, Xu F, Demirci U (2010) Advances in contaminants. Anal Bioanal Chem 397(3):
developing HIV-1 viral load assays for 1113–1135
resource-limited settings. Biotechnol Adv 17. Al-Soud WA, Rådström P (2001) Purification
28:770–781 and characterization of PCR-inhibitory com-
4. Chin CD, Linder V, Sia SK (2012) ponents in blood cells. J Clin Microbiol
Commercialization of microfluidic point-of- 39(2):485–493
care diagnostic devices. Lab Chip 12: 18. Opel KL, Chang D, McCord BR (2010) A
2118–2134 study of PCR inhibition mechanisms using real
5. Myers FB, Henrikson RH, Bone J, Lee LP time PCR. J Forensic Sci 55(1):25–33
(2013) A handheld point-of-care genomic 19. Wiedbrauk DL, Werner JC, Drevon AM
diagnostic system. PLoS One 8(8):e70266 (1995) Inhibition of PCR by aqueous and vit-
6. Giljohann DA, Mirkin CA (2009) Drivers of reous fluids. J Clin Microbiol 33(10):
biodiagnostic development. Nature 462(26): 2643–3646
461–464 20. Burns DE, Ashwood ER, Burtis CA (2007)
7. Gubala V, Harris LF, Ricco AJ, Tan MX, Fundamentals of molecular diagnostics.
Williams DE (2012) Point of care diagnostics: Saunders, St. Louis
status and future. Anal Chem 84(2):487–515 21. Zhang C, Xing D (2007) Minaturized chips
8. Fu E, Yager P, Fioriano PN, Christodoulides for nucleic acid amplification and analysis: lat-
N, McDevitt J (2011) Perspectives on diag- est advances and future trends. Nucleic Acids
nostics for global health. IEEE Pulse 2(6): Res 35(13):4223–4237
40–50 22. Park S, Zhang Y, Lin S, Wang T-H, Yang S
9. Mairfair J, Roppert K, Ertl P (2009) (2011) Advances in microfluidic PCR for
Microfluidic systems for pathogen sensing: a point-of-care infectious disease diagnostics.
review. Sensors 9:4804–4823 Biotechnol Adv 29:830–839
10. Mondal S, Venkataraman V (2007) 23. Lee J-G, Cheong KH, Huh N, Kim S, Choi
Miniaturized devices for DNA amplification J-W, Ki C (2006) Microchip-based one step
and fluorescence based detection. J Indian Inst DNA extraction and real-time PCR in one
Sci 87(3):309–332 chamber for rapid pathogen identification. Lab
11. Yoon J-Y, Kim B (2012) Lab-on-a-chip patho- Chip 6:886–895
gen sensors for food safety. Sensors 12: 24. Wang J, Chen Z, Corstjens PL, Mauk MG,
10713–10741 Bau HH (2006) A disposable microfluidic cas-
12. Tourlousse DM, Ahmad F, Stedtfeld RD, sette for DNA amplification and detection.
Seyrig G, Tiedje JM, Hashham SA (2012) A Lab Chip 6(1):46–53
polymer microfluidic chip for quantitative 25. Chen Z, Mauk MG, Wang J, Abrams WR,
detection of multiple water and foodborne Corstjens PL, Niedbala RS, Malamud D, Bau
pathogens using real-time fluorogenic loop- HH (2007) A microfluidic system for saliva-
mediated isothermal amplification. Biomed based detection of infectious diseases. Ann N Y
Microdevices 14:769–778 Acad Sci 1098:429–436
13. Sakamoto C, Yamaguchi N, Yamada M, 26. Chen D, Mauk M, Qiu X, Liu C, Kim J,
Nagase H, Seki M, Nasu M (2007) Rapid Ramprasad S, Ongagna S, Abrams WR,
quantification of bacterial cells in potable water Malamud D, Corstjens PLAM, Bau HH
using a simplified microfluidic device. J (2010) An integrated, self-contained microflu-
Microbiol Methods 68:643–647 idic cassette for isolation, amplification, and
14. Lochhead MJ, Todorof K, Delaney M, Ives JT, detection of nucleic acids. Biomed
Greef C, Moll K, Rowley K, Vogel K, Myatt C, Microdevices 12(4):705–719
Zhang X-Q, Logan C, Benson C, Reed S, 27. Qiu X, Mauk MG, Chen D, Liu C, Bau HH
Schooley RT (2011) Rapid multiplexed immu- (2010) A large volume, portable, real-time
noassay for simultaneous serodiagnosis of PCR reactor. Lab Chip 10(22):3170–3177
38 Michael G. Mauk et al.
28. Qiu X, Chen D, Liu C, Mauk MG, Kientz T, 41. Piepenburg O, Williams CH, Stemple DL,
Bau HH (2011) A portable, integrated ana- Ames NA (2006) DNA detection using recom-
lyzer for microfluidic-based molecular analysis. bination proteins. PLoS Biol 4(7):e204
Biomed Microdevices 13:809–817 42. Lutz S, Weber P, Focke M, Faltin B, Hoffman
29. Liu C, Mauk MG, Hart R, Qiu X, Bau HH J, Müller C, Mark D, Roth G, Munday P,
(2011) A self-heating cartridge for molecular Armes N, Piepenberg O, Zengerle R, von
diagnostics. Lab Chip 11:2686–2692 Stetten F (2010) Microfluidic lab-on-foil for
30. Chang C-C, Chen C-C, Wei S-C, Lu H-H, nucleic acid analysis based on isothermal
Liang Y-H, Lin C-W (2012) Diagnostic recombinase polymerase amplification (RPA).
devices for isothermal nucleic acid amplifica- Lab Chip 10:887–893
tion. Sensors 12:8319–8337 43. Vincent M, Xu Y, Kong H (2004) Helicase-
31. Duarte C, Salm E, Dorvel B, Reddy B Jr, dependent isothermal DNA amplification.
Bashir R (2013) On-chip parallel detection of EMBO Rep 5(8):795–800
foodborne pathogens using loop-mediated 44. Chow WH, McCloskey C, Tong Y, Hu L, You
isothermal amplification. Biomed Microdevices Q, Kelly CP, Kong H, Tang YW, Tang W
15(5):821–830 (2008) Application of isothermal helicase-
32. Fang X, Liu Y, Kong J, Jiang X (2010) Loop- dependent amplification with a disposable
mediated isothermal amplification integrated detection device in a simple sensitive stool test
on microfluidic chips for point-of-care quanti- for toxigenic Clostridium difficile. J Mol Diagn
tative detection of pathogens. Anal Chem 10(5):452–458
82:3002–3006 45. Mahalanabis M, Do J, Al-Muayad H, Zhang
33. Wu Q, Jin W, Zhou C, Han S, Yang W, Zhu Q, JY, Klapperich CM (2010) An integrated dis-
Jin Q, Mu Y (2011) Integrated glass microde- posable device for DNA extraction and heli-
vice for nucleic acid purification, loop- case dependent amplification. Biomed
mediated isothermal amplification, and online Microdevices 12:353–359
detection. Anal Chem 83:3336–3342 46. Francois P, Tangomo M, Hibbs J, Bonetti E-J,
34. Xu L-Y, Jin W, Zhu Q-Y, Yang W-X, Wu Q-Q, Boehme CC, Notomi T, Perkins MD, Shrenzel
Jin Q-H, Mu Y (2011) Real-time detection of J (2011) Robustness of loop-mediated isother-
loop-mediated isothermal amplification reac- mal amplification reaction for diagnostics
tion on microfluidic chip. 2011 5th applications. FEMS Immunol Med Microbiol
International conference on bioinformatics 62:41–48
and biomedical engineering, Wuhan, China, 47. Liu C, Geva E, Mauk MG, Qiu X, Abrams
IEEE, Piscataway, New Jersey, 2011 WR, Malamud D, Curtis K, Owen SM, Bau
35. Notomi T, Okayama H, Masubuchi H, HH (2011) An isothermal amplification reac-
Yonekawa T, Watanabe K, Amino N, Hase T tor with an integrated isolation membrane for
(2000) Loop-mediated isothermal amplifica- point-of-care detection of infectious diseases.
tion of DNA. Nucleic Acids Res 28(12):E63 Analyst 136:2069–2076
36. Mori Y, Kanda H, Notomi T (2013) Loop- 48. Liu C, Mauk MG, Hart R, Bonizzoni M, Yan
mediated isothermal amplification (LAMP): G, Bau HH (2012) A low-cost microfluidic
recent progress in research and development. J chip for rapid genotyping of malaria-transmitted
Infect Chemother 19(3):404–411 mosquitoes. PLoS One 7(8):e42222
37. Parida M, Posadas G, Inoue S, Hasebe F, 49. Stevens DY, Petri CR, Osborn JL, Spicar-
Morita K (2004) Real-time reverse transcrip- Mihalic P, McKenzie KG, Yager P (2008)
tion loop-mediated isothermal amplification Enabling a microfluidic immunoassay for the
for rapid detection of West Nile virus. J Clin developing world by integration of on-card dry
Microbiol 42(1):257–263 reagent storage. Lab Chip 8(12):2038–2045
38. Curtis KA, Rudoph DL, Owen SM (2008) 50. Fridley GE, Le HQ, Fu E, Yager P (2012)
Rapid detection HIV-1 by reverse- Controlled release of dry reagents in porous
transcription, loop-mediated isothermal ampli- media for tunable temporal and spatial distri-
fication (RT-LAMP). J Virol Methods 151: bution upon rehydration. Lab Chip 12(21):
264–270 4321–4327
39. Compton J (1991) Nucleic acid sequence- 51. Hitzbleck M, Gervais L, Delamarche E (2011)
based amplification. Nature 350:91–92 Controlled release of reagents in capillary-
40. Gulliksen A, Solli LA, Drese KS, Sörensen O, driven microfluidics using reagent integrators.
Karlsen F, Rogne H, Hovig E, Sirevåg R Lab Chip 11:2680–2685
(2005) Parallel nanoliter detection of cancer 52. Kim J, Byun D, Mauk MG, Bau HH (2009) A
markers using polymer chips. Lab Chip 5(4): disposable, self-contained PCR chip. Lab Chip
416–420 9(4):606–612
Microfluidic Devices for Nucleic Acid (NA) Isolation, Isothermal NA… 39
53. Kim J, Mauk MG, Chen D, Qiu X, Kim J, Gale 68. Boom R, Sol CJ, Salimans MM, Jansen CL,
B, Bau HH (2010) A PCR reactor with an Wertheim-van Dillen PM, van der Noordaa J
integrated alumina membrane for nucleic acid (1990) Rapid and simple method for purifica-
isolation. Analyst 135:2408–2414 tion of nucleic acids. J Clin Microbiol
54. Ziober B, Mauk M, Chen Z, Bau HH (2008) 28:495–503
Lab-on-a-chip for oral cancer screening and 69. Greenfield L, White TJ (1993) Sample prepa-
diagnosis. Head Neck 30(1):111–121 ration methods. In: Persing DH, Smith TF,
55. Becker H, Gärtner C (2008) Polymer micro- Tenover FC, White TJ (eds) Diagnostic molec-
fabrication technologies for microfluidic systems. ular biology: principles and applications.
Anal Bioanal Chem 390:89–111 American Soc. Microbiology, Washington,
56. Black I (1998) Laser cutting of perspex. DC, pp 122–137
J Mater Sci Lett 17:1531–1533 70. Kemp BM, Monroe C, Smith DG (2006)
57. Pfleging W, Baldus O (2006) Laser patterning Repeat silica extraction: a simple technique for
and welding of transparent polymers for the removal of PCR inhibitors from DNA
microfluidic device fabrication. Laser-based extracts. J Archaeol Sci 33:1680–1689
micropackaging. Proc SPIE 6107:e610705 71. Breadmore MC, Wolfe KA, Arcibal IG, Leung
58. Steigert J, Haeberle S, Brenner T, Müller C, WK, Dickson D, Giordano BC, Power ME,
Steinert CP, Koltay P, Gottschlich N, Reinecke Ferrance JP, Feldman SH, Norris PM, Landers
H, Rühe J, Zengerle R, Ducrée J (2007) Rapid JP (2003) Microchip-based purification of
prototyping of microfluidic chips in COC. DNA from biological samples. Anal Chem
J Micromech Microeng 17:333–341 75:1880–1886
59. Snakenborg D, Klank H, Kutter JP (2004) 72. Chen X, Cui DF, Liu C (2006) High purity
Microstructure fabrication with a CO2 laser DNA extraction with a SPE microfluidic chip
system. J Micromech Microeng 14:182–189 using KI as the binding salt. Chin Chem Lett
60. Tsao C-W, DeVoe DL (2009) Bonding of 17(8):1101–1104
thermoplastic polymer microfluidics. 73. Chen X, Cui D, Sun J, Zhang L, Li H (2013)
Microfluid Nanofluid 6:1–16 Microdevice-based DNA extraction method
61. Hsu Y-C, Chen T-Y (2007) Applying Taguchi using green reagent. Key Eng Mater
methods for solvent-assistant PMMA bonding 562–565:1111–1115
technique for static and dynamic μ-TAS 74. Cady NC, Stelick S, Batt CA (2003) Nucleic
devices. Biomed Microdevices 9:513–522 acid purification using microfabricated struc-
62. Umbrecht F, Müller D, Gattiker F, Boutry tures. Biosens Bioelectron 30(19):59–66
CM, Neuenschwander J, Sennhauser U, 75. Wen J, Legendre LA, Bienvenue JM, Landers
Hierold C (2009) Solvent assisted bonding of JP (2008) Purification of nucleic acids in
polymethylmethacrylate: characterization using microfluidic devices. Anal Chem 80:
the response surface methodology. Sens 6472–6479
Actuators A Phys 156:121–128 76. Wolfe KA, Breadmore MC, Ferrance JP, Power
63. Kodzius R, Xiang K, Wu J, Yi X, Gong X, ME, Conroy JF, Norris PM, Landers JP
Foulds IG, Wen W (2012) Inhibitory effect of (2002) Toward a microchip-based solid-phase
common microfluidic materials on PCR. Sens extraction method for isolation of nucleic
Actuators B 161:349–358 acids. Electrophoresis 23:727–733
64. Giordano BC, Copeland ER, Landers JP 77. Chung YC, Jan M-S, Lin Y-C, Lin J-H, Cheng
(2001) Towards dynamic coating of glass W-C, Fan C-Y (2004) Microfluidic chip for
microchip chambers for amplifying DNA via high efficiency DNA extraction. Lab Chip
polymerase chain reaction. Electrophoresis 4:141–147
22:334–340 78. Gudnason H, Dufva M, Bang DD, Wolff A
65. Gonzalez A, Grimes R, Walsh EJ, Dalton T, (2007) Comparison of multiple DNA dyes for
Davies M (2007) Interaction of quantitative real-time PCR: effects of dye concentration
PCR with polymeric surfaces. Biomed and sequence composition on DNA amplifica-
Microdevices 9:261–266 tion and melting temperature. Nucleic Acids
66. Lou XJ, Panaro NJ, Wilding P, Fortina P, Kricka Res 35(19):e127
LJ (2004) Increased amplification efficiency of 79. Leone G, van Schijndel H, van Gemen B,
microchip-based PCR by dynamic surface pas- Kramer FR, Schoen CD (1998) Molecular
sivation. Biotechniques 36:248–251 beacon probes combined with amplification by
67. Piruska A, Nikcevic I, Lee SH, Ahn C, NASBA enable homogeneous, real-time detec-
Heineman WR, Limbach PA, Seliskar CJ tion of RNA. Nucleic Acids Res 26(9):
(2005) The autofluorescence of plastic materi- 2150–2155
als and chips measured under laser fluores- 80. Lee D-S, Chang B-H, Chen P-H (2005)
cence. Lab Chip 5:1348–1354 Development of a CCD-based fluorimeter for
40 Michael G. Mauk et al.
real-time PCR machine. Sens Actuators B 87. Goto M, Honda E, Ogura A, Nomoto A,
107:872–881 Hanaki K (2009) Colorimetric detection of
81. Walczak R, Bembnowicz P, Szczepanska P, loop-mediated isothermal amplification reac-
Dziuban JA, Golonka L, Koszur J, Bang DD tion using hydroxy naphthol blue.
(2008), Miniaturized system for real-time Biotechniques 46(3):167–172
PCR in low-cost disposable LTCC chip with 88. Tomita N, Mori Y, Kanda H, Notomi T
integrated optical waveguide. Twelfth Int’l (2008) Loop-mediated isothermal amplifica-
conference on miniaturized systems for chem- tion (LAMP) of gene sequences and simple
istry and the life sciences, San Diego. pp visual detection of products. Nat Protoc
1078–1080 3(5):877–882
82. Zhu H, Yaglidere O, Su T-W, Tseng D, Ozcan 89. Liu C, Mauk M, Gross R, Bushman FD,
A (2011) Cost-effective and compact wide- Edelstein PH, Colman RG, Bau HH (2013)
field fluorescent imaging on a cell-phone. Lab Membrane-based, sedimentation-assisted
Chip 11:315–322 plasma separator for point-of-care applications.
83. Lee D-S, Chou WP, Yeh SH, Chen PJ, Chen PH Anal Chem 85:10463–10470
(2011) DNA detection using commercial mobile 90. Gallagher ML, Burke WF Jr, Orzech K (1987)
phones. Biosens Bioelectron 26:4349–4354 Carrier RNA enhancement of recovery from
84. Ahmad F, Seyrig G, Tourlousse DM, Stedtfeld dilute solutions. Biochem Biophys Res
RD, Tiedje JM, Hashsham SA (2011) A Commun 144(1):271–276
CCDbased fluorescence imaging for real-time 91. Shaw KJ, Thain L, Docker PT, Dyer CE,
loop-mediated isothermal amplification-based Greenman J, Greenway GM, Haswell SJ
rapid and sensitive detection of waterborne (2009) The use of carrier RNA to enhance
pathogens on microchips. Biomed DNA extraction from microfluidic-based silica
Microdevices 13:929–937 monoliths. Anal Chim Acta 652:231–233
85. Myers FB, Lee LP (2008) Innovations in opti- 92. Shaw KJ, Oakley J, Docker PT, Dyer CE,
cal microfluidic technologies for point-of-care Greenman J, Greenway GM, Haswell SJ
diagnostics. Lab Chip 8:2015–2031 (2008) DNA extraction, using carrier RNA,
86. Ogilvie IRG, Sieben VJ, Floquet CFA, Zmijan integrated with agarose gel-based polymerase
R, Mowlem MC, Morgan H (2010) Reduction reaction in a microfluidic device. Twelfth
of surface roughness for optical quality international conference on miniaturized sys-
microfluidic devices in PMMA and COC. tems in chemistry and the life sciences, San
J Micromech Microeng 20:065016 Diego. pp 1069–1071
Chapter 3
Abstract
Nanotechnology based diagnostics has provided improved tools for pathogen detection and sensitive and
specific characterization of antibiotic resistance signatures. Tuberculosis (TB) is caused by members of
the Mycobacterium tuberculosis Complex (MTBC) and, according to the World Health Organization, is
one of the most serious infectious diseases in the world. Recent advances in molecular diagnostics of TB
have improved both the detection time and sensitivity but they still require specialized technical person-
nel and cumbersome laboratory equipment. Diagnostics at point-of-need is crucial to TB control as it
may provide rapid identification of pathogen together with the resistance profile of TB strains, originated
from single nucleotide polymorphisms (SNPs) in different loci, allowing for a more accurate indication of
the adequate therapy.
Gold nanoparticles have been widely used in molecular diagnostics platforms. Here, we describe the
use of gold nanoprobes (oligonucleotide functionalized gold nanoparticles) to be used in a non-cross-
linking colorimetric method for the direct detection of specific DNA targets. Due to the remarkable optical
properties of gold nanoparticles, this detection system provides colorimetric detection of the pathogen
together with the potential of identification of several single nucleotide polymorphisms (SNPs) involved in
TB resistance to antibiotics. For point-of-need use, we adapted this strategy to a low-cost mobile scheme
using a paper based revelation platform and where the spectral signature is transposed to RGB data via a
smartphone device. This way, identification of pathogen and characterization of resistance signatures is
achieved at point-of-need.
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_3, © Springer Science+Business Media New York 2015
41
42 B. Veigas et al.
Fig. 1 Au-nanoprobe strategy for detection of MTBC members. Schematic representation of the detection with
gold nanoprobes. The colorimetric assay consists of visual comparison of test solutions after salt induced
Au-nanoprobe aggregation on a [MgCl2] impregnated paper plate. After color development a photo of the paper
plate is captured and a RGB and GPS analysis is performed
44 B. Veigas et al.
2 Materials
Table 1
Oligonucleotide sequences
3 Methods
3.1 Gold Nanoprobe Gold nanoprobe (see Note 4–6) synthesis was performed by deriva-
Synthesis tizing 13 nm gold nanoparticles with thiol-ssDNA oligonucle-
otides. Detailed materials and methods for the preparation and
handling of such particles can be found in Conde et al. [22].
1. Extract one volume (100–500 μl) of thiol-modified oligonu-
cleotide resuspended in 0.1 M DTT with two volumes of ethyl
acetate.
2. Discard the organic phase (i.e., upper phase) after centrifuging
for 5 min at 21,460 × g.
3. Repeat steps 1 and 2 two more times.
4. Purify the remaining aqueous phase through a desalting NAP-5
column, following the manufacturer’s instructions and using
10 mM phosphate buffer (pH 8) as eluent. Typically, after col-
umn equilibration with the eluent, 500 μl of the aqueous phase
is added and let to enter the column. Afterwards, 1000 μl of
purified thiol-modified oligonucleotide can be collected by
adding 1,000 μl of the eluent to the column.
5. Quantify the purified thiol-modified oligonucleotide by UV/
Vis spectroscopy using the extinction coefficient at 260 nm
provided by the oligonucleotide manufacturer.
6. Mix the purified thiol-modified oligonucleotide with the col-
loidal solution in a 1:160 and 1:200 AuNPs/oligo for MTC
and rpo531 probes, respectively.
7. Add AGE I solution to achieve a final concentration of 10 mM
phosphate buffer (pH 8), 0.01 % (w/v) SDS. Typically, 15.1 μl
of AGE I solution is added to a volume of 3 ml of the solution
prepared in step 6.
8. Sonicate the solution for 10 s using an ultrasound bath and
incubate at room temperature for 20 min.
9. Afterwards, sequentially increase the ionic strength of the solu-
tion in 50 mM NaCl increments by adding the respective vol-
ume of AGE II solution up to a final concentration of 10 mM
phosphate buffer (pH 8), 0.3 M NaCl, 0.01 % (w/v)
SDS. Typically, 104.1, 111.6, 119.8, 129.1, 139.4, and 151 μl
of AGE II solution are added sequentially to the 3,015.1 μl of
the solution prepared in step 7. After each increment, sonicate
the solution for 10 s and incubate at room temperature for
20 min before the next increment—see Note 7.
10. Incubate the solution overnight at room temperature.
11. Distribute the functionalized nanoparticles in 1.5 ml micro-
centrifuge tube and centrifuge for 20 min at 21,460 × g.
48 B. Veigas et al.
12. Discard the supernatant. Wash the resulting oily pellet with
1 ml/microcentrifuge tube of 10 mM phosphate buffer (pH 8)
and centrifuge for 20 min at 21,460 × g.
13. Repeat step 12.
14. Discard the supernatant, wash with 1 ml/microcentrifuge tube
of PBS solution and centrifuge for 20 min at 21,460 × g.
15. Discard the supernatant and finally resuspend the pellet in
500 μl/microcentrifuge tube of PBS solution. Gather the
resulting solutions of each microcentrifuge tube in a polypro-
pylene or glass vial with a conical skirted base.
16. Prepare aliquots of 15 or 0.3 nM of Au-nanoprobes, using
PBS as eluent. The initial nanoprobe concentration can be
determined by the Lambert–Beer law using the molar absorp-
tivity of the respective nanoparticles.
17. Store the nanoprobe stock solutions in the dark at 4 °C until
further use.
DNA Extraction 1. Heat water bath or heating block to 95 °C to use in step 6 and
with QIAamp DNA Mini Kit to 56 °C to use in step 9.
2. Centrifuge 1,000 μl of the sample at 15,493 × g for 10 min.
3. Discard supernatant and add 1,000 μl Buffer TE to the pellet.
Mix by pulse-vortexing.
4. Centrifuge at 15,493 × g for 10 min.
5. Discard supernatant and add 200 μl Buffer TE to the pellet.
Mix by pulse-vortexing.
6. Incubate at 95 °C for 20 min.
7. Pipet 20 μl of Proteinase K QIAGEN into the tube.
8. Add 200 μl Buffer AL to the sample. Mix by pulse-vortexing
for 15 s.
9. Incubate at 56 °C for 10 min.
10. Briefly centrifuge the 1.5 ml microcentrifuge tube to remove
drops from the inside of the lid.
Mobile Based Gold Nanoprobe TB Diagnostics for Point-of-Need 49
Table 2
Setup for characterization of nanoprobe aggregation
4. Add the 0.3 M MgCl2 according to Table 2, mix well and spin
down the solutions – see Note 9.
5. Incubate the solutions for 15 min at room temperature and
register their absorption spectra (350–800 nm) using a
UV–visible spectrophotometer or microplate reader.
6. Plot Abspeak/Abs600nm vs. [MgCl2]final to determine the mini-
mum salt concentration needed for a complete nanoprobe
aggregation, where Abspeak is the absorbance peak of the
initial dispersed nanoprobe (e.g., typically, 520 nm for
Au-nanoprobes). For Abspeak/Abs600nm ratios below 1, the
nanoprobe is considered to be fully aggregated. If the nano-
probes do not completely aggregate and change color within
the suggested concentrations of MgCl2, use higher concen-
trations of MgCl2. If the nanoprobe changes color and fully
aggregates at a final MgCl2 concentration of 10 mM, then
derivatization of the nanoparticles with the thiol-modified
oligonucleotides may have not been effective. If so, repeat
nanoprobe synthesis – see Note 10.
Fig. 2 Illustration of the characteristic detection assay results and colorimetric call. (a) MTBC detection: UV/
visible analysis of Au-nanoprobe alone—Blank; in presence of a complementary target—MTBC; and in pres-
ence of a non-complementary target—non-MTBC and non-related, registered 30 min after salt addition.
Results presented in Abs526nm/Abs600nm ratios of the UV/visible spectra. (b) Mutation analysis: ratio of WT probe
divided by Mut probe, calculated for rpoB 531 were values >1 indicate a WT genotype (White) and <1 a Mut
genotype (Black). The bars represent the average of three independent measurements and the error bars
indicate standard deviation
52 B. Veigas et al.
3.4 Paper Platform Starting from sheets of a cellulose substrate Whatman No. 1
Chromatography paper (Whatman International Ltd., Floram
3.4.1 Paper Platform
Park, NJ, USA), A5 standard format (210 × 148 mm) sheets were
Preparation
cut. This paper size fits directly into the manual feed tray of a com-
mercial solid ink printer (Xerox ColorQube 8570, Xerox
Corporation, Norwalk, CT, USA) designed to print a wax based
ink, originating hydrophobic barriers. Paper microplates were
designed with a standard 384 plate format in Microsoft Office
Visio1 (Microsoft Corporation, US). All the measurements were
taken from the “Microplate Dimensions Guide, Compendium of
Greiner Bio-One Microplates” (Greiner Bio-One GmbH,
Frickenhausen, Germany). The printed pattern of a 384 microplate
was placed on a hot plate (Heidolph MR Hei-Tec, Schwabach,
Germany) at 140 μC for 2 min, allowing the wax to melt and
spread vertically through the whole thickness of the paper, creating
the desired hydrophobic pattern. Each well was impregnated with
1 ml of a 0.12 M MgCl2 solution (revelation agent) and allowed to
dry at 25 °C for 10 min. The final microplates were stored at
25 μC, and wrapped in aluminum foil until use.
3.4.2 Paper Platform For assaying with the Gold on Paper platform, a total reaction mix-
Detection ture of 5 ml was used with 2.5 nM Au-nanoprobes in 10 mM
phosphate buffer (pH 8), 0.1 M NaCl and target DNA at a final
concentration of 30 mg/ml. After 10 min of denaturation at
95 °C, the mixtures were allowed to stand for 10 min at room
temperature and spotted onto the well on the paper plate. After
45 min at room temperature for color development, the paper
plate was photographed with a mobile device and RGB analysis
was performed.
3.5 Data Analysis The color pattern on the Gold on Paper was captured with an
HTC Desire android smartphone with a 5 megapixel camera
(2,592 × 1,944 pixels) with autofocus. Photos were taken with
artificial white light without flash. Blank test spots with 10 mM
phosphate buffer (pH 8), were used to normalize the data for
light conditions. The digitalized data were then analyzed without
further manipulation with a free RGB analysis application
(ColorPikr, WiseClue) and transmitted via a 3G network to a per-
sonal computer for image processing with ImageJ™. Each assay
was repeated at least three times and on four different paper
micro-well plates (Fig. 3).
Mobile Based Gold Nanoprobe TB Diagnostics for Point-of-Need 53
Fig. 3 Representative Gold on paper assay results. Digital photography and RGB
data analysis of Au-nanoprobe mix in the presence of Au-nanoprobe alone—
Blank; in presence of a complementary target—MTBC; and in presence of a
non-complementary target—non-MTBC and non-related. Ratio of aggregation
calculated in the Smartphone (ratio of average intensity of the Red and Blue
channels) for the assay mixtures. Paper microplate assay performed with—
2.5 nM Au-nanoprobe, 10 mM phosphate buffer (pH 8), 0.1 M NaCl, and sample
DNA at a final concentration of 30 μg/ml in a final volume of 5 μl per test. Image
captured after 45 min. The bars represent the average of three independent
measurements and the error bars indicate standard deviation. The horizontal line
represents the threshold of 1 considered for discrimination between positive and
negative. Statistical analysis was performed using Prism 5 graph pad, using one-
way ANOVA with Tukey’s Multiple Comparison test; ***p < 0.001, n = 3
4 Notes
References
1. Azzazy HME, Mansour MMH (2009) In vitro resource limited settings. J Microbiol Methods
diagnostic prospects of nanoparticles. Clin 84:155–160
Chim Acta 403:1–8 8. Miller LP, Crawford JT, Shinnick TM (1994)
2. Veigas B, Doria G, Baptista PV (2012) The rpoB gene of Mycobacterium tuberculosis.
Nanodiagnostics for tuberculosis. In: Cardona Antimicrob Agents Chemother 38:805–811
PJ (ed) Understanding tuberculosis—global 9. Musser JM (1995) Antimicrobial agent resis-
experiences and innovative approaches to the tance in Mycobacteria: molecular genetic
diagnosis. InTech. pp 257–276 insights. Clin Microbiol Rev 8:496–514
3. Baptista P, Pereira E, Eaton P et al (2008) 10. Soini H, Musser JM (2001) Molecular diagno-
Gold nanoparticles for the development of sis of Mycobacteria. Clin Chem 47:809–814
clinical diagnosis methods. Anal Bioanal Chem 11. Telenti A, Imboden P, Marchesi F et al (1993)
391:943–950 Detection of rifampicin-resistance mutations
4. Doria G, Conde J, Veigas B et al (2012) Noble in Mycobacterium tuberculosis. Lancet
metal nanoparticles for biosensing applica- 341:647–651
tions. Sensors 12:1657–1687 12. Baptista PV, Koziol-Montewka M, Paluch-
5. WHO (2010) Global tuberculosis control: Oles J et al (2006) Gold-nanoparticle-probe-
WHO Report 2010. WHO Press, Geneva. based assay for rapid and direct detection of
ISBN 978-92-4-156406-9 Mycobacterium tuberculosis DNA in clinical
6. Deun AV, Martin A, Palomino JC (2010) samples. Clin Chem 52:1433–1434
Diagnosis of drug-resistant tuberculosis: reli- 13. Conde J, de la Fuente JM, Baptista PV (2010)
ability and rapidity of detection. Int J Tuberc RNA quantification using gold nanoprobes—
Lung Dis 14:131–140 application to cancer diagnostics. J
7. Abebe G, Paasch F, Apers L et al (2011) Nanobiotechnology 8:5
Tuberculosis drug resistance testing by molec- 14. Costa P, Amaro A, Botelho A et al (2010)
ular methods: opportunities and challenges in Gold nanoprobes assay for identification of
56 B. Veigas et al.
mycobacteria from the Mycobacterium tuber- 19. Carrilho E, Phillips ST, Vella SJ et al (2009) Paper
culosis complex. Clin Microbiol Infect microzone plates. Anal Chem 81:5990–5998
16:1464–1469 20. Carrilho E, Martinez AW, Whitesides GM
15. Doria G, Franco R, Baptista P (2007) (2009) Understanding wax printing: a simple
Nanodiagnostics: fast colorimetric method for micropatterning process for paper-based
single nucleotide polymorphism/mutation microfluidics. Anal Chem 81:7091–7095
detection. IET Nanobiotechnol 1:53–57 21. Kent PT, Kubica GP (1985) Mycobacteriology: a
16. Veigas B, Machado D, Perdigão J et al guide for the level III laboratory. US Dept of
(2010) Au-nanoprobes for detection of Health and Human Services, Public Health
SNPs associated with antibiotic resistance in Service, Centers for Disease Control, Atlanta, GA
Mycobacterium tuberculosis. Nanotechnology 22. Conde J, Doria G, de la Fuente JM, Baptista
21:415101 PV (2012) RNA quantification using noble
17. Silva LB, Veigas B, Doria G et al (2011) metal nanoprobes: simultaneous identification
Portable optoelectronic biosensing platform of several different mRNA targets using color
for identification of mycobacteria from the multiplexing and application to cancer diag-
Mycobacterium tuberculosis complex. Biosens nostics. Methods Mol Biol 906:71–87
Bioelectron 26:2012–2017 23. Doria G, Baumgartner BG, Franco R et al
18. Veigas B, Jacob JM, Costa MN (2012) Gold (2010) Optimizing Au-nanoprobes for specific
on paper-paper platform for Au-nanoprobe TB sequence discrimination. Colloids Surf B
detection. Lab Chip 12:4802–4808 Biointerfaces 77:122–124
Chapter 4
Abstract
A immunofluorescence microtip sensor was developed for specific detection of Mycobacterium cells in
sputum samples by the combination of electric field, streaming flow, and immuno-affinity binding. The
detection limit was 200 CFU/mL in human sputum, which was comparable to PCR but without requiring
bacteriological culture, centrifugation, or nucleic acid amplification. In spite of the complex nature of
physical, chemical, and biological mechanisms, the simple operation of “dipping and withdrawal” of tips
will allow for screening by minimally trained personnel within 30 min. In addition, the minimal power
requirement (5 W) combined with low assay cost is ideal for point-of-care (POC) screening in resource-
limited settings.
Key words Point-of care diagnosis, Tuberculosis, Sputum, Immunosensor, Electric field, Microtip
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_4, © Springer Science+Business Media New York 2015
57
58 Jong-Hoon Kim et al.
Streaming flow
Electrohydrodynamic flow
microtips
Dielectrophoresis
solution
a
Top view
Microtip
b
0.004
0.002
Microtip
0
−0.01
−0.005
0 0.008
0.006
0.005 0.004
0.01 0.002
Front view 0
−2
0 (Unit : meter)
2
Y axis
8
Side view
10
0 1 2 3 4 5 6 7 8 9 10
X axis
Fig. 3 (a) PIV analysis of two-dimensional flow for each plane, which is generated by longitudinal vibration of
an aluminum well. (b) Three-dimensional flow that is generated by combining two-dimensional flows (reprinted
with permission [41])
2 Materials
2.2 Reagents 1. Phosphate buffered saline (1× PBS, pH 7.4, KH2PO4 1.05 mM,
NaCl 155.17 mM, Na2HPO4-7H2O 2.96 mM).
2. Polyclonal IgY antibodies raised against Mycobacterium tuber-
culosis complex.
3. Polyethyleneimine solution (PEI, 1 % (w/v) in DI water).
4. Biotin-conjugated bovine serum albumin (BSA–biotin,
10 mg/mL in 1× PBS).
5. Streptavidin from Streptomyces avidin (streptavidin, 1 mg/mL
in 1× PBS, greater than 13 units/mg protein, streptavidin has
four binding sites for biotins).
62 Jong-Hoon Kim et al.
3 Methods
a
Si3N4 film
Si wafer
Photoresist
UV lithography Remove the Resist
RIE KOH etching
Linear motor
Microtip
holder
Microtip
Aluminum
well
Vibration motor
3.3 BCG and MTB 5 mL cultures of Difco Middlebrook 7H9 broth with ADC enrich-
(H37Ra) Growth ment, 0.2 % glycerol and 0.05 % Tween 20 were inoculated with
and Preparation the Bacillus Calmette-Guérin (BCG) strain of Mycobacterium bovis
(or MTB; H37Ra) and incubated in a 37 °C incubator for between
64 Jong-Hoon Kim et al.
3.6 Microtip The dipping method was utilized for the functionalization.
Functionalization
1. First, the microtip was immersed into 1 % PEI for 1 min and
(Surface Chemistry)
withdrawn at a rate of 100 μm/s. It was coated as an adhesive
layer.
2. The PEI treated tip was then immersed in biotinylated bovine
serum albumin (10 mg/mL in PBS) for 5 min.
Microtip Immunosensor 65
Aluminum well
Microchip
Sputum NALC
Negative
Glass beads Shaker
Shaker 3mm
SDS Positive
Fig. 6 25 min test procedure of a microtip immunofluorescence sensor; (1) sputum was processed to reduce
the viscosity in a simple 15-min procedure; (2) target bacteria in the 1-mL processed sample were specifi-
cally concentrated onto a microtip by a combination of convective flows, an AC field, and affinity binding for
a total of 3 min; and (3) after binding fluorescein-labeled antibodies, the fluorescence signal was processed
for 7 min. In total, 25 min were required for a single assay, from sample cup to a numerical result (reprinted
with permission [41])
3.7 Sputum Sample Sputum samples from people known not to have TB were pur-
Preparation chased from BioReclamation, Inc. To dissipate the sputum, we used
NALC and SDS with bead-beating as illustrated in Fig. 6.
1. Sputum spiked with MTB (total 600 μL) was mixed with
300 μL NALC (4 mg/mL) and 3 mm glass beads.
2. The sample was vortexed for 5 min at 200 × g .
3. 300 μL SDS (4 %) was added to the mixture. Additional
vortexing for 5 min was conducted for complete liquefaction.
In the process, the final concentration of BCG in the processed
sputum was the half of each initial concentration.
4. Out of total volume of 1.2 mL, 1 mL was transferred to an
aluminum well without glass beads.
3.8 Concentration The test was conducted by three simple steps (Fig. 6):
and Detection
1. Microtips were dipped into the sample with a vibration and an
applied AC field (20 Vpp at 5 MHz) for a 2-min concentration
step. Subsequently, the microtips were withdrawn at a rate of
100 μm/s (see Note 1).
2. The microtips were immersed in a fluorescein conjugated
antibody solution (10 μL, 2 mg/mL) for 5 min.
66 Jong-Hoon Kim et al.
8000
(N=3) SF : Streaming flow
7000
Number of cells (CFU/mm2)
6000
5000
4000
3000
2000
1000
0
E-field, SF SF E-field None
Fig. 7 Effect of each concentration mechanism. The values are number of captured
cells per microtip (reprinted with permission [41])
Microtip Immunosensor 67
a b
10
87 683 (N=4)
8
Normalized value
6
0
3107 5503 Control 2x102 2x103 2x104
MTB H37Ra in sputum [CFU/mL]
c
10
(N=24)
Normalized value 8
6
11793 15356
4
0
Control 2x102
MTB H37Ra in sputum [CFU/mL]
Fig. 8 Test results of a microtip immunosensor. (a) Digitized images from sputum samples spiked with BCG.
The concentrations of BCG and the average numbers of white pixels are shown at the bottom and top in the
images, respectively. The scale bar is 50 μm. (b) Dose response for MTB spiked in sputum samples.
(c) Reproducibility test at an MTB concentration of 200 CFU/mL in sputum. Compared with 6 negative controls,
23 out of 24 samples show positive signals. The total success rate is 96 % (reprinted with permission [41])
4 Notes
Acknowledgements
References
1. WHO (2010) Global tuberculosis control: 12. Elliott AM, Luo N, Tembo G, Halwiindi B,
WHO Report 2010. WHO, Geneva Steenbergen G, Machiels L et al (1990) Impact
2. WHO (2009) Global tuberculosis control: sur- of HIV on tuberculosis in Zambia—a cross-
veillance, planning, financing. WHO, Geneva sectional study. Br Med J 301:412–415
3. Lienhardt C, Rowley J, Manneh K, Lahai G, 13. Garland LH (1959) Studies on the accuracy of
Needham D, Milligan P et al (2001) Factors diagnostic procedures. Am J Roentgenol
affecting time delay to treatment in a tubercu- Radium Ther Nucl Med 82:25–38
losis control programme in a sub-Saharan 14. Rodriguez-Lazaro D, D’Agostino M,
African country: the experience of The Gambia. Herrewegh A, Pla M, Cook N, Ikonomopoulos
Int J Tuberc Lung Dis 5:233–239 J (2005) Real-time PCR-based methods for
4. Golub JE, Bur S, Cronin WA, Gange S, Baruch detection of Mycobacterium avium Subsp
N, Comstock GW et al (2006) Delayed tuber- paratuberculosis in water and milk. Int J Food
culosis diagnosis and tuberculosis transmission. Microbiol 101:93–104
Int J Tuberc Lung Dis 10:24–30 15. Thomson LM, Traore H, Yesilkaya H, Doig C,
5. Aber VR, Allen BW, Mitchison DA, Ayuma P, Steingrimsdottir H, Garcia L et al (2005) An
Edwards EA, Keyes AB (1980) Quality-control extremely rapid and simple DNA-release
in tuberculosis bacteriology. 1. Laboratory method for detection of M. tuberculosis from
studies on isolated positive cultures and the clinical specimens. J Microbiol Methods 63:
efficiency of direct smear examination. Tubercle 95–98
61:123–133 16. Choi YJ, Hu Y, Mahmood A (1996) Clinical
6. Boyd JC, Marr JJ (1975) Decreasing reliability significance of a polymerase chain reaction
of acid-fast smear techniques for detection of assay for the detection of Mycobacterium
tuberculosis. Ann Intern Med 82:489–492 tuberculosis. Am J Clin Pathol 105:200–204
17. Durmaz R, Aydin A, Durmaz B, Aydin NE,
7. Steingart KR, Ramsay A, Pai M (2007)
Akbasak BS, Gunal S (1997) Sensitivity of
Optimizing sputum smear microscopy for the
two-stage PCR amplification for detection of
diagnosis of pulmonary tuberculosis. Expert
Mycobacterium tuberculosis in paraffin-
Rev Anti Infect Ther 5:327–331
embedded tissues. J Microbiol Methods 29:
8. Perkins MD, Small PM (2006) Partnering for 69–75
better microbial diagnostics. Nat Biotechnol 18. Krambovitis E, Lock PE, Mcillmurray MB,
24:919–921 Hendrickse W, Holzel H (1984) Rapid diagno-
9. Keeler E, Perkins MD, Small P, Hanson C, sis of tuberculous meningitis by latex particle
Reed S, Cunningham J et al (2006) Reducing agglutination. Lancet 2:1229–1231
the global burden of tuberculosis: the contri- 19. Tamminen M, Joutsjoki T, Sjoblom M, Joutsen
bution of improved diagnostics. Nature M, Palva A, Ryhanen EL et al (2004) Screening
444(Suppl 1):49–57 of lactic acid bacteria from fermented vegeta-
10. Lazcka O, Del Campo FJ, Munoz FX (2007) bles by carbohydrate profiling and PCR-
Pathogen detection: a perspective of traditional ELISA. Lett Appl Microbiol 39:439–444
methods and biosensors. Biosens Bioelectron 20. Nassau E, Parsons ER, Johnson GD (1976)
22:1205–1217 The detection of antibodies to Mycobacterium
11. Torres-Chavolla E, Alocilja EC (2009) tuberculosis by microplate enzyme-linked
Aptasensors for detection of microbial and viral immunosorbent assay (ELISA). Tubercle 57:
pathogens. Biosens Bioelectron 24:3175–3182 67–70
Microtip Immunosensor 69
21. Delacourt C, Gobin J, Gaillard JL, Deblic J, 32. Huang Y, Joo S, Duhon M, Heller M, Wallace
Veron M, Scheinmann P (1993) Value of B, Xu X (2002) Dielectrophoretic cell separation
ELISA using antigen-60 for the diagnosis of and gene expression profiling on microelectronic
tuberculosis in children. Chest 104:393–398 chip arrays. Anal Chem 74:3362–3371
22. Middlebrook G, Reggiardo Z, Tigertt WD 33. Yeo WH, Chung JH, Liu YL, Lee KH (2009)
(1977) Automatable radiometric detection of Size-specific concentration of DNA to a nano-
growth of Mycobacterium-tuberculosis in selec- structured tip using dielectrophoresis and capil-
tive media. Am Rev Respir Dis 115:1066–1069 lary action. J Phys Chem B 113:10849–10858
23. Gamboa F, Manterola JM, Lonca J, Matas L, 34. Yeo WH, Liu S, Chung JH, Liu YL, Lee KH
Vinado B, Gimenez M et al (1997) Detection (2009) Rapid detection of Mycobacterium
and identification of mycobacteria by amplifica- tuberculosis cells by using microtip-based
tion of RNA and DNA in pretreated blood and immunoassay. Anal Bioanal Chem 393:
bone marrow aspirates by a simple lysis method. 1593–1600
J Clin Microbiol 35:2124–2128 35. Yeo WH, Chou FL, Fotouhi G, Oh K, Stevens
24. Cambau E, Wichlacz C, Truffot-Pernot C, BT, Tseng HY et al (2010) Size-selective
Jarlier V (1999) Evaluation of the new MB immunofluorescence of Mycobacterium tuber-
Redox system for detection of growth of myco- culosis cells by capillary- and viscous forces.
bacteria. J Clin Microbiol 37:2013–2015 Lab Chip 10:3178–3181
25. Qin DL, He XX, Wang KM, Tan WH (2008) 36. Wei Q, Qi H, Luo W, Tseng D, Ki SJ, Wan Z
Using fluorescent nanoparticles and SYBR et al (2013) Fluorescent imaging of single
Green I based two-color flow cytometry to nanoparticles and viruses on a smart phone.
determine Mycobacterium tuberculosis avoid- ACS Nano 7:9147–9155
ing false positives. Biosens Bioelectron 24: 37. Miller AR, Davis GL, Oden ZM, Razavi MR,
626–631 Fateh A, Ghazanfari M et al (2010) Portable,
26. Griffiths D, Hall G (1993) Biosensors—what battery-operated, low-cost, bright field and
real progress is being made. Trends Biotechnol fluorescence microscope. PLoS One 5:4
11:122–130 38. Minion J, Pai M, Ramsay A, Menzies D,
27. Owen VM (1994) Market requirements for Greenaway C (2011) Comparison of LED and
advanced biosensors in healthcare. Biosens conventional fluorescence microscopy for
Bioelectron 9:xxix–xxxv detection of acid fast bacilli in a low-incidence
28. Bown MR, Meinhart CD (2006) AC electro- setting. PLoS One 6:e22495
osmotic flow in a DNA concentrator. 39. Trusov A, Bumgarner R, Valijev R, Chestnova
Microfluid Nanofluidics 2:513–523 R, Talevski S, Vragoterova C et al (2009)
29. Wong PK, Chen CY, Wang TH, Ho CM Comparison of Lumin LED fluorescent attach-
(2004) Electrokinetic bioprocessor for concen- ment, fluorescent microscopy and Ziehl-
trating cells and molecules. Anal Chem Neelsen for AFB diagnosis. Int J Tuberc Lung
76:6908–6914 Dis 13:836–841
30. Asbury CL, van den Engh G (1998) Trapping 40. Wayne LG (1976) Dynamics of submerged
of DNA in nonuniform oscillating electric growth of Mycobacterium tuberculosis under
fields. Biophys J 74:1024–1030 aerobic and microaerophilic conditions. Am
31. Gimsa J (2001) A comprehensive approach to Rev Respir Dis 114:807–811
electro-orientation, electrodeformation, dielec- 41. Kim J-H, Yeo W-H, Shu Z, Soelberg SD,
trophoresis, and electrorotation of ellipsoidal Inoue S, Kalyanasundaram D et al (2012)
particles and biological cells. Bioelectrochemistry Immunosensor towards low-cost, rapid diagnosis
54:23–31 of tuberculosis. Lab Chip 12:1437–1440
Chapter 5
Abstract
Optical detection technologies based on mobile devices can be utilized to enable many mHealth applica-
tions, including a reader for lateral-flow immunoassay (LFIA). However, an intrinsic challenge associated
with LFIA for clinical diagnostics is the limitation in sensitivity. Therefore, rapid and simple specimen
processing strategies can directly enable more sensitive LFIA by purifying and concentrating biomarkers.
Here, a binary reagent system is presented for concentrating analytes from a larger volume specimen to
improve the malaria LFIA’s limit of detection (LOD). The biomarker enrichment process utilizes
temperature-responsive gold–streptavidin conjugates, biotinylated antibodies, and temperature-responsive
magnetic nanoparticles. The temperature-responsive gold colloids were synthesized by modifying the
citrate-stabilized gold colloids with a diblock copolymer, containing a thermally responsive poly(N-
isopropylacrylamide) (pNIPAAm) segment and a gold-binding block composed of NIPAAm-co-N,N-
dimethylaminoethylacrylamide. The gold–streptavidin conjugates were synthesized by conjugating
temperature-responsive gold colloids with streptavidin via covalent linkages using carbodiimide chemistry
chemistry. The gold conjugates formed half-sandwiches, gold labeled biomarker, by complexing with bio-
tinylated antibodies that were bound to Plasmodium falciparum histidine-rich protein 2 (PfHRP2), a
malaria antigen. When a thermal stimulus was applied in conjunction with a magnetic field, the half-
sandwiches and temperature-responsive magnetic nanoparticles that were both decorated with pNIPAAm
formed large aggregates that were efficiently magnetically separated from human plasma. The binary
reagent system was applied to a large volume (500 μL) specimen for concentrating biomarker 50-fold into
a small volume and applied directly to an off-the-shelf malaria LFIA to improve the signal-to-noise ratio.
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_5, © Springer Science+Business Media New York 2015
71
72 James J. Lai and Patrick S. Stayton
Fig. 1 Depiction of the magnetic enrichment lateral-flow immunoassay. A biotinylated antibody is added to a
plasma sample containing the target biomarker(s). An equal volume of buffer containing temperature-
responsive gold–streptavidin conjugates, pNIPAAm magnetic nanoparticles, and free pNIPAAm polymer is
added. Upon heating, the mixed gold–magnetic nanoparticle aggregates are separated by a magnet. After the
supernatant is discarded, the captured aggregates are redissolved into a smaller volume of cool buffer, result-
ing in particle disaggregation and 50-fold enrichment. The enriched mixture is then applied directly to an
immunochromatographic assay membrane with functionalized test and control line antibody regions.
Reproduced from ACS Nano 2012 with permission from ACS Publications
74 James J. Lai and Patrick S. Stayton
2 Materials
3 Methods
Fig. 2 Targeted bioconjugate gold colloid design. Gold colloids were modified with a diblock copolymer pro-
duced via two-step RAFT polymerization. The polymer’s semi-telechelic carboxyl group was conjugated to
lysine groups on streptavidin, enabling linkage to a biotinylated antibody. The conjugate design allowed facile
multiplexed detection by simply mixing different biotinylated antibodies with the sample before addition of the
universal streptavidin-gold detection reagent. Reproduced from ACS Nano 2012 with permission from ACS
Publications
76 James J. Lai and Patrick S. Stayton
3.1.1 Synthesis of mCTA, 1. Polymer targeted ca. 200 degree of polymerization (DP) was
pNIPAAm prepared by mixing in a round-bottom flask 3.11 g
(27.5 mmol) of recrystallized NIPAAm (see Note 1), 85 mg
(0.134 mmol) of DCT, and 3.4 mg (13.4 μmol) of AIBN in
6 g 1,4-dioxane.
2. The flask was sealed with a rubber septum and the solution
was raised to ca. 30–40 °C using a warm tap water bath, while
purging with N2 for ≥20 min prior to the polymerization.
Then, the polymerization was carried out by heating the solu-
tion in the sealed flask via an oil bath at 60 °C for 18 h.
3. The resulting polymers were diluted in 15 mL THF, and then
isolated by precipitation into 200 mL pentane. The precipitate
was filtered, dried in vacuo, dialyzed against DI water at 4 °C
for 72 h with MWCO 6–8 kDa dialysis membrane, and
lyophilized.
4. The polymer molecular weight was characterized using size
exclusion chromatography with multiangle light scattering
detection using dn/dc = 0.076.
3. The solution was titrated to pH 8.2 using 0.1 N NaOH, and then
1.2 mL of 10 mg/mL diblock copolymer solution was added.
4. After the polymer addition, the flask was purged with N2 for
45 min and stirred at room temperature (ca. 22 °C) for 24 h
in darkness.
5. The solution was added with 1 g of NaCl and stirred for an
additional 24 h at room temperature.
6. The particles were then concentrated using Amicon stirred
cells with 100 kDa NMWL Ultracel regenerated cellulose
membranes under 35–40 psi of N2.
7. The temperature-responsive gold colloids on the membrane
were collected by washing with 2 mL of 0.1 M MES buffered
saline at pH 5.0.
3.2 Synthesis The magnetic nanoparticle synthesis will utilize micelles comprised
of Temperature- of DMP-pNIPAAm, pNIPAAm with a dodecyl (hydrophobic)
Responsive Magnetic chain end, for dimensional confinement as previously described
Nanoparticles [24]. The polymer synthesis was carried out using RAFT polymer-
ization with DMP CTAs.
3.3 Synthesis Biotinylation of the IgG antibodies was performed using an NHS-
of Biotinylated activated biotin containing a chromophore linker.
Anti-PfHRP2 IgG
1. The NHS-chromogenic-biotin dissolved at 10 mg/mL in anhy-
drous DMF was added in 7.5 M excess (8.1 μL) to 2 mL of the
IgG antibodies at 1 mg/mL in PBS, at room temperature.
2. After 3 h incubation at room temperature, the unreacted
NHS-chromogenic- biotin was removed using a Zeba spin
desalting column.
3. The degree of biotinylation was estimated by measuring the
ratio of the biotin-chromophore extinction (ε = 29,000 M−1 cm−1
at 354 nm) to the IgG extinction (ε = 210,000 M−1 cm−1 at
280 nm) (see Note 7).
3.4 Lateral-Flow 1. To remove the flow strip, plastic cassette of the lateral-flow test
Device Modification was opened using a utility knife (Fig. 3a).
2. The dried gold conjugate pad (pink) and absorbent pad
(Fig. 3b) were removed and discarded, and then the flow strip
was removed from the plastic cassette and remounted onto a
strip of adhesive-coated mylar (4 mm wide × 38 mm long)
using a pair of tweezers (Fig. 3b).
3. A porous rectangular sheet of cellulose fiber was placed on the
assay membrane upstream of the capture lines (Fig. 3c) to
Biomarker Enrichment 79
3.5 Magnetic PfHRP2 enrichment utilized the recently developed binary reagent
Enrichment of PfHRP2 system, including temperature-responsive magnetic nanoparticles
for Lateral-Flow and temperature-responsive gold–streptavidin conjugate, in con-
Immunoassays junction with the biotinylated antibodies. While the enrichment
protocol described here utilized a lab-based incubator for heating,
the process is suitable is suitable for low-resource setting environ-
ment by using battery-powered heater.
1. Biotinylated anti-PfHPR2 IgG (50 μL, 10 nM) was added
into the tube containing 250 μL human plasma specimen,
and then followed sequentially by 50 μL PBS, 50 μL
temperature-responsive gold colloid–streptavidin conjugates
(20 nM), 50 μL temperature-responsive magnetic nanoparti-
cles (10 mg/mL), and 50 μL mCTA (15 mg/mL).
80 James J. Lai and Patrick S. Stayton
Fig. 5 Comparison of magnetic enrichment and commercial assay. Flow strip images from a 50-fold magnetic
enrichment immunoassay (top row) and from the unmodified commercial assay performed with no enrichment
(bottom row). Reproduced from ACS Nano 2012 with permission from ACS Publications
4 Notes
References
1. Anderson NL (2010) The clinical plasma pro- 5. Yager P, Domingo GJ, Gerdes J (2008) Point-
teome: a survey of clinical assays for proteins in of-care diagnostics for global health. Annu Rev
plasma and serum. Clin Chem Biomed Eng 10:107–144
56(2):177–185 6. Balsam J et al (2014) Thousand-fold fluores-
2. Ng OT et al (2012) Accuracy and user- cent signal amplification for mHealth diagnos-
acceptability of HIV self-testing using an oral tics. Biosens Bioelectron 51:1–7
fluid-based HIV rapid test. PLoS One 7. Istepanian R, Jovanov E, Zhang YT (2004)
7(9):e45168 Introduction to the special section on
3. Hay Burgess DC, Wasserman J, Dahl CA M-Health: beyond seamless mobility and
(2006) Global health diagnostics. Nature global wireless health-care connectivity. IEEE
444(Suppl 1):1–2 Trans Inf Technol Biomed 8(4):405–414
4. Urdea M et al (2006) Requirements for high 8. Mudanyali O et al (2012) Integrated rapid-
impact diagnostics in the developing world. diagnostic-test reader platform on a cellphone.
Nature 444(Suppl 1):73–79 Lab Chip 12(15):2678–2686
Biomarker Enrichment 83
38. Hsu YH (1984) Immunogold for detection of 41. Frens G (1973) Controlled nucleation for reg-
antigen on nitrocellulose paper. Anal Biochem ulation of particle-size in monodisperse gold
142(1):221–225 suspensions. Nat Phys Sci 241(105):20–22
39. Nash MA et al (2010) “Smart” diblock copo- 42. Khlebtsov NG (2008) Determination of size
lymers as templates for magnetic-core gold- and concentration of gold nanoparticles
shell nanoparticle synthesis. Nano Lett from extinction spectra. Anal Chem 80(17):
10(1):85–91 6620–6625
40. Nash MA et al (2010) Laboratory-scale protein 43. Haiss W et al (2007) Determination of size
striping system for patterning biomolecules and concentration of gold nanoparticles
onto paper-based immunochromatographic from UV-vis spectra. Anal Chem 79(11):
test strips. Lab Chip 10(17):2279–2282 4215–4221
Chapter 6
Abstract
This chapter describes the development of microfluidic toner-based analytical devices (μTADs) to perform
clinical diagnostics using a scanner or cell-phone camera. μTADs have been produced in a platform com-
posed of polyester and toner by the direct-printing technology (DPT) in a matter of minutes. This technol-
ogy offers simplicity and versatility, and it does not require any sophisticated instrumentation. Toner-based
devices integrate the current generation of disposable analytical devices along paper-based chips. The cost
of one μTAD has been estimated to be lower than $0.10. In addition, these platforms are lightweight and
portable thus enabling their use for point-of-care applications. In the last 5 years, great efforts have been
dedicated to spread out the use of μTADs in bioassays. The current chapter reports the fabrication of
printed microplates and integrated microfluidic toner-based devices for dengue diagnostics and rapid colo-
rimetric assays with clinically relevant analytes including cholesterol, triglycerides, total proteins, and glu-
cose. The use of μTADs associated with cell-phone camera may contribute to the health care, in special, to
people housed in developing regions or with limited access to clinics and hospitals.
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_6, © Springer Science+Business Media New York 2015
85
86 Karoliny Almeida Oliveira et al.
2 Materials
2.3 ELISA 1. Commercial kit (Bioeasy) for Dengue IgM Capture ELISA
Procedures and anti-dengue IgG solution for indirect ELISA were kept at
4–8 °C.
2. Anti-human IgG: 20 μg/mL IgG solution dissolved in carbon-
ate buffer at pH 9.6.
3. Dengue antigen solution: dengue antigen diluted in carbonate
buffer (1:16 v:v) at pH 9.6.
4. Serum samples from patients infected and non-infected with
dengue virus were supplied by a local Tropical Diseases
Hospital (HDT, Goiânia, GO, Brazil). The serum samples
were kept at −80 °C.
88 Karoliny Almeida Oliveira et al.
2.4 Clinical 1. Cellulose paste: cellulose powder and ultrapure water (1:4 m:m).
Diagnostics 2. Solution 1: 0.6 M potassium iodide and 0.3 M trehalose dis-
solved in 100 mM phosphate buffer at pH 6.0.
3. Solution 2: five parts of 181 U/mg glucose oxidase and one
part of 73 U/mg horseradish peroxidase.
4. Solution 3: 0.5 mM sodium cholate, 0.5 mM 4-aminoantipyrine,
300 U of cholesterol esterase, 204 U cholesterol oxidase,
828 U of horseradish peroxidase, and 35 mM piperazine-
N,N′-bis(2-ethanesulfonic acid) (PIPES) buffer at pH 7.0.
5. Solution 4: 0.12 mM Coomassie brilliant blue dye.
6. Solution 5: 6 mM 4-chlorophenol, 5 mM magnesium chloride,
0.75 mM 4-aminoantipyrine, 0.9 mM ATP, 132 kU of lipase,
1.5 kU glycerokinase, 4.02 kU glycerol 3-phosphate oxidase,
282 U of peroxidase and, 50 mM PIPES buffer at pH 7.5.
3 Methods
b
a
c 8
d
6
Height (mm)
2
toner barrier width
0
-2
0.0 0.5 1.0 1.5 2.0
Distance (mm)
Fig. 1 Representation of the (a) fabrication scheme of microplates based on laser-printing step, (b) layout of a
toner-based 96-zone microplate for immunoassays, (c) toner barrier profile, and (d) typical printed microplate
with sample added in test zones. (Reproduced with permission from ref. [13]. Copyright 2013The Royal Society
of Chemistry)
3.2 Fabrication 1. The fabrication of μTADs basically involves three steps related
of μTADs to (a) the drawing step, where the μTAD layout is designed,
(b) the laser printing which deposits a toner layer on the poly-
ester surface to form the channels, and (c) the sealing stage to
promote the bonding of two or more pieces of printed
channels.
2. During the drawing stage in a graphic software (see Note 1),
the colors black and white are often used to define the areas to
represent the microfluidic walls and channels, respectively.
When the file is sent to the printer, the black area will form a
toner layer with thickness about 7 μm. Oppositely, the laser
90 Karoliny Almeida Oliveira et al.
printer ideally does not deposit toner for the white lines or
reservoirs (also called of detection zones) previously defined in
the desirable layout. Consequently, the microchannels and
detection zones are easily created with a simple laser printing
step.
3. Besides the printed layout, its mirror image is also printed dur-
ing the same step to increase the channel depth.
4. A cut-through polyester film, with the same microfluidic struc-
ture from the layout, has been prepared by using a CO2 laser
ablation machine (see Note 2) and added between both printed
images in order to enhance the channel depth. This strategy
has been adopted to ensure the fluidic transport by capillary
action through printed channels.
5. Before laminating the polyester pieces, the detection zones in
the upper polyester piece were perforated with a paper punch
to have access the microfluidic channels.
6. Afterwards, the three polyester pieces were aligned and ther-
mally laminated at 150 °C in a laminator machine (see Note 3).
Figure 2 shows the 3D and cross-section views of a μTAD.
7. Finally, a paste made of cellulose powder and ultrapure water
was added to each test zone and allowed to dry at room tem-
perature during 30 min (see Note 4). The layout of the pro-
posed device (35 mm × 35 mm) consisted of four test zones
interconnected by microfluidic channels and one central inlet
zone to sample distribution (Fig. 2). All channels were 10-mm
long, 1-mm wide and ca. 100-μm deep. Figure 3 displays a real
image of a μTAD in comparison with the size of a quarter dol-
lar coin.
Fig. 2 Simplified fabrication process of toner-based microfluidic devices for clinical assay in (a) 3D and (b)
cross-sectional views and (c) layout of a typical device used to demonstrate the capability of performing colo-
rimetric assays. Reprinted with permission from ref. [12]. Copyright 2012 American Chemical Society
Fig. 3 Optical micrograph showing a real image of a μTAD in comparison with the
size of a quarter dollar coin
92 Karoliny Almeida Oliveira et al.
b 5
0
(1) (2)
0 5 10 15 20 25 30 35 40
Time (s)
Fig. 4 Examples of (a) the experimental arrangement for the determination of the flow rate magnitude and
(b) typical conductivity signal monitored over time. In (a), the labels E0 and E1 mean the excitation and
received electrodes, respectively. In (b), the labels (1) and (2) indicate the injection time and the time required
for an aqueous solution move from the injection point to the detector, respectively
3.4.1 Capture ELISA 1. The anti-human IgM antibodies were provided by a local labo-
ratory of monoclonal antibodies production (CEPRACO,
Goiânia, GO, Brazil). First, anti-human IgM was immobilized
on microzones using 10-μL aliquots. The microplate was kept
overnight at 4–8 °C (see Note 5).
2. Afterwards, the microplate was washed three times with PBS-T
solution and dried at room temperature.
3. Each printed zone was individually blocked by adding 20 μL of
blocking buffer during 2 h at 37 °C and washed again with
PBS-T solution.
4. Then, 10-μL aliquots of diluted samples and controls were
added to each zone. The microplate was incubated at 37 °C
during 1 h and washed with buffer.
5. After the drying step, 10-μL aliquots of the lyophilized mix-
ture containing dengue antigen and conjugated anti-dengue
were added to each zone and kept during 1 h at 37 °C.
6. Lastly, the microplate was washed and aliquots of 10 μL of the
substrate solution were added to each zone. After keeping
10 min at room temperature, the assay was stopped by the
addition of 10 μL/zone of stop solution.
3.4.2 Indirect ELISA 1. Firstly, the dengue antigen was immobilized on microzones
Procedures using 10-μL aliquots and kept overnight at 2–8 °C. The stages
associated with the washing, blocking and addition of sam-
ples/controls were performed similarly to the steps described
for the capture ELISA assay (Subheading 3.4.1).
2. Afterwards, the anti-human IgG (10-μL aliquots) was added
on each zone and kept at 37 °C during 1 h.
3. Then, 10-μL aliquots containing the peroxidise-conjugated sec-
ondary antibody (rabbit anti-mouse IgG) diluted in blocking
buffer were added to each zone and kept at 37 °C during 1 h.
4. Finally, the zones were washed and aliquots of 10 μL of the
substrate solution were added to each zone as described in the
capture ELISA procedure.
3.4.3 Simultaneous The procedure to carry out simultaneous colorimetric tests for
Colorimetric Tests clinical glucose, cholesterol, proteins, and triglycerides were quite
on μTADs similar. The μTADs were prepared to perform the bioassays by
adding the specific color reagents in different detection zones, as
described below
1. For glucose assay, the detection zone was first spotted with
8 μL of solution 1 (Subheading 2.4) and dried at room tem-
perature during 10 min. Afterwards, the zone was spotted with
8 μL of solution 2 (Subheading 2.4) and dried for 10 min dur-
ing additional 10 min.
94 Karoliny Almeida Oliveira et al.
3.5 Detection A cell-phone camera was used to measure the resulting colorimetric
System information related to the detection of IgM and IgG in samples
from patients infected with dengue virus on printed microplates. On
the other hand, the colorimetric detection of glucose, cholesterol,
proteins, and triglycerides on integrated μTADs was performed with
the scanner mode of a Deskjet multifunction printer.
1. The images captured with the cell-phone camera may be digi-
tally filtered using software to minimize interferences associ-
ated light brightness and scattering.
2. For the detection of cholesterol, glucose, proteins, and triglyc-
erides using the scanner, μTADs were placed and fixed on the
glass of scanner. In this arrangement, the image was captured
in the bottom part of the device.
3. The images recorded with both cell-phone camera and scanner
were converted to a CMYK color scale in graphic software. For
the color developed inside each zone, the arithmetic mean of
pixel intensity (Fig. 6) was used to quantify the concentration
of the analytes (see Note 6).
Fig. 6 Example of the graphic tool usually explored to extract the mean color intensity. This information is
required to quantify the analyte concentration in samples
Disposable Devices for Clinical Diagnostics 95
a
70
60
Color Intensity
50
40
30
20
10
0
B NC PC PS NS
Test samples
b
120
100
Color Intensity
80
60
40
20
B NC PC PS NS
Test samples
Fig. 7 ELISA data recorded with cell phone camera showing the detection of
(a) IgM and (b) IgG based on IgM capture and indirect ELISA procedures, respec-
tively. The labels B, NC, and PC mean blank sample, negative control, and posi-
tive control, respectively. Positive (PS) and negative (NS) samples are related to
infected and non-infected patients. The captured images are inset in the figures
96 Karoliny Almeida Oliveira et al.
3.6.2 Clinical Assays 1. μTADs have been explored to detect glucose, total proteins,
with Artificial Sera cholesterol, and triglycerides in artificial sera samples. Glucose
Samples is the main indicator of diabetes in biological fluids like urine,
serum and blood. The levels of total protein can be helpful to
diagnose renal diseases or disorders. On the other hand, the
concentration levels of cholesterol and triglycerides can be
indicative factors of coronary risks. Based on the high impor-
tance of all analytes mentioned, the capability of μTADs per-
form simultaneous assays can contribute to an early diagnostic
of the patient
2. Before demonstrating the simultaneous analysis, each bioassay
was separately performed in a single device. In this case, all
four detection zones were prepared according to the proce-
dures described in Subheading 3.2.
3. The use of four detection zones has allowed investigating the
zone-to-zone repeatability for each assay. The standard relative
deviation (RSD) values for each assay have been lower than
8 %. Based on the reaction time optimization, the images
should be captured after 10 min to obtain reliable analytical
concentrations.
4. Once demonstrated good reproducibility and high accuracy
for individual assays, the simultaneous tests for cholesterol,
Disposable Devices for Clinical Diagnostics 97
4 Notes
References
1. Arora A, Simone G, Salieb-Beugelaar GB, Kim fabrication of passive micromixers microfluidic
JT, Manz A (2010) Latest developments in devices using a direct-printing process. Lab
micro total analysis systems. Anal Chem Chip 5:974–978
82:4830–4847 9. Yu H, Lu Y, Zhou YG, Wang FB, Hi FY, Xia
2. Chen Y, Pépin A (2001) Nanofabrication: XH (2008) A simple disposable microfluidic
conventional and nonconventional methods. devices for rapid protein concentration and
Electrophoresis 22:187–207 purification via direct-printing. Lab Chip
3. do Lago CL, da Silva HDT, Neves CA, Brito- 8:1496–1501
Neto JGA, da Silva JAF (2003) A dry process 10. Duarte GRM, Coltro WKT, Borba JC, Price
for production of microfluidic devices based CW, Landers JP (2012) Disposable polyester-
on the lamination of laser-printed polyester. toner electrophoresis microchips for DNA
Anal Chem 75:3853–3858 analysis. Analyst 137:2692–2698
4. Gabriel EFM, do Lago CL, Gobbi AL, Carrilho 11. Duarte GRM, Price CW, Augustine BH,
E, Coltro WKT (2013) Characterization of Carrilho E, Landers JP (2011) Dynamic solid
microchip electrophoresis devices fabricated by phase DNA extraction and PCR amplification
direct-printing process with colored toner. in polyester-toner based microchip. Anal
Electrophoresis 34:2169–2176 Chem 83:5182–5189
5. Coltro WKT, de Jesus DP, da Silva JAF, Do 12. de Souza FR, Alves GL, Coltro WKT (2012)
Lago CL, Carrilho E (2010) Toner and paper- Capillary-driven toner-based microfluidic
based fabrication techniques for microfluidic devices for clinical diagnostics with colorimet-
applications. Electrophoresis 31:2487–2498 ric detection. Anal Chem 84:9002–9007
6. Coltro WKT, Carrilho E (2008) Low-cost 13. Oliveira KA, de Oliveira CR, da Silveira LA,
technologies for microfluidics applications. In: Coltro WKT (2013) Laser-printing of toner-
Landers JP (ed) Handbook of capillary and based 96-microzone plates for immunoassays.
microchips electrophoresis and associated Analyst 138:1114–1121
microtechniques, 3rd edn. New York: CRC 14. Kim AR, Kim JY, Choi K, Chung DS (2013)
Press, pp 1169–1184 On-chip immunoassay of a cardiac biomarker
7. Gabriel EFM, Duarte GF, Garcia PT, de Jesus in serum using a polyester-toner microchip.
DP, Coltro WKT (2012) Polyester-toner elec- Talanta 109:20–25
trophoresis microchips with improved analytical 15. Coltro WKT, Lima RS, Segato TP, Carrilho E,
performance and extended lifetime. de Jesus DP, do Lago CL, da Silva JAF (2012)
Electrophoresis 33:2660–2667 Capacitively coupled contactless conductivity
8. Liu AL, He FY, Wang K, Zhou T, Lu Y, Xia detection on microfluidic systems-ten years of
XH (2005) Rapid method for design and development. Anal Methods 4:24–33
Chapter 7
Abstract
mHeath technologies are recognized to play important roles in the future of personal care and medicine.
However, their full potentials have not been reached, as most of current technologies are restricted to
monitoring physical and behavioral parameters, such as body temperature, heart rate, blood pressure, and
physical movement, while direct monitoring of biomarkers in body fluids can provide much more accurate
and useful information for medical diagnostics. A major barrier to realizing the full potential of mHealth
is the high costs and long cycles of developing mHealth devices capable of monitoring biomarkers in body
fluids. To lower the costs and shorten the developmental cycle, we have demonstrated the leveraging of
the most successful portable medical monitoring device on the market, the blood glucose meter (BGM),
with FDA-approved smartphone technologies that allow for wireless transmission and remote monitoring
of a wide range of non-glucose targets. In this protocol, an aptamer-based assay for quantification of
interferon-γ (IFN-γ) using an off-the-shelf BGM is described. In this assay, an aptamer-based target rec-
ognition system is employed. When IFN-γ binds to the aptamer, it triggers the release of a reporter
enzyme, invertase, which can catalyze the conversion of sucrose (not detected by BGM) to glucose. The
glucose being produced is then detected using a BGM. The system mimics a competitive enzyme-linked
immunosorbent assay (ELISA), where the traditional immunoassay is replaced by an aptamer binding
assay; the reporter protein is replaced by invertase, and finally the optical or fluorescence detector is
replaced with widely available BGMs.
Key words Blood glucose meter (BGM), Aptamer, Biosensor, Point-of-care diagnostic, Electrochemical
sensor
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_7, © Springer Science+Business Media New York 2015
99
100 Tian Lan et al.
Fig. 1 Design of the aptamer based assay for detection of IFN-γ using a BGM. The assay system is composed
of a DNA-modified invertase (blue DNA sequence), hybridized with an IFN-γ aptamer (black DNA sequence),
which also hybridizes with a biotin modified DNA (green DNA sequence) that is bound to streptavidin-coated
magnetic beads (brown sphere ). In the absence of IFN-γ, complementary DNA hybridization between the three
DNA sequences keeps the enzyme invertase bound on the magnetic beads. Upon addition of IFN-γ, the binding
of IFN-γ to aptamer results in the dehybridization of thiol-modified DNA, leading to the release of invertase
from the magnetic beads. The released invertase can be separated from the remaining bead-bound invertase
by magnetic separation. The released invertase is then used to produce glucose for BGM detection, from
sucrose (which is undetectable by BGM)
2 Materials
3 Methods
Table 1
DNA sequences used in the study
Fig. 2 Conjugation chemistry for covalently attachment of a thiol-modified DNA to invertase. The primary group
on the surface of invertase is first reacted with a bifunctional linker (Sulfo-SMCC), which contains an amine-
reactive N-hydroxysuccinimide ester moiety and a thiol-reactive maleimide group. The invertase-SMCC is then
reacted with the thiol-modified DNA to complete this step of conjugation
Fig. 3 Representative data of IFN-γ detection in buffer, with human serum albumin as negative control (a); and
in 20 % human serum (b)
BGM for Protein Detection 107
4 Notes
Acknowledgements
References
1. mHealth G. Device Listing (2012) A global list 10. Nie Z, Deiss F, Liu X, Akbulut O, Whitesides
of commercially available devices in the health- GM (2010) Integration of paper-based micro-
care sector, updated fortnightly 2012. http:// fluidic devices with commercial electrochemi-
www.gsma.com/connectedliving/wp-content/ cal readers. Lab Chip 10(22):3163–3169,
uploads/2012/04/22-November12_ PMCID: 3060706
MobileHealth_Device_Listing.pdf 11. Su J, Xu J, Chen Y, Xiang Y, Yuan R, Chai Y
2. Chin CD, Cheung YK, Laksanasopin T, (2012) Personal glucose sensor for point-of-
Modena MM, Chin SY, Sridhara AA et al care early cancer diagnosis. Chem Commun
(2013) Mobile device for disease diagnosis and (Camb) 48(55):6909–6911
data tracking in resource-limited settings. Clin 12. Yan L, Zhu Z, Zou Y, Huang Y, Liu D, Jia S
Chem 59(4):629–640 et al (2013) Target-responsive “sweet” hydro-
3. iBG STAR, 510(k) Substantial Equivalence gel with glucometer readout for portable and
Determination Decision Summary (510(k) quantitative detection of non-glucose targets.
number: k103544) J Am Chem Soc 135(10):3748–3751
4. iBGSTAR (2011) iBGStar® blood glucose 13. Boehm U, Klamp T, Groot M, Howard JC
meter. Sanofi (updated 2011; cited 2011 Dec (1997) Cellular responses to interferon-
1) http://www.bgstar.com/web/ibgstar gamma. Annu Rev Immunol 15:749–795
5. Telcare (2013) Telcare (updated 2013; cited 14. Liu Y, Kwa T, Revzin A (2012) Simultaneous
2013 09/09) https://www.telcare.com/ detection of cell-secreted TNF-alpha and IFN-
6. Hones J, Muller P, Surridge N (2008) The gamma using micropatterned aptamer-modified
technology behind glucose meters: test strips. electrodes. Biomaterials 33(30):7347–7355
Diabetes Technol Ther 10:S10–S26 15. Liu Y, Yan J, Howland MC, Kwa T, Revzin A
7. Xiang Y, Lu Y (2011) Using personal glucose (2011) Micropatterned aptasensors for con-
meters and functional DNA sensors to quan- tinuous monitoring of cytokine release from
tify a variety of analytical targets. Nature Chem human leukocytes. Anal Chem 83(21):8286–
3(9):697–703, PMCID: 3299819 8292, PMCID: 3235337
8. Xiang Y, Lu Y (2012) Using commercially 16. Lee PP, Ramanathan M, Hunt CA, Garovoy
available personal glucose meters for portable MR (1996) An oligonucleotide blocks
quantification of DNA. Anal Chem interferon-gamma signal transduction.
84(4):1975–1980, PMCID: 3302979 Transplantation 62(9):1297–1301
9. Xiang Y, Lu Y (2012) Portable and quantita- 17. Balasubramanian V, Nguyen LT,
tive detection of protein biomarkers and small Balasubramanian SV, Ramanathan M (1998)
molecular toxins using antibodies and ubiqui- Interferon-gamma-inhibitory oligodeoxynu-
tous personal glucose meters. Anal Chem cleotides alter the conformation of interferon-
84(9):4174–4178, PMCID: 3341531 gamma. Mol Pharmacol 53(5):926–932
BGM for Protein Detection 109
18. Tuleuova N, Jones CN, Yan J, Ramanculov 21. Acevedo B, Perera Y, Ruiz M, Rojas G, Benitez
E, Yokobayashi Y, Revzin A (2010) J, Ayala M et al (2002) Development and vali-
Development of an aptamer beacon for dation of a quantitative ELISA for the mea-
detection of interferon-gamma. Anal Chem surement of PSA concentration. Clin Chim
82(5):1851–1857 Acta 317(1–2):55–63
19. Stoltenburg R, Reinemann C, Strehlitz B 22. Vessella RL (1993) Trends in immunoassays of
(2007) SELEX–a (r)evolutionary method to prostate-specific antigen: serum complexes
generate high-affinity nucleic acid ligands. and ultrasensitivity. Clin Chem 39(10):
Biomol Eng 24(4):381–403 2035–2039
20. Wilson AB, McHugh SM, Deighton J, Ewan 23. Matsumoto K, Konishi N, Hiasa Y, Kimura E,
PW, Lachmann PJ (1993) A competitive inhi- Takahashi Y, Shinohara K et al (1999) A highly
bition ELISA for the quantification of human sensitive enzyme-linked immunoassay for
interferon-gamma. J Immunol Methods serum free prostate specific antigen (f-PSA).
162(2):247–255 Clin Chim Acta 281(1–2):57–69
Chapter 8
Abstract
Ovarian cancer is a leading cause of death from gynecologic cancers in the USA, and early diagnosis can
potentially increase 5-year survival rate. Detection of biomarkers derived from hyperplasia of epithelial tis-
sue by enzyme-linked immunosorbent assay (ELISA) proves to be a practical way of early diagnosis of
ovarian cancer. However, ELISA is commonly performed in a laboratory setting, and it cannot be used in
a clinical setting for on-site consultation. We have shown a microchip ELISA that detects HE4, an ovarian
cancer biomarker, from urine using a cell phone integrated with a mobile application for imaging and data
analysis. In microchip ELISA, HE4 from urine was first absorbed on the surface; the primary and second-
ary antibodies were subsequently anchored on the surface via immuno-reaction; and addition of substrate
led to color development because of enzymatic labeling. The microchip after color development was
imaged using a cell phone, and the color intensity was analyzed by an integrated mobile application. By
comparing with an ELISA standard curve, the concentration of HE4 was reported on the cell phone
screen. The presented microchip ELISA coupled with a cell phone is portable as opposed to traditional
ELISA, and this method can facilitate the detection of ovarian cancer at the point-of-care (POC).
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_8, © Springer Science+Business Media New York 2015
111
112 ShuQi Wang et al.
Fig. 1 Workflow of microchip ELISA coupled with cell phone detection (Reproduced from ref. 5 with permis-
sion from The Royal Society of Chemistry). (a) 100 µL of urine sample containing HE4 was pipetted into a
microchannel. (b) Formation of an indirect ELISA immune-complex on a microchip. (c) Blue color with varying
intensities was developed in microchannels and imaged using a cell phone. (d) The blue color (marked with
rectangles) developed in microchannels was auto-recognized by a mobile application, and R pixel values were
reported. The R pixel value of background was used for normalization. The concentration of HE4 in urine was
calculated with a reference to the standard curve and then reported on the screen
Microchip ELISA Coupled with Cell Phone to Detect Ovarian Cancer HE4 Biomarker… 113
2 Materials
3 Methods
Fig. 2 (a) Assembly of a microchip with three straight microchannels on a polystyrene petri dish. (b) Actual
image of an assembled microchip with color development. Adapted from ref. [5] with permission from The
Royal Society of Chemistry
3.2 Construction To avoid the interference of external light, we built a cell phone-
of a Black Box for Cell imaging instrument (Fig. 3). This setup consisted of one black
Phone Imaging acrylic box, one imaging stage, and an imaging hole on the top.
1. To provide internal light, two LEDs were installed under the
imaging stage.
2. One layer of PMMA (1.5 mm thick) was mounted as the top of
the stage.
3. One piece of white print paper was attached to the bottom of
the PMMA layer to provide a white background.
3.3 Microchip ELISA 1. Pure HE4 peptide antigen was serially twofold diluted in
sodium bicarbonate (0.1 M, pH 9.7) to obtain quantification
standards (1,250, 625.0, 312.5, 156.3, 78.1, 39.1, and
19.5 ng/mL).
2. 100 μL of each HE4 quantification standard was injected into
a microchannel and incubated at room temperature for 1 h
(see Note 3).
Microchip ELISA Coupled with Cell Phone to Detect Ovarian Cancer HE4 Biomarker… 115
Fig. 3 (a) Design of a black box for cell phone imaging. (b) Actual image of the black box. Adapted from ref.
[5] with permission from The Royal Society of Chemistry
Fig. 4 Validation of on-chip HE4 ELISA compared to conventional 96-well microplate ELISA (Adapted from
ref. 5 with permission from The Royal Society of Chemistry). (a) The standard curve obtained from microchip
ELISA was compared with that obtained from a 96-well microplate ELISA. The absorbance of the resultant
color solution obtained from both methods was measured using a spectrophotometer (BioTek, Winooski, VT) at
a wavelength of 450 nm. (b) The standard curve of HE4 microchip ELISA. R pixel values were measured using
a mobile application, which was correlated with the color intensity obtained at varying concentrations. Data
are presented as mean ± standard deviation (n = 8)
3.4 Microplate ELISA As a standard method, conventional 96-well microplate ELISA was
for Detection of HE4 also performed. The testing procedure was the same except for the
follow steps:
1. The washing steps were performed using a plate-washer.
2. The incubation of TMB in microwells was 15 min prior to addi-
tion of 100 μL of stop solution.
3. The readout of absorption (optical density, OD) was measured
using a microplate reader at a wavelength of 450 nm.
3.5 Quantitative 1. The microchip with blue color development at the end of 9-min
Image Processing by incubation was placed in the black box as shown in Fig. 3.
Matlab 2. A cell phone (Sony Ericson i790) with a 3.2 megapixel camera
was used to image the on-chip color development through the
hole on top of the black box.
3. The images were exported to a computer, and the manually
cropped image regions with color development were analyzed
using a customized MATLAB (MathWorks, Natick, MA) code
(Table 1). Red, green and blue pixel values were reported within
seconds as mean value ± standard deviation.
4. Red (R) pixel values were used for data analysis, since they cor-
related well with the concentrations of HE4 loaded to micro-
channels (Fig. 4b).
Microchip ELISA Coupled with Cell Phone to Detect Ovarian Cancer HE4 Biomarker… 117
Table 1
The Matlab code for image analysis
for i=1:34
str=strcat(int2str(i),'.jpeg');
I=imread(str);
Ired=I;
Igreen=I;
Iblue=I;
Ired(:,:,2)=[];
Ired(:,:,2)=[];
Igreen(:,:,1)=[];
Igreen(:,:,2)=[];
Iblue(:,:,1)=[];
Iblue(:,:,1)=[];
r=mean2(Ired);
g=mean2(Igreen);
b=mean2(Iblue);
std=std2(Iblue);
rgb(i,1)=r;
rgb(i,2)=g;
rgb(i,3)=b;
rgb(i,4)= std;
end
rgb
xlswrite('rgb.xls',rgb);
3.6 Quantification Microchip ELISA was first validated by comparing to 96-well plate
of Clinical Samples ELISA. The blue color solution from microchannels was trans-
with Unknown ferred to a 96-well plate using a pipet. The optical density of each
Concentrations of HE4 well was measured using a spectrophotometer and the readouts
were plotted as shown in Fig. 4a. The result indicated that the
microchip ELISA can be used for HE4 quantification. To elimi-
nate the use of a spectrophotometer, we developed a cell-phone
based imaging and analysis system. By measuring the red pixel
118 ShuQi Wang et al.
Fig. 5 Mobile application for image processing and data report. (a) Automatic selection of channel region for
image processing. (b) Batch processing to report R pixel values versus the concentrations of HE4 (quantifica-
tion standards). (c) Plot of standard curve for microchip ELISA
Table 2
Comparison of Red pixel values obtained by MATLAB and mobile application
Note: The detection limit was 19.5 ng/mL. For each standard point, we performed
three replicates. Reproduced from ref. [5] with permission from The Royal Society of
Chemistry
4 Notes
1. The power of the laser cutter used for cutting PMMA was
75 % at a speed of 7 %.
2. The power of the laser cutter used for cutting double-sided
adhesive was 10 % at a speed of 6 %.
3. Microchips were wrapped with Parafilm during incubation
steps wherever applied to avoid evaporation.
4. Homogeneous color distribution was achieved by pipetting
the blue solution inside the microchannel back and forth for a
couple of times. Generation of air bubbles should be avoided.
5. Homogeneous color distribution was obtained prior to cell
phone imaging.
6. One hundred microliters of sample or reagent was a standard
volume as a comparison to the control method, i.e., 96-well
plate ELISA. Compared to smaller volumes, this volume was
more tolerant to sampling errors and allowed us to collect
more reliable blue color images.
7. The quantification standards were serially diluted in sodium
bicarbonate to construct the standard curve for quantifying
the concentration of HE4 in urine. Although this sample pro-
cessing may cause a systemic variation in terms of absolute
quantification, it would not affect the comparison results
between healthy and cancerous urine samples using a defined
threshold. Nevertheless, a pooled negative urine sample would
be ideal to dilute quantification standards to reduce the varia-
tion of quantification.
8. For validation, the blue solution was transferred to an
Eppendorf tube using a pipette. The blue solution was stopped
using 100 μL of 1 M H2SO4 and subsequently measured using
a microplate reader at a wavelength of 450 nm.
9. Standard images must to be loaded in an order starting from
higher concentrations to lower ones for regions to be assigned
to the right concentration values.
10. The application reports R values and concentrations for each
selected image.
Acknowledgments
References
1. Clarke-Pearson DL (2009) Clinical practice. 7. Moon S et al (2009) Integrating microfluid-
Screening for ovarian cancer. N Engl J Med ics and lensless imaging for point-of-care
361(2):170–177 testing. Biosens Bioelectron 24(11):
2. Visintin I et al (2008) Diagnostic markers for 3208–3214
early detection of ovarian cancer. Clin Cancer 8. Hellstrom I et al (2010) Detection of the HE4
Res 14(4):1065–1072 protein in urine as a biomarker for ovarian neo-
3. Wang S, Xu F, Demirci U (2010) Advances in plasms. Cancer Lett 296(1):43–48
developing HIV-1 viral load assays for resource- 9. Wang S et al (2014) Micro-a-fluidics ELISA
limited settings. Biotechnol Adv 28(6):770–781 for rapid CD4 cell count at the point-of-care.
4. Wang S et al (2013) Point-of-care assays for Sci Rep 4:3796
tuberculosis: role of nanotechnology/micro- 10. Cramer DW et al (2011) Ovarian cancer bio-
fluidics. Biotechnol Adv 31(4):438–449 marker performance in prostate, lung, colorec-
5. Wang S et al (2011) Integration of cell phone tal, and ovarian cancer screening trial
imaging with microchip ELISA to detect ovar- specimens. Cancer Prev Res (Phila) 4(3):
ian cancer HE4 biomarker in urine at the 365–374
point-of-care. Lab Chip 11(20):3411–3418 11. Zhu CS et al (2011) A framework for evaluat-
6. Kim YG et al (2009) Quantum dot-based HIV ing biomarkers for early detection: validation
capture and imaging in a microfluidic channel. of biomarker panels for ovarian cancer. Cancer
Biosens Bioelectron 25(1):253–258 Prev Res (Phila) 4(3):375–383
Chapter 9
Abstract
The sparse cells that are shed from tumors into peripheral circulation are an increasingly promising resource
for noninvasive monitoring of cancer progression, early diagnosis of disease, and serve as a tool for improv-
ing our understanding of cancer metastasis. However, the extremely sparse concentration of circulating
tumor cells (CTCs) in blood (~1–100 CTC in 7.5 mL of blood) as well as their heterogeneous biomarker
expression has limited their detection using conventional laboratory techniques. To overcome these chal-
lenges, we have developed a microfluidic chip-based micro-Hall detector (μHD), which can directly mea-
sure single, immunomagnetically tagged cells in whole blood. The μHD can detect individual cells even in
the presence of vast numbers of blood cells and unbound reactants, and does not require any washing or
purification steps. Furthermore, this cost-effective, single-cell analytical technique is well suited for minia-
turization into a mobile platform for low-cost point-of-care use. In this chapter, we describe the methodol-
ogy used to design, fabricate, and apply these chips to cancer diagnostics.
Key words Hybrid microfluidic, Microelectronic chips, BioMEMS, Sensors, Microfluidics, Magnetic
nanomaterials
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_9, © Springer Science+Business Media New York 2015
123
124 David Issadore
Table 1
Comparison of soluble proteins, circulating nucleic acid, and circulating tumor
cells (CTCs) for cancer monitoring
Fig. 1 Summary of our detection method. (a) Our hybrid chip consists of a GaAs substrate with microfabricated
Hall sensors. Built directly on top of the semiconductor chip is a microfluidic network to controllably deliver
cells to the sensors. The microfluidics are fabricated using soft-lithography and are permanently bonded to the
GaAs. (b) Circulating tumor cells are magnetically labeled based on a panel of relevant cancer biomarkers
(EGFR, HER2/neu, etc.) and magnetic nanoparticles (MNPs). The cells are passed one-by-one via hydrody-
namic flow structures over μHall sensors for highly sensitive detection
2 Materials
2.1 μHall Chip The sensors were built on a pseudomorphic high electron mobility
Farbrication GaAs wafer using standard semiconductor processing (Fig. 2a).
A mesa was etched onto the GaAs wafer heterostructure (Fig. 2b).
The mesa was defined using photolithography and followed by an
Rare Cell Cancer Diagnostics 127
Fig. 2 Summary of our fabrication method. The cartoons on the left are side-view
schematics of the chip in the various stages of the fabrication (a–g). The images
on the right are schematics of the top-view. (g) A micrograph of the finished
micro-Hall chip. The scale bar is 250 μm. (h) A cross-sectional schematic of the
instrumentation that connect to the μHall chip (i) A photograph of the slot-
connector/magnet holder, where the μHD chip sits. A shielded ribbon cable
connects the output of the μHD to the custom printed circuit board
128 David Issadore
Fig. 3 Electronics to process the micro-Hall signal. (a) The hall resistance R was measured versus the applied
magnetic field B, and a sensitivity of 78 Ω/T was calculated. (b) The input-referred noise of the μHall sensors
as measured with a spectrum analyzer. (c) A schematic of the electronics to condition the μHall signal prior to
software analysis. Each pair of μHall sensors are driven with a current source. The differential output of each
μHall sensor is AC coupled to a two-stage amplifier (gain = 30 × 30) before analog-to-digital conversion
ADC. Once digitized the signal is sent to either a computer or mobile device for software analysis
2.3 Magnetic The μHD is designed to detect magnetically labeled cellular tar-
Nanomaterials gets. By targeting cells with MNPs and subjecting them to an
external magnetic field B, each cell acquired a magnetic moment m
that is directly proportional to both the number of biomarkers
N and the magnetic moment of the MNPs mp (m = N × mp).
The induced magnetic fields of these cells are then measured using
the microfabricated Hall sensors.
130 David Issadore
2.4 List of Materials Here, we list the materials used in the construction of the μHD. The
list is organized into the following sections: micro-Hall (μHall)
chip, magnetic labeling reagents, and auxiliary electronics.
3 Methods
3.1 Finite Element To aid in the design and characterization of our microfabricated
Simulations: Magnetic Hall sensors, we constructed a numerical model to describe the
spatial response of the sensors to magnetically labeled cells
(Matlab). The model treats each cell as a magnetic dipole moment
mtot located at the center of the cell, aligned with the external mag-
netic field in the z direction (Fig. 4a). The Hall voltage
(VH = I × RH × B⊥), where I is the input current to the sensor and RH
is the characteristic Hall resistance of the device, is calculated by
analytically solving for the demagnetization field B, and averaging
B⊥ over the sensing area of the Hall element. This numerical model
was used to determine the optimum sensor size that maximizes
signal-to-noise ratio (SNR) (Fig. 4b). We found that for cells with
an average diameter of 12 μm, the SNR was maximal when the
sensors had a detection area similar to that of the cells (8 × 8 μm2).
The spatial response of the Hall voltage VH was also simulated.
As the dipole was moved away from the sensor surface, the VH
signal steeply declined (Vh∝d−3) (Fig. 4c) Our numerical simula-
tion showed that the VH was >100-fold larger for a cell brought to
the sensor surface (d = 4 μm) than for a cell placed at the center of
the microfluidic channel (d = 7.5 μm), thus motivating accurate
positioning of the cell to the bottom of the channel with hydrody-
namic focusing.
132 David Issadore
Fig. 4 Summary of our detection method. (a) A computer model was used to design and optimize the μHall
chip. The software calculates the magnetic field B⊥ normal to the chip’s surface, which is produced by a
magnetic dipole at a specific position (x, y, z). (b) VH is a function of the sensor size (w, l) for a magnetic dipole
at a height d = 4 μm above the chip surface. (c) Simulation of the normalized VH as a function of the height of
a magnetic dipole at (x, y) = (0, 0). The signal strength was found to decay rapidly with distance (~d−3) from the
sensor surface
3.2 Finite Element To controllably stream cells over the μHall sensors, we design a
Simulations: Fluidic two-stage flow focusing structure (Fig. 5a). Cells are directed lat-
erally towards the center of the fluid channel via coplanar sheath
flows. Cells are pushed vertically toward the bottom of the chan-
nel using hydrodynamic forces generated with Chevron patterned
cut-out of the roof of the channel. The physical channel was
500 μm wide and 30 μm high, and designed to operate under
very large flow rates (φ ~ 1 mL/h). The use of hydrodynamic
focusing allows the physical channel to be much larger than the
cells, which in turn lowers the fluidic resistance and reduce the
risk of channel clogging.
To design this two-stage flow focusing structure we utilized an
iterative finite element analysis (Comsol). A cross section of the
microfluidic channel after lateral flow focusing is plotted (Fig. 5a,
inset). The sample, which includes the cells, is focused laterally
towards the center of the channel. After the chevron pattern the
sample is pushed vertically towards the bottom of the channel
(Fig. 5a, inset). The initial design for the chevron pattern was
modeled after a previously reported design [18]. We modified this
design for our application by including chevrons only on the top of
the channel rather than on the top and bottom as was previously
done. In this way, our chip drives cells to the bottom of the chan-
nel rather than towards the center as was done by Howell et al.
After a process of iterative simulation and redesign, which led
to a design that brought the cells to the bottom-center of the
channel, experimental verification was done. The device was built
using standard two-layer SU-8 soft lithography. The sample flow
was stained red with rhodamine and the sheath flow green with
fluorescein, and flow was analyzed using a fluorescence microscope
(Fig. 5b). As the sheath flow rate increased relative to the sample
Rare Cell Cancer Diagnostics 133
Fig. 5 To enhance the μHall signal, a hydrodynamic focusing structure was used to bring individual cells close
to the μHall sensors. Cells entering the chip though the sample input are pushed towards the center of the
channel by the lateral sheaths and are pushed vertically to the bottom of the channel by the chevron patterns.
(a) The results of a finite element computer simulation. The sheath fluid is colored blue and the sample fluid in
colored red. (b) Fluorescence micrographs, demonstrating flow focusing. The sheath fluid was labeled with
fluorescein and the sample was labeled with rhodamine. As the sheath/sample flow rate ratio increased, the
sample flow was focused to an increasingly narrow region in the center of the chip
flow rate, the width of sample stream decreased. The ratio of the
flow rate between the sheath flow and the sample streams was con-
trolled using gravitational flow, with negative pressure provided by
a syringe pump on the output (BS8000, Braintree Scientific).
Typical total flow rates were 0.1–1 mL/h.
3.3 Circulating o demonstrate clinical use of the μHall chip, we detected rare cir-
Tumor Cell Detection culating tumor cells (CTCs) in whole blood. We conducted a small
study on a cohort of ovarian cancer patients (n = 20). Each of the
patients selected had advanced disease (Stage IIIc or IV), such that
we expected to find CTCs in each of their blood samples. As a
negative control, blood samples were obtained from healthy vol-
unteers (n = 15). To measure the performance of the μHall chip,
we directly compared its results against the clinical gold standard,
the CellSearch system.
134 David Issadore
3.4 Drug Response In addition to the detection of cancer via the enumeration of rare
Measurements tumor cells in blood, the μHall chip can also be used to sensitively
measure biomarker expression on individual cells. This ability
allows the μHall chip to profile biomarker expression on samples
with limited cell numbers, such as fine needle aspirates (FNAs), for
applications such as the monitoring of disease progression during
treatment. The single cell measurements enabled by the μHall chip
obviate the need for separate experiments to normalize against an
independent cell count. To demonstrate this capability, we used
the μHall chip to monitor drug treatment efficacy on human A431
epidermoid cancer cells xenografted onto mice. Tumor samples
were obtained through fine-needle aspiration (~1,800 tumor cells
per aspirate). The aspirates were then labeled with EGFR-specific
MNPs and processed for micro-Hall measurement (Fig. 7a).
For a proof-of-concept experiment, three mice with xeno-
grafted tumors received treatment with geldanamycin, a heat shock
Rare Cell Cancer Diagnostics 135
Fig. 6 Circulating tumor detection on a μHall chip. (a) The μHall chip was used to detect CTCs in blood samples
from patients with late stage ovarian cancer. Samples were magnetically labeled using four known cancer
biomarkers: EpCAM, HER2/neu, EGFR, and MUC1. (b) Patient samples (n = 20) were split into two and profiled
using both the μHall chip (top ) and CellSearch (bottom ). (c) Additionally, n = 15 healthy controls were screened
by the μHD. The mean values of cell counts are shown as a dashed line. (d) A receiver operating curve
(ROC) for the μHD was generated from the data in (c)
136 David Issadore
Fig. 7 Molecular analysis of fine needle aspirates on a μHall chip. (a) The μHall
chip was used to profile EGFR expression of cells obtained by fine needle aspi-
rate from mice with a xenografted tumor. Mice bearing xenografted tumors were
treated with geldanamycin for 6 days or left untreated (n = 6 per group). (b) Rate
of tumor growth in untreated mice and mice treated with geldanamycin. The blue
and red data points represent untreated and treated mice, respectively. (c) Tumor
samples were screened by the μHall chip to monitor the changes in EGFR expres-
sion over the course of the drug treatment (Color figure online)
References
5. Nagrath S, Sequist LV, Maheswaran S, Bell Liao JC, Gambhir SS et al (2009) Matrix-
DW, Irimia D, Ulkus L, Smith MR, Kwak EL, insensitive protein assays push the limits of
Digumarthy S, Muzikansky A, Ryan P, Balis biosensors in medicine. Nat Med 15:
UJ, Tompkins RG, Haber DA, Toner M 1327–1332
(2007) Isolation of rare circulating tumour 13. Lee JH, Huh YM, Jun YW, Seo JW, Jang JT,
cells in cancer patients by microchip technol- Song HT, Kim S, Cho EJ, Yoon HG, Suh JS,
ogy. Nature 450:1235 Cheon J (2007) Artificially engineered mag-
6. Mettlin C, Littrup PJ, Kane RA, Murphy GP, netic nanoparticles for ultra-sensitive molecu-
Lee F, Chesley A, Badalament R, Mostofi FK lar imaging. Nat Med 13:95–99
(1994) Relative sensitivity and specificity of 14. Issadore D, Min C, Liong M, Chung J,
serum prostate specific antigen (PSA) level Weissleder R, Lee H (2011) Miniature mag-
compared with age-referenced PSA, PSA den- netic resonance system for point-of-care diag-
sity, and PSA change. Cancer 74:1615–1620 nostics. Lab Chip 11:2282–2287
7. de B, Johann S, Scher HI, Montgomery RB, 15. Issadore D, Chung HJ, Chung J, Budin G,
Parker C, Miller MC, Tissing H, Doyle GV, Weissleder R, Lee H (2013) μHall chip for
Terstappen LWWM, Pienta KJ, Raghavan D sensitive detection of bacteria. Adv Healthc
(2008) Circulating tumor cells predict survival Mater 2(9):1224–1228. doi:10.1002/adhm.
benefit from treatment in metastatic castration- 201200380
resistant prostate cancer. Clin Cancer Res 16. Weigl B, Domingo G, LaBarre P, Gerlach J
14:6302–6309 (2008) Towards non-and minimally instru-
8. Ozkumur E, Shah AM, Ciciliano JC, Emmink mented, microfluidics-based diagnostic
BL, Miyamoto DT, Brachtel E, Yu M, Chen devices. Lab Chip 8:1999–2014
P-I, Morgan B, Trautwein J et al (2013) 17. Haun JB, Devaraj NK, Hilderbrand SA, Lee
Inertial focusing for tumor antigen – depen- H, Weissleder R (2010) Bioorthogonal chem-
dent and – independent sorting of rare circu- istry amplifies nanoparticle binding and
lating tumor cells. Sci Transl Med 5:179ra47 enhances the sensitivity of cell detection. Nat
9. Gaster R, Hall D, Wang S (2011) nanoLAB: Nanotechnol 5:660–665
an ultraportable, handheld diagnostic labora- 18. Howell J, Peter B, Mott DR, Fertig S, Kaplan
tory for global health. Lab Chip 11:950–956 CR, Golden JP, Oran ES, Ligler FS (2005) A
10. Issadore D, Westervelt RM (eds) (2013) Point microfluidic mixer with grooves placed on the
of care diagnostics on a chip. Springer, Berlin, top and bottom of the channel. Lab Chip
Heidelberg. 5:524–530
11. Ejsing L, Hansen MF, Menon AK, Ferreira HA, 19. Rhim AD, Mirek ET, Aiello NM, Maitra A,
Graham DL, Freitas PP (2004) Planar Hall Bailey JM, McAllister F, Reichert M, Beatty
effect sensor for magnetic micro- and nanobead GL, Rustgi AK, Vonderheide RH et al (2012)
detection. Appl Phys Lett 84:4729–4731 EMT and dissemination precede pancreatic
12. Gaster RS, Hall DA, Nielsen CH, Osterfeld tumor formation. Cell 148:349–361
SJ, Yu H, Mach KE, Wilson RJ, Murmann B,
Chapter 10
Abstract
Flow cytometry is used for cell counting and analysis in numerous clinical and environmental applications.
However flow cytometry is not used in mHealth mainly because current flow cytometers are large, expensive,
power-intensive devices designed to operate in a laboratory. Their design results in a lack of portability and
makes them unsuitable for mHealth applications. Another limitation of current technology is the low volu-
metric throughput rates that are not suitable for rapid detection of rare cells.
To address these limitations, we describe here a novel, low-cost, mobile flow cytometer based on
wide-field imaging with a webcam for large volume and high throughput fluorescence detection of rare
cells as a simulation for circulating tumor cells (CTCs) detection. The mobile flow cytometer uses a com-
mercially available webcam capable of 187 frames per second video capture at a resolution of 320 × 240
pixels. For fluorescence detection, a 1 W 450 nm blue laser is used for excitation of Syto-9 fluorescently
stained cells detected at 535 nm. A wide-field flow cell was developed for large volume analysis that allows
for the linear velocity of target cells to be lower than in conventional hydrodynamic focusing flow cells
typically used in cytometry. The mobile flow cytometer was found to be capable of detecting low concen-
trations at flow rates of 500 μL/min, suitable for rare cell detection in large volumes. The simplicity and
low cost of this device suggests that it may have a potential clinical use for mHealth flow cytometry for
resource-poor settings associated with global health.
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_10, © Springer Science+Business Media New York 2015
139
140 Joshua Balsam et al.
2 Materials
2.4 Cell Culture 1. THP-1 human monocytes, ATCC #TIB-202 (ATCC Manassas, VA).
and Fluorescence 2. Culture media RPMI-1640 medium supplemented with 10 %
Staining (v/v) heat-inactivated FBS, 1 % antibiotic–antimycotic solu-
tion, and 2 mM glutamine.
3. SYTO-9 dye (Molecular Probes Eugene, OR).
142 Joshua Balsam et al.
3 Methods
3.1 Wide-Field Flow The flow cytometer for fluorescence detection (Fig. 1) consists of
Cytometer for mHealth four main elements: (1) a webcam used as an optical detector,
Applications (2) a laser excitation source for illumination, (3) a wide-field flow
cell, and (4) an optical stage to hold each module in alignment for
stable and clear imaging. The optical detector consists of the elec-
tronics of a webcam which was disassembled in order to remove
the original lens, a 12 mm f/1.2 CCTV lens which enables
improved light collection and image magnification (see Note 1), a
green emission filter for fluorescent imaging, and a computer to
collect and analyze data. The excitation source is a 1 W 450 nm
blue laser (see Note 2). The optical elements must be vertically
centered (see Notes 2 and 3). The fluidics module consists of a
flow cell and a programmable syringe pump. The flow cytometer
platform shown in Fig. 1a consists of a stationary platform and a
moveable stage for focusing (using a screw mechanism). The flow
cytometer platform was constructed using 0.5 in. thick clear acrylic
sheet and nylon rod (McMaster-Carr, Robbinsville, NJ) which was
used as a rail for focusing. The webcam electronic circuitry and
optics were attached to the stationary platform while the flow cell
was attached to the translating stage. It is important to enclose the
device to reduce background light (see Note 4).
a b
Fig. 1 Schematic of mobile wide-field flow cytometer for mHealth. (a) The flow cytometer consists of four
modules: (1) a webcam as an optical detector, (2) a blue 435 nm excitation source for illumination, (3) a wide-
field flow cell, and (4) a stage to focus the image ant to hold each module in alignment. The sensing element
(1) consists of the electronic elements of a webcam, a 12 mm f/1.2 CCTV lens, a green emission filter. It is
connected to a computer to collect and analyze data. The excitation source (2) is a 450 nm 1 W blue laser.
The sample handling module consists of the wide-field flow cell, a programmable syringe pump, and a waste
container. (b) A schematic of the wide-field flow cell with key elements labeled with the illuminated field of
detection
Mobile Flow Cytometer for mHealth 143
3.2 Wide-Field Flow The flow cell (Fig. 1b) consists of a 4 mm wide imaging channel
Cell Fabrication through which fluorescent labeled samples are injected via an inlet,
excited by a laser, and then imaged by the webcam before being
collected at the outlet. The wide-field flow cell was fabricated using
an Epilog Legend CO2 65 W laser cutter (Epilog, Golden, CO) (see
Note 5) using similar techniques described in our previous work
[11, 21–25]. The flow cell consisted of three functional layers. The
upper layer fabricated of a 3 mm thick clear acrylic plate (75 × 25 mm)
with laser machined inlet and outlet ports into which 18GA needles
were pressed and bonded to allow for sample injection and waste
collection. The middle layer, which defines the shape of the fluid
flow, is a laser machined channel fabricated from 3M 9770 double-
sided adhesive transfer tape, which results in a wide-field flow cell
with width 4 mm, length 45 mm, and depth of approximately
0.075 mm. The bottom layer consisted of a single glass microscope
slide which was bonded to the acrylic with the double-sided adhe-
sive transfer tape micromachined flow channel (see Note 6).
A Fusion100 syringe pump connected to the flow cell was used
for flow rate control. The maximum flow rate achievable through
this flow cell is 10 mL/min. The adhesive may fail at higher flow
rate due to the dynamic pressures required. The flow rate in experi-
ments presented in this paper was limited to 500 μL/min due to
the maximum frame rate of the webcam employed for sensing. For
very faint cells the flow rate should be decreased in order to improve
sensitivity. Sensitivity also increases if exposure time is increased to
allow cell images to form short streaks, with length equal to the
length of the cell image plus one pixel (detailed in Subheading 3.7).
3.3 Optical System The optical system includes the blue laser for fluorophore excitation,
for Webcam-Based webcam detector, and excitation filter.
Cytometer Platform
Laser Excitation. For fluorescent imaging of a wide field, a 1 W
consumer laser was used to project an elliptical spot which covered
the width of the flow cell. The laser illuminates the flow cell at an
angle of incidence of approximately 45° (see Note 2). A laser such
as this is fairly expensive (~$300) for a device designed for use in a
low-resource setting. To reduce the cost of the laser, a lower power
laser with line generator optics could be used to further focus the
laser spot. This could allow the critical parameter of photon flux to
remain unchanged while reducing overall power. Alternatively,
high power LEDs could be utilized. This would require the addi-
tion of an excitation filter as well as collimating optics, leading to a
more complex and potentially more expensive configuration.
Webcam detector and emission filter. A Sony PlayStation® Eye web-
cam was used as the photodetector in this platform by converting
the webcam to a microscope. The device was disassembled and
the main circuit board (with attached image sensor and USB
cable) was removed. A C-mount CCTV lens (Pentax 12 mm f/1.2)
144 Joshua Balsam et al.
3.4 Cell Culture Fluorescently stained THP-1 human monocytes were used as a
Staining and Dilution model to simulate rare CTC. Though monocytes themselves are
not rare, they were diluted to levels similar to those of rare cells
such as CTCs. Cells were cultured with RPMI-1640 medium sup-
plemented with 10 % (v/v) heat-inactivated FBS, 1 % antibiotic–
antimycotic solution, and 2 mM glutamine (see Note 7) in a 5 %
CO2 environment at 37 °C. Cells were removed from an active
culture, pelleted by centrifugation and resuspended in deionized
water at an approximate concentration of 106 cells/mL (staining
protocol for Syto-9 dye advises against the use of phosphate buffer
solutions). 10 μL of Syto-9 dye (3.34 mM stock concentration)
was added to 1 mL of suspended cells and allowed to rest at room
temperature in the dark for 20 min. Washing of cells to remove
excess dye is not necessary due to the low intrinsic quantum yield
when not bound to nucleic acids (<0.01), and because of the later
dilution of this stock solution by factors of 104–106.
One known factor which contributed substantially to the
decreasing counting efficiency at lower concentrations is the cell
staining protocol. The protocol recommended by the dye manu-
facturer was followed. However, dye which had entered the cell
but not yet bound to DNA was noted to diffuse out of cells over
time, leading to diminishing cell brightness. Because all dilutions
were prepared simultaneously and then analyzed sequentially from
highest to lowest concentration, lower dilutions exhibited cells
with lower fluorescent intensity. Allowing the cells to equilibrate at
a low concentration for several hours would allow this diffusion
process time to complete.
The stock solution of cells after staining was diluted to a level
of approximately 1–10 cell/μL (measured by microscopy) to allow
for accurate manual counting. Cell concentration was measured by
placing 3 μL sample droplets on a microscope slide and counting
cells in the droplet in real time under laser excitation using the
same imaging platform employed to image the flow cells in these
experiments. This was repeated many (N > 20) times. An average
cell concentration of 3.73 cells/μL with a standard error of 0.3 is
an example of typical population estimate resulting from these
measurements. From this relatively high concentration, lower con-
centration samples of 100, 10, and 1 cell/mL were generated by
Mobile Flow Cytometer for mHealth 145
3.5 Imaging Flow The webcam sensor was connected to a 32-bit Windows-based
Cytometry laptop computer via a USB2 port. The drivers and software con-
trolling the webcam were developed and freely distributed by Code
Laboratories, Inc. (Henderson, NV). The camera control software
(CL-Eye Test) enables setting several camera parameters (exposure
time, frame rate, and gain), and the capture of video in uncom-
pressed AVI format. The imaging parameters for the camera (expo-
sure time and frame rate) depend on the brightness of the
fluorescently labeled cells and the flow rate (see Note 8). A single
video frame of THP-1’s stained with SYTO-9 dye in wide-field
flow cell at a flow rate of 500 μL/min is shown in Fig. 2a. The flow
cell geometry and fluorescent detection optics allow for a high sig-
nal to noise ratio for easy detection of these cells (Fig. 2b).
Fig. 2 Cell imaged using wide-field flow cytometer and webcam-based flow focusing cytometry. (a) Single
video frame of fluorescent labeled THP-1 human monocytes in wide-field flow cell at 500 μL/min, and (b) 3D
visualization of a
146 Joshua Balsam et al.
First, the distance between the imaging lens and the flow cell
should be set such that images of cells are projected onto at most a
3 × 3 array of pixels. Distance should not be so great that a cell
image is less than one pixel in size. At those distances, photon flux
per pixel begins to diminish and SNR drops quickly. When cells are
imaged on more than one pixel, photon flux per pixel is a constant
and independent of the distance between flow cell and lens. The
largest possible distance should be chosen in order to maximize
field of view (FOV). Set the flow cell width so that it covers the
FOV of the image sensor at this lens-to-flow cell distance. This
allows the largest possible flow cell width to be used which mini-
mizes flow velocity and maximizes SNR at a given flow rate.
Next, the relationship between average cell velocity and flow
rate should be determined empirically. The maximum velocity
achievable will be determined later and is a function of cell image
signal to noise ratio (SNR) (e.g., higher SNR allows higher veloc-
ity). This in turn depends on the cell staining characteristics
(e.g., quantum yield of the dye, number of fluorophores bound,
and excitation field intensity), the camera used (e.g., camera sensi-
tivity, characteristic noise levels, and maximum frame rate), and the
lens used (e.g., focal ratio).
Exposure time should be set such that cell image brightness is
maximized. For a given average flow velocity and average cell
brightness (i.e., photon emission rate), there will be a maximum
cell image brightness that can be produced. This is based on the
number of photons than can strike a pixel as the cell image passes
over it. The minimum exposure time that will yield this maximum
image intensity will be the time required for a cell image to traverse
a number of pixels equal to its image length in pixels (L) plus one
pixel, thereby producing an image streak of length 2L + 1.
Finally, volumetric flow rate should be set such that the entire
distribution of cells is significantly above the noise background of
the system (this typically corresponds to an SNR > 5). At higher
flow rates image SNR will decrease. Above a certain flow rate, cells
at the faint end of the distribution will be indistinguishable from
background noise and the average cell count will decrease. In order
to set flow rate, a stock solution of fluorescent cells should be pre-
pared and counted beginning at a high flow rate and decreasing
until the average cell count per sample reaches a constant value.
As previously mentioned this maximum flow rate will depend on
several factors, including the inherent brightness of cell staining
and the excitation photon flux. The parameters of exposure time
and frame rate depend directly on flow rate, so these parameters
will need to be updated as flow rate is varied in order to maintain
similar levels of device sensitivity across various tests.
Fig. 3 Background subtraction to improve imaging. (a) Median pixel value from 2,000 frames showing average
background autofluorescence from the flow cell, (b) maximum pixel value from 2,000 video frames showing a
single cell moving through the laser spot (marked with circled in yellow), (c) result of subtracting image b from
image c, allowing for improved visualization of cell movement and faint cell images
positions along its path). The median image of all the frames is
shown in Fig. 3a (this represents the average background
signal), and the maximum image is shown in Fig. 3b which
shows the highest signal recorded at each pixel during the video
with the cell position in each frame marked. In order to improve
cell identification, especially for lower SNR images than this
example, the median image shown in Fig. 3a, which contains
only background signal and no information from the cells, was
subtracted from the maximum image to produce Fig. 3c, which
allows for improved visualization of cell movement by removing
background autofluorescence from the flow cell.
4. Cell streaking. Cell streaking is imaging of moving cells at long
exposure times such that they produce line images, as seen in
Fig. 4a. This cell image was captured at a flow rate of 20 mL/
min with a frame rate of 20 fps. The primary benefit of cell
streaking is that it allows pixels to integrate over the entire
length of a cell as its moves image moves over them. Shorter
exposure times result in only a portion of the available photon
flux to be integrated by a pixel, reducing image SNR. Figure 4a
shows significant streaking, which is not necessary to realize
this benefit. The minimum streak length required such that at
least one pixel measures the maximum possible signal is equal
to twice the length of the cell image in pixels before streaking
(L) plus one pixel (i.e., for a cell image of 3 × 3 pixels, a streak
length of 7 is required). This effect is shown graphically in
Fig. 4b, where a cell of length 3 pixels is seen to move a dis-
tance of 4 pixels, thereby producing an image streak with
length 7 pixels with a single bright pixel in its center. An actual
cell streak image is shown in Fig. 4c for a similar cell image size
and similar flow conditions, along with a plot of actual pixel
brightness along the center of the cell streak in Fig. 4d. Cell
streaking such as this results in at least one pixel measuring the
maximum possible signal from the cell.
Mobile Flow Cytometer for mHealth 149
d 20
15
Gray Value
10
0 2 4 6 8
Distance (pixels)
Fig. 4 Cells in a streaking mode. (a) An image of cells traveling in a flow rate of
10 mL/min captured in 20 frames per second imaged as lines. (b) Schematic of
a cell traversing a number of pixels equal to its length plus one pixel, showing a
maximum brightness achieved in the pixel at the image center. (c) An actual cell
image is shown under these approximate flow conditions (125 μL/min, 20 fps),
along with (d) a plot of its brightness along the center line of pixels
150 Joshua Balsam et al.
a 1000
Before Optimization
Measured Cell Count (cells/mL)
100
b
10 Inlet
1
0.1
0.01
Field of View
100 10 1
c 1000 Laser
After Optimization
Measured Cell Count (cells/mL)
100
10
0.1 Outlet
0.01
100 10 1 0.1
Target Cell Count (cells/mL)
Fig. 5 Rare cell counting using streaking mode. (a) Results of counting dilutions of rare cells using the first
generation flow cell depicted in Fig. 1b. The large error bars evident at the level of 1 cell/mL prompted further
optimization of the flow cell geometry and operating parameters. (b) An updated flow cell design which
improves image SNR by reducing cell velocity. Field of view was also improved. (c) Following optimization,
performance of the wide-field flow cytometer improved substantially at the level of 1 cell/mL, and utility was
expanded to measurements as low as 0.1 cell/mL. In each case, measurements made via microscopy (sparsely
filled columns) and those made via wide-field flow cytometry (dense fill) were not significantly different at the
95 % confidence level
Mobile Flow Cytometer for mHealth 151
4 Notes
6. For strong bonding, all air bubbles between the surfaces and
the adhesive tape should be pressed out manually. The adhesive
layer should be attached to the acrylic side first to ensure that
proper alignment exists between the inlet and outlet ports.
Bubbles between these layers can be pressed out before the pro-
tective paper layer on the back of the adhesive tape is removed.
After this, with the protective paper removed, the glass layer
can be bonded. Bubbles between the glass and adhesive layer
can be removed by carefully applied pressure, or by use of a
heated press. Care must be taken to ensure even pressure
because of the risk of glass breakage. To avoid this, the glass
layer can be replaced by another layer of acrylic. However the
high autofluorescent emission rate of acrylic will result in
reduced SNR and diminished dynamic range of the sensor. For
improved sensitivity, two quartz microscope slides can be used
in place of the acrylic layer and the glass slide. Inlet and outlet
holes must be cut using a glass cutting drill bit or other appro-
priate method. High pressure ports can be constructed by
pressing an 18GA needle into a laser cut hole in a small square
of acrylic sheet, sealing with cyanoacrylate glue, and bonding
over the inlet and outlet holes in the quartz slide using double
sided adhesive transfer tape with a center hole cut out.
7. Cell cultures with very high density may result in cell anucle-
ation (ghost cells), cells with low fluorescence signal when
labeled with nucleic acid stain.
8. Imager Qualification: webcam performance varies depending
both on the device and the application used to collect the
images from the camera. Prior to preparing a sample for mea-
surement it is important to understand the performance of the
camera and application being used. First, block any light from
reaching the camera sensor by taping a layer of aluminum foil
over the lens aperture. Take two images and open them in
ImageJ or a comparable software package. If a color sensor is
used, use only the green channel for analysis. Convert the
images to 32-bit float format, and subtract one image from the
other. Finally, use the software package to produce a histogram
of all pixel values. This histogram should have an approxi-
mately Gaussian profile. If the original images had pixel values
that were predominantly zero valued, you must change the
settings of the camera capture software so that the black end of
the sensor histogram is not being artificially clipped. This clip-
ping behavior is typically set by an adjustment labeled bright-
ness. If the images captured by the sensor with no light arriving
cannot be made to show values above zero with an approxi-
mately Gaussian profile, it is likely that the device is not suit-
able for performing sensitive fluorescence measurements of
faint objects.
Mobile Flow Cytometer for mHealth 153
References
1. Golden JP, Kim JS, Erickson JS, Hilliard LR, 13. Zhu H, Mavandadi S, Coskun AF, Yaglidere O,
Howell PB, Anderson GP et al (2009) Multi- Ozcan A (2011) Optofluidic fluorescent imaging
wavelength microflow cytometer using groove- cytometry on a cell phone. Anal Chem 83(17):
generated sheath flow. Lab Chip 9(13): 6641–6647, Epub 2011/07/22
1942–1950, Epub 2009/06/18 14. Wei Q, Qi H, Luo W, Tseng D, Ki SJ, Wan Z
2. Howell PB Jr, Golden JP, Hilliard LR, Erickson et al (2013) Fluorescent imaging of single
JS, Mott DR, Ligler FS (2008) Two simple and nanoparticles and viruses on a smart phone. ACS
rugged designs for creating microfluidic sheath Nano 7(10):9147–9155, Epub 2013/09/11
flow. Lab Chip 8(7):1097–1103, Epub 15. Coskun AF, Nagi R, Sadeghi K, Phillips S,
2008/06/28 Ozcan A (2013) Albumin testing in urine using
3. Taitt CR, Anderson GP, Ligler FS (2005) a smart-phone. Lab Chip 13(21):4231–4238,
Evanescent wave fluorescence biosensors. Epub 2013/09/03
Biosens Bioelectron 20(12):2470–2487, Epub 16. Navruz I, Coskun AF, Wong J, Mohammad S,
2005/04/28 Tseng D, Nagi R et al (2013) Smart-phone
4. Ngundi MM, Qadri SA, Wallace EV, Moore based computational microscopy using multi-
MH, Lassman ME, Shriver-Lake LC et al frame contact imaging on a fiber-optic array. Lab
(2006) Detection of deoxynivalenol in foods Chip 13(20):4015–4023, Epub 2013/08/14
and indoor air using an array biosensor. Environ 17. Zhu H, Sencan I, Wong J, Dimitrov S, Tseng
Sci Technol 40(7):2352–2356 D, Nagashima K et al (2013) Cost-effective
5. Moreno-Bondi MC, Taitt CR, Shriver-Lake and rapid blood analysis on a cell-phone. Lab
LC, Ligler FS (2006) Multiplexed mea- Chip 13(7):1282–1288, Epub 2013/02/09
surement of serum antibodies using an 18. Zhu H, Sikora U, Ozcan A (2012) Quantum
array biosensor. Biosens Bioelectron 21(10): dot enabled detection of Escherichia coli using
1880–1886 a cell-phone. Analyst 137(11):2541–2544,
6. Ligler FS, Sapsford KE, Golden JP, Shriver- Epub 2012/03/08
Lake LC, Taitt CR, Dyer MA et al (2007) The 19. Zhu H, Yaglidere O, Su TW, Tseng D, Ozcan A
array biosensor: portable, automated systems. (2011) Wide-field fluorescent microscopy on a
Anal Sci 23(1):5–10 cell-phone. Conference proceedings: Annual
7. Kostov Y, Sergeev N, Wilson S, Herold KE, international conference of the IEEE engineering
Rasooly A (2009) A simple portable electrolu- in medicine and biology society, IEEE Engineering
minescence illumination-based CCD detector. in Medicine and Biology Society Conference,
Methods Mol Biol 503:259–272, Epub 2011. pp 6801–6804. Epub 2012/01/19
2009/01/20 20. Zhu H, Yaglidere O, Su TW, Tseng D, Ozcan
8. Sapsford KE, Sun S, Francis J, Sharma S, A (2011) Cost-effective and compact wide-
Kostov Y, Rasooly A (2008) A fluorescence field fluorescent imaging on a cell-phone. Lab
detection platform using spatial electrolumi- Chip 11(2):315–322, Epub 2010/11/11
nescent excitation for measuring botulinum 21. Rasooly A, Bruck HA, Kostov Y (2013) An
neurotoxin A activity. Biosens Bioelectron ELISA lab-on-a-chip (ELISA-LOC). Methods
24(4):618–625, Epub 2008/07/23 Mol Biol 949:451–471, Epub 2013/01/19
9. Sun S, Francis J, Sapsford KE, Kostov Y, Rasooly 22. Rasooly A, Kostov Y, Bruck HA (2013)
A (2010) Multi-wavelength Spatial LED illumi- Charged-coupled device (CCD) detectors for
nation based detector for in vitro detection of lab-on-a chip (LOC) optical analysis. Methods
botulinum neurotoxin A activity. Sens Actuators Mol Biol 949:365–385, Epub 2013/01/19
B 146(1–8):297–306, Epub 2010/05/26 23. Balsam J, Bruck HA, Rasooly A (2013)
10. Sun S, Ossandon M, Kostov Y, Rasooly A Capillary array waveguide amplified fluores-
(2009) Lab-on-a-chip for botulinum neuro- cence detector for mHealth. Sens Actuators B
toxin a (BoNT-A) activity analysis. Lab Chip 186:711–717, Epub 2013/09/17
9(22):3275–3281, Epub 2009/10/30 24. Balsam J, Rasooly R, Bruck HA, Rasooly A
11. Sun S, Yang M, Kostov Y, Rasooly A (2010) (2014) Thousand-fold fluorescent signal
ELISA-LOC: lab-on-a-chip for enzyme-linked amplification for mHealth diagnostics. Biosens
immunodetection. Lab Chip 10(16):2093– Bioelectron 51:1–7, Epub 2013/08/10
2100, Epub 2010/06/15 25. Balsam J, Ossandon M, Bruck HA, Lubensky I,
12. Zhu H, Ozcan A (2013) Wide-field fluorescent Rasooly A (2013) Low-cost technologies for
microscopy and fluorescent imaging flow medical diagnostics in low-resource settings.
cytometry on a cell-phone. J Vis Exp. Expert Opin Med Diagn 7(3):243–255, Epub
doi:10.3791/50451, Epub 2013/04/23 2013/03/14
Chapter 11
Abstract
Oral and cervical cancers are a growing global health problem that disproportionately impacts women and
men living in the developing world. The high death rate in developing countries is largely due to the fact
that these countries do not have the appropriate medical infrastructure and resources to support the orga-
nized screening and diagnostic programs that are available in the developed world. Diffuse reflectance
spectroscopy (DRS) with a fiber-optic probe can noninvasively quantify the optical properties of epithelial
tissues and has shown the potential as a cost-effective, easy-to-use, and sensitive tool for diagnosis of early
precancerous changes in the cervix and oral cavity. However, current fiber-optic DRS systems have not
been designed to be robust and reliable for use in developing countries. They are subject to various sources
of systematic or random errors, arising from the uncontrolled probe–tissue interface and lack of real-time
calibration, use bulky and expensive optical components, and require extensive training. This chapter
describes a portable DRS device that is specifically designed for detection of oral and cervical cancers in
resource-poor settings. The device uses an innovative smart fiber-optic probe to eliminate operator bias,
state-of-the-art photonics components to reduce size and power consumption, and automated software to
reduce the need of operator training. The size and cost of the smart fiber-optic DRS system may be further
reduced by incorporating a smartphone based spectrometer.
Key words Diffuse reflectance spectroscopy, Fiber-optic sensor, Smartphone, Oral and cervical
cancers
1 Introduction
Each year there are 528,000 cases of cervical cancer with 274,000
deaths worldwide [1]. The majority of these cases (approxi-
mately 85 %) occur in resource-poor countries [1, 2]. Cervical
cancer is the second most common cancer among women. The
highest incidence rates of cervical cancer are found in Central
and South America, Africa, and Asia where rates may exceed 50
cases per 100,000 women. Because of the lack of modern pre-
vention practices in these areas, the global mortality rate of
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_11, © Springer Science+Business Media New York 2015
155
156 Bing Yu et al.
Fig. 1 Illustration of the principle of VIS-DRS with a fiber-optic probe and the spectrum analysis procedures.
(Reprinted with permission. Copyright Austin Publishing Group 2014)
2 Materials
2.1 Optical Systems 1. Multichannel USB spectrometer with two visible channels
and Components (range: 400–635 nm, resolution: 1.8 nm), one NIR channel
(range: 750–932 nm, resolution: 0.25 nm), (AvaSpec-2048-
USB2, Avantes, Inc., Broomfield, CO, USA).
2. Cary 300 UV–VIS spectrophotometer (Agilent Technologies,
Inc., Santa Clara, CA, USA).
3. High power white LED module, 1,000 μm plastic fiber pigtail
with FC/PC connector, (Doric Lenses, Inc., Québec, Canada).
4. 850 nm LED, 50/125 μm fiber pigtail, FWHM 50 nm, output
power ~150 μW, FC/PC receptacle, (Appointech Inc., Taiwan).
5. PowerPuck 350 mA LED Drive Module, (LED Supplies, Inc.,
Randolph, VT, USA).
6. DynaOhm constant current LED driver, 25 mA, (LED
Supplies, Inc., Randolph, VT, USA).
7. Large core optical fibers, high-OH silica/silica, 200/220/
239 μm, NA = 0.22, (FVP200220240, Polymicro Technologies,
Inc., Phoenix, AZ, USA).
8. Multimode fibers, 50/125-μm, NA = 0.22 (Corning, Inc.,
Corning, NY, USA).
9. 2 × 2 multimode fiber coupler, 850 nm, Corning multimode
fiber 50/125 μm, coupling ratio 50/50, FC/PC connectors,
(AC Photonics, Inc., Santa Clara, CA, USA).
10. Spectralon® Diffuse Reflectance target, reflectance factor 99 %,
wavelength range 250–2,500 nm, (SRS-99, Labsphere, Inc.,
North Sutton, NH, USA).
2.2 Computer 1. Dell laptop with USB ports, Windows 7, (Dell, Inc., Round
and Software Rock, TX, USA).
2. LabVIEW 2011, (National Instruments Corp., Austin, TX, USA).
3. Matlab 2011b with Curve Fitting Toolbox and Optimization
Toolbox, (Mathworks, Inc., Natick, MA, USA).
160 Bing Yu et al.
3 Methods
3.1 Mobile Fiber- A smart fiber-optic sensor system has been developed for perform-
Optic Sensor System ing DRS measurements in developing countries. A schematic of
the main components of the smart fiber-optic probe and the instru-
ment to which it is coupled are shown in Fig. 2a. The instrument
consists of an 850 nm LED, a high power white LED, a three-
channel fiber-optic USB spectrometer, and a laptop with custom
LabVIEW and Matlab software. All these components are low
power consuming and can be powered by batteries. The smart
probe integrates a VIS-DRS channel, a self-calibration (SC)
channel [38, 39], and a diaphragm-based Fabry–Perot interfero-
metric (DFPI) pressure sensor [40] into a single fiber-optic probe.
The SC channel records a calibration spectrum that can be used for
correction of instrument drifts and probe bending loss in real-time,
while the pressure sensor provides feedback on the probe pressure
so that the operator can manually adjust the force applied on it.
The DRS and calibration channels share the white LED as the light
source, while the pressure sensor uses the 850 nm LED as its
light source. The two visible channels of the USB spectrometer
(Spec A and B) are used for detection of the tissue DRS and SC
spectra, respectively. The NIR channel of the USB spectrometer
(Spec C) is used for detection of the interferograms from the DFPI
pressure sensor.
At the distal end, the DRS channel uses a single fiber (the blue
fiber) for DRS detection (connected to Spec A) and six source
fibers (the red fibers), forming a ring around the detection fiber for
tissue illumination. For epithelial mucosal tissue, a source–detector
separation (the radius of the illumination ring) of 640 μm was
used, which provides a simulated sensing depth of 0.5–2 mm. The
SC source fiber (one of the seven red fibers from the white LED)
is looped back, by the 99 % Spectralon diffuse reflectance coating
Mobile Fiber-Optic Sensor for Detection of Oral and Cervical Cancer… 161
Fig. 2 A smart optic sensor system: (a) schematic of the probe and instrument; (b) schematic of the DFPI pressure
sensor head; and (c) photograph of the portable instrument and smart probe. LED light emitting diode, Spec
spectrometer, R1 and R2 reflection, P external pressure, D diameter of the glass diaphragm [41]. (Reprinted
with permission. Copyright the Optical Society of America 2014)
inside the rigid part of the probe housing, into a SC detection fiber
(the pink fiber) that is connected to the Spec B. All fibers for the
tissue and SC channels use the same type of 200/220-μm fiber. Six
inches of the fiber-optic cable immediately after the rigid probe tip
was mounted inside a gooseneck tube so that the probe can be easily
bent to different angles inside an oral cavity.
The DFPI sensor head, as shown in Fig. 2b, is basically a low-
coherence Fabry–Perot interferometer formed by the cleaved end
face of the lead in/out fiber and the inner surface of a glass dia-
phragm [40]. The broadband NIR light from the 850 nm LED is
launched into a DFPI sensor through a 2 × 2 fiber-optic coupler
whose other input leg (the green fiber) is connected to Spec C
(Fig. 2a). The tip of the unused output leg is immersed in index
matching gel to minimize unnecessary back reflection. All fibers
used in the NIR channel are 50/125-μm multimode fibers. The
lead in/out fiber, fused silica ferrule and tube, as well as the dia-
phragm are bonded together using high temperature epoxy (Epoxy
MS-907). The reflected lights from the two air–glass interfaces (R1
and R2) propagate back to Spec C, generating interferogram.
External pressure P applied on the outer surface of the diaphragm
deflects it towards the fiber tip, thus reducing the cavity length of
the DFPI and causing a shift in the peak positions and a reduction
162 Bing Yu et al.
3.2 Software A LabVIEW program was used to automate the data collection
and analysis processes so that minimum training is required to
operate the instrument. The LabVIEW program includes the fol-
lowing function modules: (1) initializing the spectrometers,
selecting the probe and target type (phantom or tissue subject),
and loading saved probe configuration information; (2) acquiring
phantom/tissue, calibration and pressure spectra; (3) calling the
interferogram analysis algorithm in Matlab to calculate the probe
pressure P from the NIR spectrum; (4) performing self-calibra-
tion; (5) calling a Monte Carlo inversion model in Matlab to ana-
lyze the tissue spectrum if the pressure is within the preset range;
and (6) displaying the raw spectra as well as calculated probe
pressure and extracted tissue parameters, such as the hemoglobin
concentrations (HbO2, Hb, and THb), oxygen saturation (SO2),
and wavelength averaged reduced scattering coefficient. The time
required to measure and analyze the spectra from a tissue sample
is approximately 1–2 s. If and only if the measured probe pressure
falls within the preset range, e.g., 1–2 psi, the collected DRS and
SC spectra will be stored and analyzed. Otherwise, another scan
will be taken.
3.3 Data Analysis To correct for instrument drifts and fiber bending loss, the DRS
data is analyzed according to the flowchart in Fig. 3. On Day 1, a
single measurement is taken from a Spectralon® diffuse reflectance
target and is used to generate a correction factor:
On the day for tissue study (Day 2), DRS and SC spectra will
be collected from the tissue only. No separate calibration mea-
surement is necessary. The self-calibrated phantom and tissue
spectra, both corrected by Fcorr(λ), are input into an inverse Monte
Carlo model of reflectance, which extracts the tissue μa(λ) and
μs’(λ) [28]. From μa(λ), the tissue absorber concentrations, such
as [HbO2] and [Hb], can be accurately calculated using the Beer–
Lambert Law. The total hemoglobin concentration THb
(= [HbO2] + [Hb]) and blood oxygenation SO2 (= [HbO2]/THb)
can also be determined.
3.4 Pressure The pressure response of the smart sensor was measured in a pres-
Sensor Test sure test tube. The probe tip was mounted and sealed into the test
tube through a fitting. The gas pressure in the test tube was pro-
vided by compressed nitrogen through a gas pipe, tuned using a
pressure regulator, and monitored by a digital pressure gauge with
a resolution of 0.1 psi within 0–30 psi. Figure 4a shows the mea-
sured air cavity length (L) of a DFPI sensor vs. the applied nitrogen
pressure (P). A pressure sensitivity of 49 nm/psi was calculated
from the L–P curve.
To find out how well the probe pressure on an in vivo tissue
sample can be controlled with the smart sensor, the probe was
brought in contact with a volunteer’s finger. The preset probe
164 Bing Yu et al.
a 112.8 b 5
112.7 L = - 0.049193*P + 112.66
50 out of 170
within 2-3 psi
Sensor Cavity Length (µm)
112.6 4
112.3
2 Min
112.2
112.1 1
112
0
111.9
111.8 -1
0 5 10 15 0 20 40 60 80 100 120 140 160
Fig. 4 Pressure test results of a smart fiber-optic sensor: (a) measured DFPI sensor cavity length as a function
of the nitrogen pressure and (b) measured probe pressure during 170 continuous scans with best effort to
maintain a preset range of 2–3 psi. Negative pressure indicates that the probe was not in good contact with
the tissue
pressure was set to 2–3 psi and best effort was taken to maintain
the pressure within this range. A total of 170 pressure measure-
ments have been taken within ~200 s. The measured probe pres-
sures are plotted in Fig. 4b. Among the 170 measurements, 50 fell
within the preset range. The other measurements were either
above or below the preset pressure range which were mainly caused
by the shaking of the hand holding the probe during the scans.
Therefore, it requires an average of approximately four scans to
catch one valid measurement (within the preset pressure range),
which takes less than 40 ms.
Extracted µa (cm-1)
5
1
1 1.5 2 2.5 3 3.5 4 4.5 5 5.5
Expected µa (cm-1)
12
Extracted µs' (cm-1)
10
5 6 7 8 9 10 11 12
Fig. 5 Extracted vs. expected <μa(λ)> and <μs’(λ)> (wavelength averaged over 420–630 nm) of the 12 phan-
toms. The error bars were resulted from different phantom for reference
3.6 Measurement To evaluate the response of oral mucosa to increasing probe pres-
of Oral Mucosa sure, the probe was cleaned using 2 % chlorhexidine digluconate
solution and was then placed in contact with the mucosal tissue
from the inner cheek of a healthy volunteer. During the optical
measurements the volunteer was asked to close the mouth to main-
tain a relatively constant probe temperature through the study.
Room lights were dimmed during the measurements to minimize
the background light. Reflectance measurements were taken at
multiple pressure levels from 0.5 ± 0.5 psi up to 9.5 ± 0.5 psi. Prior
to the reflectance measurements a test run was conducted the vol-
unteer was trained on how to hold the probe, read the pressure
readings on the laptop screen, and maintain the probe pressure
within the preset range. At each pressure level ten pairs of DRS and
SC spectra were acquired sequentially at a 1–2-s interval. The first
measurement started immediately after the desired probe pressure
was reached and best effort was made to maintain the pressure at
the desired value during the ten scans. The probe pressure at each
valid scan was recorded. The DRS spectra measured from the
mucosal tissues analyzed following the procedures in Fig. 3.
166 Bing Yu et al.
30 15
data
10
HbO2 (µM)
Hb (µM)
5
10
0 data
cubic fit
0 −5
0 2 4 6 8 10 0 2 4 6 8 10
Pressure (PSI) Pressure (PSI)
30 120
data data
100
5th degree fit
THb (µM)
SO2 (%)
80
60
10
40
0 20
0 2 4 6 8 10 0 2 4 6 8 10
Pressure (PSI) Pressure (PSI)
16
data
14 6th degree fit
µs (cm−1)
12
10
8
0 2 4 6 8 10
Pressure (PSI)
Fig. 6 The tissue HbO2, Hb, THb, SO2, and <μs’(λ)> measured from the left cheek of a healthy volunteer under
increased probe pressure (ten pressure levels from 0.5 ± 0.5 psi to 9.5 ± 0.5 psi with ten measurements under
each pressure level)
Figure 6 shows the tissue HbO2, Hb, THb, SO2, and <μs’(λ)>
measured from the left cheek of the volunteer with a probe pres-
sure from 0.5 ± 0.5 to 9.5 ± 0.5 psi. A trend of decrease in HbO2,
THb, SO2, and μs’ and increase in Hb with probe pressure has
been identified. The sharpest changes in all tissue parameters
occur within 0–3 psi. These changes may be attributed to the
compression of the blood vessels which causes reduced blood
flow and alterations in the metabolism of the tissue as well as a
change in the density of the scatterers. THb also showed a
rebound above 4 psi which was likely due to the return of blood
to the compressed area.
DRS spectra have also been collected continuously from the
right cheek of the volunteer under a constant probe pressure of
1–2 psi to investigate how the tissue parameters of the oral mucosa
respond to constant probe pressure. Figure 7 shows the typical tis-
sue parameters extracted from 100 repeated scans from the volun-
teer. Significant fluctuations in the THb and SO2 have been
observed in the first 30 s (corresponding to first 20–25 data points).
The scattering coefficients were relatively constant through the
measurements. This suggested that DRS measurements should not
start until the tissue physiology stabilizes.
Mobile Fiber-Optic Sensor for Detection of Oral and Cervical Cancer… 167
HbO2 (µM) 15 10
Hb (µM)
10
5
5
0 0
0 20 40 60 80 100 0 20 40 60 80 100
Measurement # Measurement #
30 100
THb (µM)
SO2 (%)
20
50
10
0 0
0 20 40 60 80 100 0 20 40 60 80 100
Measurement # Measurement #
14
µs (cm−1)
12
10
8
0 20 40 60 80 100
Measurement #
Fig. 7 Tissue parameters measured from the right cheek of a volunteer under a constant probe pressure
of 1–2 psi
4 Notes
References
1. Ferlay J et al (2010) Estimates of worldwide comparison to clinical measurements.
burden of cancer in 2008: GLOBOCAN J Biomed Opt 11(6):064027
2008. Int J Cancer 127(12):2893–2917 16. Cardenas-Turanzas M et al (2007) The clinical
2. Vizcaino AP et al (2000) International trends effectiveness of optical spectroscopy for the
in incidence of cervical cancer: II. Squamous- in vivo diagnosis of cervical intraepithelial neo-
cell carcinoma. Int J Cancer 86(3):429–435 plasia: where are we? Gynecol Oncol 107
3. Westra WH (2009) The changing face of head (1 Suppl 1):S138–S146
and neck cancer in the 21st century: the impact 17. Freeberg JA et al (2007) The performance of
of HPV on the epidemiology and pathology of fluorescence and reflectance spectroscopy for
oral cancer. Head Neck Pathol 3(1):78–81 the in vivo diagnosis of cervical neoplasia;
4. Parkin DM et al (2005) Global cancer statis- point probe versus multispectral approaches.
tics, 2002. CA Cancer J Clin 55(2):74–108 Gynecol Oncol 107(1 Suppl 1):S248–S255
5. Licitra L et al (2002) Cancer of the orophar- 18. Wang A, Nammalavar V, Drezek R (2007)
ynx. Crit Rev Oncol Hematol 41(1):107–122 Targeting spectral signatures of progressively
6. Dahlstrand HM, Dalianis T (2005) Presence dysplastic stratified epithelia using angularly
and influence of human papillomaviruses variable fiber geometry in reflectance Monte
(HPV) in Tonsillar cancer. Adv Cancer Res Carlo simulations. J Biomed Opt 12(4):
93:59–89 044012
7. Chang VT et al (2009) Quantitative physiology 19. Arifler D et al (2005) Reflectance spectroscopy
of the precancerous cervix in vivo through opti- for diagnosis of epithelial precancer: model-
cal spectroscopy. Neoplasia 11(4):325–332 based analysis of fiber-optic probe designs to
resolve spectral information from epithelium
8. Muller MG et al (2003) Spectroscopic detec- and stroma. Appl Opt 44(20):4291–4305
tion and evaluation of morphologic and bio-
chemical changes in early human oral 20. Lane PM et al (2006) Simple device for the
carcinoma. Cancer 97(7):1681–1692 direct visualization of oral-cavity tissue fluores-
cence. J Biomed Opt 11(2):024006
9. de Veld DC et al (2005) Autofluorescence and
diffuse reflectance spectroscopy for oral oncol- 21. Pavlova I et al (2009) Fluorescence spectros-
ogy. Lasers Surg Med 36(5):356–364 copy of oral tissue: Monte Carlo modeling
with site-specific tissue properties. J Biomed
10. Sharwani A et al (2006) Assessment of oral Opt 14(1):014009
premalignancy using elastic scattering spec-
troscopy. Oral Oncol 42(4):343–349 22. Hasina R, Lingen MW (2001) Angiogenesis in
oral cancer. J Dent Educ 65(11):1282–1290
11. Amelink A et al (2008) Non-invasive measure-
ment of the morphology and physiology of 23. Georgakoudi I et al (2002) Trimodal spectros-
oral mucosa by use of optical spectroscopy. copy for the detection and characterization of
Oral Oncol 44(1):65–71 cervical precancers in vivo. Am J Obstet
Gynecol 186(3):374–382
12. Rahman M et al (2008) Low-cost, multimodal,
portable screening system for early detection of 24. Liu Q, Ramanujam N (2006) Sequential esti-
oral cancer. J Biomed Opt 13(3):030502 mation of optical properties of a two-layered
epithelial tissue model from depth-resolved
13. Schwarz RA et al (2008) Autofluorescence and ultraviolet-visible diffuse reflectance spectra.
diffuse reflectance spectroscopy of oral epithe- Appl Opt 45(19):4776–4790
lial tissue using a depth-sensitive fiber-optic
probe. Appl Opt 47(6):825–834 25. Zonios G et al (1999) Diffuse reflectance spec-
troscopy of human adenomatous colon polyps
14. Mirabal YN et al (2002) Reflectance spectros- in vivo. Appl Opt 38(31):6628–6637
copy for in vivo detection of cervical precancer.
J Biomed Opt 7(4):587–594 26. Farrell TJ, Patterson MS, Wilson B (1992) A
diffusion theory model of spatially resolved,
15. Arifler D et al (2006) Spatially resolved reflec- steady-state diffuse reflectance for the nonin-
tance spectroscopy for diagnosis of cervical vasive determination of tissue optical proper-
precancer: Monte Carlo modeling and ties in vivo. Med Phys 19(4):879–888
170 Bing Yu et al.
27. Volynskaya Z et al (2008) Diagnosing breast 39. Yu B, Fu HL, Ramanujam N (2011)
cancer using diffuse reflectance spectroscopy Instrument independent diffuse reflectance
and intrinsic fluorescence spectroscopy. spectroscopy. J Biomed Opt 16(1):011010
J Biomed Opt 13(2):024012 40. Yu B et al (2003) Fiber Fabry-Perot sensors for
28. Palmer GM, Ramanujam N (2006) Monte detection of partial discharges in power trans-
Carlo-based inverse model for calculating tis- formers. Appl Opt 42(16):3241–3250
sue optical properties. Part I: Theory and vali- 41. Yu B et al (2014) Diffuse reflectance spectros-
dation on synthetic phantoms. Appl Opt copy of epithelial tissue with a smart fiber-optic
45(5):1062–1071 probe. Biomed Opt Express 5(3):675–689
29. Thueler P et al (2003) In vivo endoscopic tis- 42. Qi B et al (2003) Novel data processing tech-
sue diagnostics based on spectroscopic niques for dispersive white light interferome-
absorption, scattering, and phase function
ter. Opt Eng 42:3165–3171
properties. J Biomed Opt 8(3):495–503 43. Zhang J (2008) Spectrometers miniaturized
30. Wang Q et al (2008) Measurement of internal for working with cellular phones and other
tissue optical properties at ultraviolet and visi- portable electronic devices. Int. Patent
ble wavelengths: development and implemen- Application No. PCT/US2008/083613
tation of a fiberoptic-based system. Opt 44. Wang S, Zhou X (2009) Spectroscopic sensor
Express 16(12):8685–8703 on mobile phone. US Patent 7,420,663
31. Reif R, A’Amar O, Bigio IJ (2007) Analytical 45. Chu K et al (2011) Microscopy and spectros-
model of light reflectance for extraction of the copy on a cell phone. In: Bio-optics: design
optical properties in small volumes of turbid and application, Monterey, California United
media. Appl Opt 46(29):7317–7328 States. p BWA3
32. Utzinger U, Richards-Kortum RR (2003) 46. Gallegos D et al (2013) Label-free biodetec-
Fiber optic probes for biomedical optical spec- tion using a smartphone. Lab Chip 13(11):
troscopy. J Biomed Opt 8(1):121–147 2124–2132
33. Reif R et al (2008) Analysis of changes in 47. Sumriddetchkajorn S, Somboonkaew A,
reflectance measurements on biological tissues Chanhorm S (2012) Mobile device-based digi-
subjected to different probe pressures. J tal microscopy for education, healthcare, and
Biomed Opt 13(1):010502 agriculture. In: 9th International conference
34. Ti Y, Lin WC (2008) Effects of probe contact on electrical engineering/electronics, com-
pressure on in vivo optical spectroscopy. Opt puter, telecommunications and information
Express 16(6):4250–4262 technology (ECTI-CON), Phetchaburi
35. Chan EK et al (1996) Effects of compression 48. Tseng D et al (2010) Lensfree microscopy on
on soft tissue optical properties. IEEE J Sel a cellphone. Lab Chip 10(14):1787–1792
Top Quantum Electron 2(4):943–950 49. Ayas S et al (2013) Counting molecules with a
36. Nichols MG, Hull EL, Foster TH (1997) mobile phone camera using plasmonic
Design and testing of a white-light, steady- enhancement. ACS Photonics. doi:10.1021/
state diffuse reflectance spectrometer for deter- ph400108p
mination of optical properties of highly 50. Wei Q et al (2013) Fluorescent imaging of
scattering systems. Appl Opt 36(1):93–104 single nanoparticles and viruses on a smart
37. Marin NM et al (2006) Calibration standards phone. ACS Nano 7(10):9147–9155
for multicenter clinical trials of fluorescence 51. Colaner S (2013) Public labs open source smart-
spectroscopy for in vivo diagnosis. J Biomed phone spectrometer let’s you find your wave-
Opt 11(1):014010 length. HotHardware, 18 Nov 2013. http://
38. Yu B et al (2008) Diffuse reflectance spectroscopy hothardware.com/News/Public-Labs-Open-
with a self-calibrating fiber optic probe. Opt Source-Smartphone-Spectrometer-Lets-You-
Lett 33(16):1783–1785 Find-Your-Wavelength/
Chapter 12
Abstract
Blood analysis is one of the most important clinical tests for medical diagnosis. Flow cytometry and optical
microscopy are widely used techniques to perform blood analysis and therefore cost-effective translation
of these technologies to resource limited settings is critical for various global health as well as telemedicine
applications. In this chapter, we review our recent progress on the integration of imaging flow cytometry
and fluorescent microscopy on a cell phone using compact, light-weight and cost-effective opto-fluidic
attachments integrated onto the camera module of a smartphone. In our cell-phone based opto-fluidic
imaging cytometry design, fluorescently labeled cells are delivered into the imaging area using a disposable
micro-fluidic chip that is positioned above the existing camera unit of the cell phone. Battery powered
light-emitting diodes (LEDs) are butt-coupled to the sides of this micro-fluidic chip without any lenses,
which effectively acts as a multimode slab waveguide, where the excitation light is guided to excite the
fluorescent targets within the micro-fluidic chip. Since the excitation light propagates perpendicular to the
detection path, an inexpensive plastic absorption filter is able to reject most of the scattered light and create
a decent dark-field background for fluorescent imaging. With this excitation geometry, the cell-phone
camera can record fluorescent movies of the particles/cells as they are flowing through the microchannel.
The digital frames of these fluorescent movies are then rapidly processed to quantify the count and the
density of the labeled particles/cells within the solution under test. With a similar opto-fluidic design, we
have recently demonstrated imaging and automated counting of stationary blood cells (e.g., labeled white
blood cells or unlabeled red blood cells) loaded within a disposable cell counting chamber. We tested the
performance of this cell-phone based imaging cytometry and blood analysis platform by measuring the
density of red and white blood cells as well as hemoglobin concentration in human blood samples, which
showed a good match to our measurement results obtained using a commercially available hematology
analyzer. Such a cell-phone enabled opto-fluidics microscopy, flow cytometry, and blood analysis platform
could be especially useful for various telemedicine applications in remote and resource-limited settings.
Key words Flow cytometry, Fluorescent microscopy, Imaging, Cell-phone based diagnostics, Blood
analysis, Telemedicine, Global health
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_12, © Springer Science+Business Media New York 2015
171
172 Hongying Zhu and Aydogan Ozcan
a b
c
Glass (or PDMS)
(n1=1.45 (or 1.41))
Liquid (n2=1.33)
Glass (n1=1.45)
Battery Battery
powered Fluorescence emission powered
LED LED
Absorption filter
External lens
Fig. 1 (a) Schematic diagram of the opto-mechanical attachment for fluorescent imaging cytometry on a cell
phone. (b) The picture of our opto-fluidic fluorescent imaging cytometer on a cell phone. (c) Schematic illustra-
tion for opto-fluidic pumping/excitation scheme and the cell-phone based optical imaging system. (Reprinted
with permission from Zhu, H., Mavandadi, S., Coskun, A. F., Yaglidere, O., and Ozcan, A., Opto-fluidic Fluorescent
Imaging Cytometry on a Cellphone, Anal. Chem., 83, 6641–6647, 2011. Copyright 2011 American Chemical
Society)
determine the total count of the labeled cells flowing through the
imaging field of view.
Using the same opto-fluidic pumping/excitation scheme, we
are also able to obtain fluorescent microscopic images of stationary
cells loaded into a cell counting chamber or a disposable micro-
fluidic device. These microscopic images can be directly processed
on the smartphone using a custom-written application to deter-
mine the number of cells within a given region of interest.
In this chapter, we introduce the basic operation principles and
the design of this cell-phone based flow-cytometry and optical
174 Hongying Zhu and Aydogan Ozcan
2 Materials
3 Methods
3.1 Cell-Phone First, warm the SYTO16 solution to room temperature and then
Based Flow Cytometry briefly centrifuge it in a microcentrifuge tube to bring the dimethyl
sulfoxide (DMSO) solution to the bottom of the vial. Gently rotate
3.1.1 Fluorescent
the whole blood sample and mix it. After this step, add 20 μL of
Labeling of White Blood
SYTO16 to 200 μL of whole blood. Incubate the whole blood and
Cells in Whole Blood
dye solution mixture in dark for approximately 30 min. To avoid
sedimentation, the sample is gently rotated during incubation.
Opto-Fluidics Based Microscopy and Flow Cytometry on a Cell Phone… 175
Note that no red blood cell lysing step is performed. In addition, since
the intrinsic fluorescence quantum yield of SYTO16 is very low,
the unbound SYTO16 is not separated from the whole blood.
After incubation, dilute the labeled whole blood sample by tenfold
using PBS buffer which is then delivered into our micro-fluidic
chip using a syringe pump with a typical flow rate of 1 μL per min.
3.1.2 Optical Design The cell-phone based flow-cytometry device (see Fig. 1) consists of
for Cell-Phone Based Flow a camera phone and an opto-mechanical attachment, which is
Cytometry designed by autoCAD and printed with a 3D printer using
ABSplus™ modeling thermoplastic material. Sony Ericsson U10I
Aino™, which has an 8 Mpixel color RGB sensor installed on it, is
chosen as the base cell-phone device to build our first-generation
opto-fluidic fluorescent flow cytometry unit (see Note 1). A single
plano-convex lens with a focal length of f2 is placed in front of the
existing lens (f ~ 4.65 mm) of cell-phone camera as shown in Fig. 1
to create an opto-fluidic fluorescent flow cytometry attachment
unit for the cell phone (see Note 2). In the initial experiments, an
aspherical lens with f2 = 4.5 mm is used to get 1× magnification,
which provides us a sufficiently large field-of-view and a good spa-
tial resolution that are needed for white blood cell counting. The
micro-fluidic chip is placed 4.5 mm above the external lens (see
Note 3). During the measurement process, the liquid sample of
interest flows through a PDMS based micro-fluidic chip
(see Fig. 1a, b) using a syringe pump (see Note 4). As shown in
Fig. 1c, blue LEDs are butt-coupled to the two sides of the micro-
channel. In addition to the sample delivery function, this micro-
fluidic chip also works as an opto-fluidic multimode slab waveguide
to guide the excitation light. This opto-fluidic waveguide consists
of three different refractive index layers, i.e., PDMS–liquid–glass.
Since air–glass and air–PDMS interfaces have stronger refractive
index contrast than the glass–liquid or PDMS–liquid interfaces,
the coupled LED excitation light can be tightly confined within
this multimode opto-fluidic waveguide structure to uniformly
excite the labeled cells within the micro-fluidic channel. The fluo-
rescence emission from the labeled cells is then collected perpen-
dicularly to the excitation light path. To further reject the scattered
excitation photons and create a dark-field background for the fluo-
rescent image, an inexpensive plastic filter is placed between the
microchannel and the external lens (see Note 5).
3.1.3 Digital Analysis The Near High Definition (nHD) mode of the Sony Ericsson cam-
of Fluorescent Flow Videos era is chosen to record the fluorescent videos of the flowing blood
cells. Each fluorescent movie that is recorded with the nHD mode
has a resolution of 640 × 352 pixels per frame at a frame rate of
~7–10 frames per second (fps). To count the number of the parti-
cles/cells flowing through our image field of view, these recorded
video frames are post-processed through a PC using a repeating
176 Hongying Zhu and Aydogan Ozcan
3.1.4 White Blood Cell (a) WBCs in fresh whole blood samples were first labeled with
(WBC) Density SYTO®16 fluorescent nucleic acid dyes and then diluted the
Measurement Using labeled blood in PBS buffer using the protocol described in
Cell-Phone Based Subheading 3.1.1.
Opto-Fluidic Fluorescent (b) The diluted and labeled blood samples were then flowed
Flow Cytometry through a disposable micro-fluidic chip using a syringe pump
at a flow rate of ~1 μL/min.
(c) The cell-phone camera continuously recorded a video of fluo-
rescent emission from the flowing WBCs. To improve the
counting accuracy, we defined a cascade of five counters with a
distance separation of ~0.3 mm from each other along the
microchannel; and these digital counters dynamically monitored
the number of WBCs that flow through the microchannel.
(d) To further improve our counting accuracy, the program
counted the number of WBCs that passed through each coun-
ter line over a period of 210 frames and the number of the
counted WBCs from these five independent line counters was
averaged to get the final number of WBCs.
(e) Based on the volume flow rate and the average number of the
counted WBCs, the WBC density (#/μL) in the blood sample
can be estimated. The same process was repeated for 5–6 min
with continuous blood flow and the WBC density for each
sample was plotted as a function of time as shown in Fig. 2a, b.
(f) The average WBC density for each blood sample was calcu-
lated by further averaging each one of these dynamic curves
over 5–6 min. Our WBC density measurements were com-
pared against the standard test results obtained from a com-
mercially available hematology analyzer (i.e., Sysmex KX-21N).
As shown in Fig. 2a, b, cell-phone based flow-cytometry WBC
density measurement results match very well with the standard
test results within <5 % error.
Opto-Fluidics Based Microscopy and Flow Cytometry on a Cell Phone… 177
a b
White blood cell density (# of cells/µL)
6000 9000
5000 8000
4000 7000
Cellphone cytometry WBC counting results Cellphone cytometry WBC counting results
Cellphone cytometry WBC average counting results (4947/µL) Cellphone cytometry WBC average counting results (7354/µL)
3000 Sysmex KX-21N WBC counting results (5000/µL) 6000
Sysmex KX-21N WBC counting results (7800/µL)
2000 5000
0 50 100 150 200 250 300 0 50 100 150 200 250
Time (s) Time (s)
Fig. 2 WBC counting results obtained using our cell-phone based imaging flow-cytometer. (a) For a lower WBC
density sample (5,000 cells/μL) and (b) for a higher WBC density sample (7,800 cells/μL). (Reprinted with per-
mission from Zhu, H., Mavandadi, S., Coskun, A. F., Yaglidere, O., and Ozcan, A., Opto-fluidic Fluorescent Imaging
Cytometry on a Cellphone, Anal. Chem., 83, 6641–6647, 2011. Copyright 2011 American Chemical Society)
a b
7500
Cellphone cytometry WBC
6500 0
6000 Bias= −339
−400
5500
5000 −800
4500
−1200 95% limit
4000
4500 5000 5500 6000 6500 7000 7500 8000 4500 5000 5500 6000 6500 7000 7500 8000
Sysmex KX-21N WBC counting Average of WBC counting by
results (# of cells /µL) both methods (# of cells/µL)
Fig. 3 (a) Comparison of WBC density measurement results obtained with our cell-phone based imaging
cytometer against the results of a commercially available hematology analyzer for 12 patients. The error bar
for each data point is generated from blood density measurements taken over 200 s. (b) The Bland–Altman
analysis results, evaluating the accuracy of the cell-phone based cytometry for WBC density measurements
versus a standard hematology analyzer. (Reprinted with permission from Zhu, H., Mavandadi, S., Coskun, A. F.,
Yaglidere, O., and Ozcan, A., Opto-fluidic Fluorescent Imaging Cytometry on a Cellphone, Anal. Chem., 83,
6641–6647, 2011. Copyright 2011 American Chemical Society)
blood cell (WBC) and red blood cell (RBC) density as well as
hemoglobin concentration. The same opto-fluidic excitation
scheme described in the previous sections is also used in the design
of this automated blood analyzer. An Android OS based cell phone
(Samsung Galaxy SII) is chosen as the base for this automated
blood analyzer prototype. The camera of this phone has an 8
MPixel color (RGB) CMOS sensor installed on it and its built-in
lens has a focal length of f ~ 4 mm. The opto-mechanical attach-
ment for this blood analysis platform consists of two parts
(see Fig. 4): (a) A base attachment fixed on the top of the cell-
phone camera unit, which includes two AA batteries and a univer-
sal port for three different add-on components; and (b) Three
different attachments, each designed for WBC and RBC imaging/
counting and hemoglobin concentration measurement. Each add-
on component has an inexpensive plano-convex lens for forming
the imaging system along with the existing lens of the cell-phone
camera in addition to LEDs for sample illumination. Once the spe-
cific add-on component is clicked into the universal port of the
fixed base, the lens on it gets aligned with the fixed lens on the
cell-phone camera similar to the opto-fluidic flow-cytometry sys-
tem described in the previous section. The existing LEDs on the
attachment are then powered up by the batteries on the base to
illuminate the sample of interest.
Opto-Fluidics Based Microscopy and Flow Cytometry on a Cell Phone… 179
Fig. 4 (A-1) and (A-2) Illustration and picture of the blood analysis platform installed on a cell phone. (B-1) and
(B-2) Illustration and picture of the white blood cell counting and density measurement device. (C-1) and (C-2)
Illustration and picture of the red blood cell counting and density measurement device. (D-1) and (D-2)
Illustration and picture of hemoglobin density measurement device. (Reprinted with permission from Zhu, H.,
Sencan, I., Wong, J., Dimitrov, S., Tseng, O., Nagashima, K., and Ozcan, A., Cost-effective and Rapid Blood
Analysis on a Cell-phone, Lab Chip, 13, 1282–1288, 2013. Copyright Royal Society of Chemistry)
3.2.2 Smart Application Based on an Android phone, we are able to process the captured
for Blood Cell Counting images on the cell phone using a custom designed smart application.
and Hemoglobin This “blood analysis” smart application runs as follows (see Fig. 5):
Concentration
(a) The “blood analysis” application is first initiated by the user by
Measurements
logging into the application and starting a new test.
(b) Once a new test is selected, the user can choose one of the
three options: white blood cell, red blood cell, and hemoglo-
bin measurements. After choosing the type of test, the user will
put the corresponding add-on component onto the cell-phone
base attachment as illustrated in Fig. 4.
(c) After the specific opto-mechanical add-on is inserted to the cell
phone, the user can start to take a picture of the sample of
interest with the cell-phone camera, where the screen of the
phone shows the raw image.
(d) The user can then select the parameters required to process the
raw image. Some of these parameters include: (1) Region of inter-
est (ROI) for analysis. (2) Image pixel size, which is dependent
Fig. 5 Work flow for the smart application installed on the Android cell phone for rapid blood analysis. (Reprinted
with permission from Zhu, H., Sencan, I., Wong, J., Dimitrov, S., Tseng, O., Nagashima, K., and Ozcan, A., Cost-
effective and Rapid Blood Analysis on a Cell-phone, Lab Chip, 13, 1282–1288, 2013. Copyright Royal Society
of Chemistry)
182 Hongying Zhu and Aydogan Ozcan
3.2.3 Imaging Process (a) For the WBC counting process (see Fig. 6), the ROI is cropped
Methods for the Blood Cell from the whole field-of-view of the raw fluorescent image.
Counting and Hemoglobin (b) The cropped region is converted from RGB (red, green, blue)
Concentration channel into HSV (hue, saturation, value) space.
Measurements
(c) The saturation channel is extracted, which has the maximum
intensity contrast between the fluorescent labeled WBCs and
the dark-field background.
Fig. 6 Block diagram of the digital processing steps that are implemented on the cell phone for WBC concen-
tration measurements. (Reprinted with permission from Zhu, H., Sencan, I., Wong, J., Dimitrov, S., Tseng, O.,
Nagashima, K., and Ozcan, A., Cost-effective and Rapid Blood Analysis on a Cell-phone, Lab Chip, 13, 1282–
1288, 2013. Copyright Royal Society of Chemistry)
Opto-Fluidics Based Microscopy and Flow Cytometry on a Cell Phone… 183
Fig. 7 Block diagram of the digital processing steps that are implemented on the cell phone for RBC concentra-
tion measurements. (Reprinted with permission from Zhu, H., Sencan, I., Wong, J., Dimitrov, S., Tseng, O.,
Nagashima, K., and Ozcan, A., Cost-effective and Rapid Blood Analysis on a Cell-phone, Lab Chip, 13, 1282–
1288, 2013. Copyright Royal Society of Chemistry)
Fig. 8 Block diagram of the digital processing steps that are implemented on the cell phone for hemoglobin
concentration measurements. (Reprinted with permission from Zhu, H., Sencan, I., Wong, J., Dimitrov, S.,
Tseng, O., Nagashima, K., and Ozcan, A., Cost-effective and Rapid Blood Analysis on a Cell-phone, Lab Chip,
13, 1282–1288, 2013. Copyright Royal Society of Chemistry)
3.2.4 White Blood Cell 1. Whole blood samples from anonymous donors with WBC
Density Measurement concentrations ranging from ~3,000/μL up to ~12,000/μL
Based on Static were tested with our cell-phone based imager.
Microscopic Imaging 2. For each test, 10 μL of whole blood sample was mixed with
85 μL PBS buffer and 5 μL of nucleic acid staining (SYTO16)
at room temperature for ~20 min in dark.
3. Then 10 μL of this diluted and labeled whole blood sample
was loaded into the standard cell counting chamber that has
channel height of 0.1 mm. The counting chamber was placed
flat for approximately 2 min for the cells to sediment, after
which the cell phone took the microscopic image of the
labeled WBCs using the opto-mechanical attachment shown
in Fig. 4b-1. The fluorescent images of static WBCs were digi-
tally processed with our cell-phone application (running on
Opto-Fluidics Based Microscopy and Flow Cytometry on a Cell Phone… 185
a b
Mean+1.96SD
12000 Linear fit 1000
11000 Y=0.97x-14, R=0.98
800
y=x
10000 ⫾7% 600
(# of cells per mL)
9000 400
Mean=230
8000 200
7000
0
6000
−200
5000
−400 Mean-1.96SD
4000
−600
3000
−800
4000 6000 8000 10000 12000 4000 6000 8000 10000 12000
Sysmex KN21 White Blood Cell Counting Results Average of White Blood Cell Counting Results
(# of cells per mL) Between Cellphone and Sysmex KN21 (# of cells per mL)
Fig. 9 Automated WBC density measurement results of our cell-phone based blood analyzer. (a) Comparison
of cell-phone based WBC density measurement results against the standard test results obtained using
Sysmex KN21 for 30 different blood samples. Each data point was measured three times. (b) The Bland–
Altman analysis results for evaluation of the accuracy of the cell-phone blood analyzer against a standard
hematology analyzer (Sysmex KN21). (Reprinted with permission from Zhu, H., Sencan, I., Wong, J., Dimitrov,
S., Tseng, O., Nagashima, K., and Ozcan, A., Cost-effective and Rapid Blood Analysis on a Cell-phone, Lab Chip,
13, 1282–1288, 2013. Copyright Royal Society of Chemistry)
3.2.5 Red Blood Cell 1. 1 μL of whole blood was mixed with 999 μL PBS buffer to
Density Measurement prepare the diluted blood sample.
Based on Static 2. 10 μL of this diluted whole blood sample was loaded into the
Microscopic Imaging disposable cell counting chamber, which is the same cuvette as
the one used for the WBC counting.
3. Then the specific RBC opto-mechanical attachment shown in
Fig. 4c-1 was used to take bright-field images of the RBCs
after cell sedimentation. These bright field images of the
RBCs within the sample cuvette were then further processed
186 Hongying Zhu and Aydogan Ozcan
a b
6.0x106 5
Y=0.96x+2.5x10 , R=0.98
y=x
3x105 Mean+1.96SD
5.5x106
⫾5% 2x105
(# of cells per mL)
5.0x106 1x105
4
4.5x106 0 Mean= -2.9x10
−1x105
4.0x106
−2x105
3.5x106
−3x105 Mean-1.96SD
3.0x106
−4x105
3.0x106 3.5x106 4.0x106 4.5x106 5.0x106 5.5x106 6.0x106 3.0x106 3.5x106 4.0x106 4.5x106 5.0x106 5.5x106 6.0x106
Sysmex KN21 Red Blood Cell Counting Results Average of Red Blood Cell Counting Results
(# of cells per mL) Between Cellphone and Sysmex KN21 (# of cells per mL)
Fig. 10 Same as Fig. 9, except for RBC density measurements. Each data point in (a) was measured three
times. (Reprinted with permission from Zhu, H., Sencan, I., Wong, J., Dimitrov, S., Tseng, O., Nagashima, K., and
Ozcan, A., Cost-effective and Rapid Blood Analysis on a Cell-phone, Lab Chip, 13, 1282–1288, 2013. Copyright
Royal Society of Chemistry)
3.2.6 Hemoglobin 1. The test cuvette was filled with DI water and measured its
Concentration transmission light intensity over 9 mm2 (1000 × 1000 pixels)
Measurement Based yielding I0.
on Static Microscopic 2. Lysed blood samples were prepared by mixing 10 μL of whole
Imaging blood sample with 90 μL RBC lysing buffer solution to lyse
the RBCs. After the RBCs were lysed completely, another
900 μL of PBS buffer was added to further dilute the lysed
sample.
3. Then this diluted sample was loaded into a new cuvette and
the transmission light intensity was measured over the same
area, this time yielding I.
4. The absorbance value (A) for each blood sample was calcu-
lated by using I0 and I values. An internal linear calibration
curve/equation was generated by measuring 60 different
human blood samples with known hemoglobin concentrations
Opto-Fluidics Based Microscopy and Flow Cytometry on a Cell Phone… 187
a b
Difference of Hemoglobin Concentration Results Measured
Cellphone Measured Hemoglobin Concentration (g/dL)
18 1.0
Linear fit
by Sysmex KN21 and Cellphone (g/dL)
Mean+1.96SD
Y=1.05x-0.97, R=0.92
y=x 0.5
16
⫾5%
Mean=0.036
0.0
14
−0.5 Mean-1.96SD
12
−1.0
12 14 16 18 12 13 14 15 16 17
Sysmex KN21 Measured Hemoglobin Average of Hemoglobin Concentration Results Measured
Concentration (g/dL) by Sysmex KN21 and Cellphone (g/dL)
Fig. 11 Same as Fig. 9, except for hemoglobin density measurements. Each data point in (a) was measured
three times. (Reprinted with permission from Zhu, H., Sencan, I., Wong, J., Dimitrov, S., Tseng, O., Nagashima,
K., and Ozcan, A., Cost-effective and Rapid Blood Analysis on a Cell-phone, Lab Chip, 13, 1282–1288, 2013.
Copyright Royal Society of Chemistry)
188 Hongying Zhu and Aydogan Ozcan
4 Notes
References
1. Beck N (2009) Diagnostic hematology. device development and education applications.
Springer, Berlin PLoS One 6:e17150
2. Mellors JW, Munoz A, Giorgi JV, Margolick 10. Martinez AW, Philips ST, Carrilho E, Thomas
JB, Tassoni CJ (1997) Plasma viral load and SW, Sindi H, Whitesides GW (2008) Simple
CD4+ lymphocytes as prognostic markers of telemedicine for developing regions: camera
HIV-1 infection. Ann Intern Med 126: phones and paper-based microfluidic device for
946–954 real-time, off-site diagnosis. Anal Chem 80:
3. Wurm EM, Wellenhof RH, Wurm R, Soyer HP 3699–3707
(2008) Telemedicine and teledermatology: 11. Zhu H, Sikora U, Ozcan A (2012) Quantum
past, present and future. J Dtsch Dermatol Ges dot enabled detection of Escherichia coli using
6:106–112 a cell-phone. Analyst 137:2541–2544
4. Pamplona VF, Mohan A, Oliveira MM, Raskar 12. Mudanyali O, Dimitrov S, Sikora U,
R (2010) NETRA: interactive display for self- Padmanabhan S, Navruz I, Ozcan A (2012)
evaluation of an eye for visual accommodation Integrated rapid-diagnostic-test reader plat-
and focal range. Proc. of Frontiers in Optics, form on a cellphone. Lab Chip 12:2678–2686
2010, Paper FTuB4. 13. Preechaburana P, Gonzalez MC, Suska A,
5. Breslauer DN, Maamari RN, Switz NA, Lam Flippini D (2012) Surface plasmon resonance
WA, Fletcher DA (2009) Mobile phone based chemical sensing on cell phones. Angew Chem
clinical microscopy for global health applica- Int Ed 12:11585–11588
tions. PLoS One 4:e6320 14. Coskun A, Wong J, Khodadah D, Nagi R, Tey
6. Tseng T, Mudanyali O, Oztoprak C, Isikman A, Ozcan A (2012) A personalized food aller-
SO, Sencan I, Yaglidere O, Ozcan A (2010) gen testing platform on a cellphone. Lab Chip
Lensfree microscopy on a cell-phone. Lab Chip 13:636–640
10:1787–1792 15. You DJ, Park TS, Yoon J (2013) Cell-phone-
7. Zhu H, Yaglidere O, Su T, Tseng D, Ozcan A based measurement of TSH using Mie scatter
(2010) Cost-effective and compact wide-field optimized lateral flow assays. Biosens Bio-
fluorescent imaging on a cellphone. Lab Chip electron 40:180–185
11:315–322 16. Navruz I, Coskun AF, Wong J, Mohammad S,
8. Zhu H, Mavandadi S, Coskun AF, Yaglidere Tseng D, Nagi R, Phillips S, Ozcan A (2013)
O, Ozcan A (2011) Opto-fluidic fluorescent Smart-phone based computational microscopy
imaging cytometry on a cellphone. Anal Chem using multi-frame contact imaging on a fiber-
83:6641–6647 optic array. Lab Chip 13:4015–4023
9. Smith ZJ, Chu K, Espenson AF, Rahimzadeh 17. Zhu H, Sencan I, Wong J, Dimitrov S, Tseng
M, Gryshuk A, Molinaro M, Dwyre DM, Lane O, Nagashima K, Ozcan A (2013) Cost-
S, Mattews D, Wachsmann-Hogiu S (2011) effective and rapid blood analysis on a cell-
Cell-phone-based platform for biomedical phone. Lab Chip 13:1282–1288
190 Hongying Zhu and Aydogan Ozcan
18. Balsam J, Rasooly R, Bruck HA, Rasooly A 21. Kiesel P, Beck M, Johnson N (2011)
(2014) Thousand-fold fluorescent signal Monitoring CD4 in whole blood with an opto-
amplification for mHealth diagnostics. Biosens fluidic detector based on spatially modulated
Bioelectron 51:1–7 fluorescence emission. Cytometry A 79A:
19. International Telecommunication Union, 317–324
Market information and statistics (2013). 22. Suzuki S, Abe K (1985) Topological structural
http://www.itu.int/en/ITU-D/Statistics/ analysis of digital binary image by border fol-
Documents/facts/ICTFactsFigures2013.pdf lowing. Vis Graph Image Process 30:32–46
20. Schmidt O, Bassler M, Kiesel P, Knollenberg 23. Ozcan A, Demirci U (2008) Ultra wide-field
C, Johnson N (2007) Fluorescent spectrometer- lens-free monitoring of cells on-chip. Lab Chip
on-a-fluidic-chip. Lab Chip 7:626–629 8:98–106
Chapter 13
Abstract
A unique optofluidic lab-on-a-chip device that can detect optically encoded forward scattering signals is
demonstrated. With a unique design of a spatial mask that patterns the intensity distribution of the illumi-
nating light, the position and velocity of each travelling cell in the flow can be measured with submicrom-
eter resolution, which enables the generation of a cell distribution plot over the cross section of the
channel. The distribution of cells is highly sensitive to its size and stiffness, both being important biomark-
ers for cell classification without cell labelling. The optical-coding technique offers an easy route to classify
cells based on their size and stiffness. Because the stiffness and size of neutrophils are distinct from other
types of white blood cells, the number of neutrophils can be detected from other white blood cells and red
blood cells. Above all, the enumeration of neutrophil concentration can be obtained from only 5 μL of
human blood with a simple blood preparation process saving the usual steps of anticoagulation, centrifuga-
tion, antibody labelling, or filtering. The optofluidic system is compact, inexpensive, and simple to fabri-
cate and operate. The system uses a commodity laser diode and a Si PIN photoreceiver and digital signal
processing to extract vital information about cells and suppress the noise from the encoded optical scatter-
ing signals. The optofluidic device holds promise to be a point-of-care and home care device to measure
neutrophil concentration, which is the key indicator of the immune functions for cancer patients undergo-
ing chemotherapy.
Key words Optical-coding technique, Neutrophils, White blood cells, Microfluidics, Optofluidic,
Point-of-care, Labsel-free detection
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_13, © Springer Science+Business Media New York 2015
191
192 Tsung-Feng Wu and Yu-Hwa Lo
Fig. 1 (a) Equilibrium position of cells affected by the inertial effect, which mainly depends on the size of cells.
The larger cells experience a greater lift force that pushes them toward the center of the microchannel. (b) and
(c) show the effect of drag force affected by the velocity gradient (UA–UB) normal to the cell travel direction. The
higher deformability of softer cells makes them nearer the center of the microchannel, thus travelling at a
higher speed, than those stiffer cells
194 Tsung-Feng Wu and Yu-Hwa Lo
of cells are determined by the balance of two forces, lift force due
to the inertial effect and fluidic dynamic drag force. Larger cells
experience a greater inertial force which pushes the cells farther
away from the channel wall (Fig. 1a). On the other hand, the
greater deformability of softer cells enables a better alignment of
the cell orientation with the streamline, so softer cells experience
less drag force than stiffer cells and tend to be closer to the center
of the channel (Fig. 1b, c). As a result, the combined effects of
larger size and greater deformability separate neutrophils from
the other WBCs by their travel speed and their location in the
microchannel.
The optically encoded microfluidic device demonstrated in this
work consists of a straight microfluidic channel with spatial pat-
terns that modulate the incident light illuminating each cell pass-
ing the patterns. The spatial pattern converts the forward scattering
signal of each cell into an intensity-modulated temporal signal
according to its velocity and position [3]. As shown in Fig. 2, cells
within the microfluidic channel produce forward scattering signals
of unique features in their waveforms. For example, if the trajec-
tory of cells passing through the sensing area is close to the side
wall of the microchannel (e.g., trajectory 1 or 3 in Fig. 2), the wave
form of the forward scattering signal will be different from the
forward scattering signal for cells travelling through the center of
the microchannel (e.g., trajectory 2 in Fig. 2). Therefore, from the
ratio of W2 to W1 or W3 to W4 in the forward scattering signal, we
can find the trajectory of each cell. Here we use two pairs of
patterned slits, (W1,W2) and (W3,W4), as opposed to a single pair to
Fig. 2 Illustrations of different scattering signal waveforms from cells taking different travel paths in the micro-
fluidic channel. The white areas represent the transparent trapezoidal slits. The bases of each trapezoidal slit
are 50 μm and 100 μm, as indicated in the figure. The solid (1), semi-dashed (2) and dashed (3) red lines are
exemplary cell paths over the sensing area. The waveforms of the forward scattering signal of the cells taking
the paths 1, 2, 3 are illustrated on the right, with the characteristic widths W1, W2, W3, W4 in the signals. Such
information, as explained in the text, allows us to find the travel speed and position for each single cell within
the microchannel (Color figure online)
Optofluidic Device for Label-Free Cell Classification from Whole Blood 195
2 Materials
2.1 System 1. Laser beam (λ = 488 nm (or 650 nm) diode laser, 40 mW,
for Detection Spectra-physics).
of Space-Time 2. Silicon photoreceiver (PDA36A, Thorlabs, NJ, USA).
Coded Optical
3. Monochrome CCD camera, C-mount (XC-ST30, Sony, Japan).
Scattering Signal
4. XY translation stage with standard micrometers (460A-XY,
Newport, CA, USA).
5. Optical table.
6. LabView Express (2010, National Instruments, TX, USA).
7. DAQ Broad (BNC-2110, National Instruments, TX, USA).
8. Syringe pump (11 Elite, Harvard Apparatus, MA, USA).
2.2 Sample Solution 1. 10× red blood cell lysing buffer for multi-species (00-4300-54,
eBioscience, CA, USA) is diluted with deionized water to form
1× lysing buffer before RBC lysing.
2. 10 mM ethylenediaminetetraacetic acid (EDTA), 1 wt% bovine
serum albumin (BSA), and 1× phosphate buffered saline (PBS)
are mixed as the cell suspension solution.
3. Whole blood is collected from healthy donors by using a finger
prick (see Note 1).
2.3 Device Material 1. Polydimethylsiloxane kit (Sylgard 184, Dow Corning) was
purchased directly and mixed prior to use.
196 Tsung-Feng Wu and Yu-Hwa Lo
3 Methods
3.1 Setup A 488 nm (or 650 nm) laser light is introduced from the bottom
of the Microfluidic of the microfluidic device. The CCD camera is used to assure align-
Device ment between the laser beam and the spatial patterns on the glass
slide to let the laser beam illuminate cells inside the microchannel.
The schematic and photo image of the system are shown in Fig. 3a, b,
respectively. The laser light and CCD camera have been aligned
with each other. In the actual operation, the microfluidic device is
placed on the XY translational stage for device alignment. An
attenuator is inserted in front of the laser source to avoid saturating
the CCD camera. By adjusting the XY translation stage, the entire
pattern is uniformly illuminated by the laser beam as shown in
Fig. 3c. Figure 3c also shows the exact features of the spatial mask
pattern, where four trapezoidal slits are aligned with the 100 μm
Fig. 3 (a) The schematic of the optical-coding system, consisting of a PIN photoreceiver, a diode laser, a micro-
fluidic device, and a computation system for data acquisition and analysis. (b) The actual image of the optical-
coding system shows the detail of the optics setup. (c) The CCD image of the spatial patterns on the microfluidic
device under uniform laser illumination. The red arrows show the channel width. (d) A time-domain encoded
forward scattering signal from a cell collected by a silicon PIN photodetector (Color figure online)
Optofluidic Device for Label-Free Cell Classification from Whole Blood 197
2. PDMS Replica.
(a) PDMS (Sylgard 184, Dow Corning) is mixed with the cur-
ing agent at the weight ratio of 15:1.
(b) The pre-polymer mixture is poured onto the SU-8 mold
on silicon wafer and baked in the oven at 60 °C for 4 h.
(c) The PDMS replica can be easily cut off with the blazer for
further use.
3. Mask Pattern.
(a) Mask silts design: Four transparent trapezoidal slits are
designed as shown in Fig. 2. Each individual trapezoidal
slit has its base lengths of 50 μm and 100 μm and is sepa-
rated from the neighboring silts by 50 μm. The total pat-
tern length for the sensing area is 450 μm.
(b) Negative photoresist, NR9-1500, is spun on a 1 × 2 in.
glass substrate at 4,000 rpm for 30 s.
(c) After the soft bake at 150 °C for 1 min, the glass slide is
exposed under I-line for 90 s.
(d) As-exposed glass slide is placed on a hotplate at 100 °C for
1 min, followed by immersion of the slide in the developer
solution for 6 s. Rinse with deionized water after
development.
(e) A spatial mask is formed by sputtering a Ti/Au
(100 nm/200 nm) metal film on the patterned glass, fol-
lowed by the lift-off process to form the four trapezoidal
slit patterns.
4. Assembly of microfluidic devices.
(a) The PDMS replica from Subheading 3.2, step 2 and the
patterned glass slide from Subheading 3.2, step 3 are
treated with ozone plasma.
(b) After the ozone plasma treatment, the PDMS replica is
bonded onto the patterned glass slide with careful align-
ment to form the microfluidic device.
3.3 Determination 1. Use a finger prick to take 5 μL of whole blood from healthy
of the Number donors. The amount of blood is calibrated with a micropipette.
of Neutrophils 2. The 5 μL whole blood is first diluted with 1 mL of buffer solu-
tion described in Subheading 2.2, item 2.
3. Without any centrifuge, 500 μL of RBC lysing buffer is added
to 150 μL of diluted whole blood sample to dilute the white
blood cell concentration by 870 times.
Optofluidic Device for Label-Free Cell Classification from Whole Blood 199
a 140 b 20
19 30
120 18
Position along
100 V>30cm/s 17
y-axis (mm)
16 20
Cell Count
80 15
60 14
13 10
40
12 Neutrophils
20 11
10 0
0
0 20 40 60 80 100
15 20 25 30 35 40
Velocity (cm/s) Position along x-axis (mm)
c400000
Neutrophils
300000
SSC-A
200000
Monocytes
100000 Lymphocytes
0
0 300000 600000 900000 1200000 1500000
FSC-A
Fig. 4 (a) Travelling speed histogram of white blood cells from 5 μL RBC-lysed whole blood sample. The histo-
gram indicates a distinguishable population of neutrophils due to their high travel speed resulted from the
higher deformability. The event, which speed is less than 30 cm/s, is the signal from other WBC types plus RBC
residues since the sample did not completely filter out by the centrifuge. (b) The spatial distribution of white
blood sample over the cross section of the channel. A separate band for neutrophils (as the gating shows)
separates them from the other cells. Due to the property of symmetry, only the upper half of the channel cross
section is shown. (c) Scatter plot of 5 μL diluted whole blood sample measured with a commercial flow cytom-
eter (Accuri C6)
4. After 10 min in the ambient, the Tygon tubings (ID: 0.02 in.
OD: 0.03 in.) are inserted into the inlet and outlet of
microfluidic devices. The diluted white blood cell sample is
directly introduced into the microfluidic device by using a
syringe pump (Pump Elite 11, Harvard Apparatus, MA,
USA) at the flow rate of 75 μL/min.
5. Record signals from the white blood cell sample for 90 s, which
means a sample volume of 112.5 μL is interrogated.
6. Digital signal processing is applied to obtain the speed of each
cell, as shown in Fig. 4a. The neutrophils have faster speed
than other WBCs.
7. In combination with the cell position information, the velocity
of cell can be further converted into cell distribution over the
cross section of the channel, as shown in Fig. 4b.
200 Tsung-Feng Wu and Yu-Hwa Lo
Table 1
Summary of test results from 870× diluted RBC-lysed blood samples
using our device and a commercial flow cytometer (Accuri C6)
Trial I II III IV
Our device (neutrophil 5.80 7.74 5.72 6.20
counts/μL)
Accuri C6 (neutrophil 4.38 7.09 6.39 6.77
counts/μL)
8. Repeat the above procedures for four samples from the healthy
donors and measure the samples with a commercial flow
cytometer (Accuri C6) as shown in Fig. 4c.
9. The comparison between the optofluidic device and the com-
mercial flow cytometer is shown in Table 1.
4 Notes
References
1. Cheung K, Gawad S, Renaud P (2005) Impedance 4. Keisel P, Bassler M, Beck M, Johnson N (2009)
spectroscopy flow cytometry: on-chip label-free Spatially modulated fluorescence emission from
cell differentiation. Cytometry A 65A:124–132 moving particles. Appl Phys Lett 94:041107
2. Sipsas NV, Bodey GP, Kontoyiannis DP (2005) 5. Mei Z, Wu TF, Pion-Tonachini L, Qiao W, Zhao
Perspectives for the management of febrile neu- C, Liu ZW, Lo YH (2011) Applying an optical
tropenic patients with cancer in the 21st century. space-time coding method to enhance light scat-
Cancer 103:1103–1113 tering signals in microfluidic devices.
3. Wu TF, Mei Z, Pion-Tonachini L, Zhao C, Qiao Biomicrofluidics 5:034116
W, Arianpour A, Lo YH (2011) An optical- 6. Wu TF, Mei Z, Lo YH (2012) Optofluidic
coding method to measure particle distribution device for label-free cell classification from whole
in microfluidic devices. AIP Adv 1:022155 blood. Lab Chip 12:3791–3797
Chapter 14
Abstract
A portable chemical sensing system that integrates sample preconcentration, separation and detection as
well as wireless communication functionalities into a compact, wearable format can provide continuous
and real-time monitoring of volatile organic compounds in the environment. The sensing modality relies
on tuning forks coated with molecularly imprinted polymers that, in conjunction with sample preconcen-
tration, offer selective detection down to parts-per-billion levels. The use of capillary columns allows indi-
vidual components of complex mixtures to be detected at these highly sensitive levels even in the presence
of interferents. The wireless capability facilitates the utilization of a paired smartphone as the user interface
as well as a vehicle for additional processing and storage of the measured data. This integrated approach
offers a cost-effective and reliable platform for personal exposure assessment.
Key words Environmental monitoring, Chemical sensors, Volatile organic compounds, Wearable
detection system, Wireless sensing, Personal exposure assessment
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_14, © Springer Science+Business Media New York 2015
201
202 Cheng Chen et al.
Air Interference
inlets V1 filter V2 V3
Zero
Precon-
Electronics
filter V4
centrator circuitry
Pump GC
and
column
batteries
Fig. 1 A schematic representation of an integrated chemical detection system, consisting of modules for: flu-
idic and thermal control (with a pump. filters and valves V1–V4), sample preconcentration, sample separation
(by a gas chromatographic (GC) column), sample detection (by tuning fork sensors), and electronics
2 Materials
2.1 Sample 1. Stainless steel tubing (outer diameter: 0.0625 in., inner
Preconcentration diameter: 0.0335 in.).
Module 2. Graphitized carbon black (Sigma-Aldrich Co. LLC, St. Louis,
MO).
3. Thermally conductive compound (Resbond 920, Cotronics
Corp., Brooklyn, NY).
4. Heating wire (Kanthal Nikrothal 80 Plus, 35 Ω/m; Sandvik
Materials Technology, Clarks Summit, PA).
2.4 Fluidic 1. Micro vane pump (SP 135 FZ-LC; Schwarzer Precision USA,
and Thermal Control Madison, CT).
Module 2. Miniature three-way valves, total of 4 (LHLA0531211H; The
Lee Co., Essex, CT).
3. Particle filter (custom-made with commercial glass fiber).
4. Zeroing filter (custom-made from activated carbon).
5. Interference filter (custom-made using polyester fiber).
6. PTFE tubing (EW-06605-27, outer diameter: 0.125 in., inner
diameter: 0.0625 in.; Cole-Parmer, Vernon Hills, IL).
7. Connectors (L410-J1A, elbow tube fitting; Value Plastics,
Inc., Fort Collins, CO).
2.6 User Interface 1. Smartphone (Q9h; Motorola Mobility LLC, Chicago, IL).
2. Application software (custom-made using Microsoft Visual
Studio).
A Wearable Sensing System for Assessment of Exposures… 205
3 Methods
3.1 Monitoring The preconcentrator is built using a 1-in. section of stainless steel
System Setup tubing with an outer diameter of 0.0625 in. The tubing is packed
with about 3 mg of the graphitized carbon black to provide a high
3.1.1 Preconcentration
density of binding sites for adsorbing VOCs. The outside of the
Module
tubing was coated with a thermal conductive material, and a heat-
ing wire was wrapped around the tubing. Then another layer of the
same thermal conductive material was coated on the heating wire
along with a thermistor. The resistance value of the thermistor is
monitored and converted into a temperature reading by the elec-
tronics module, which is used to turn on and off the heating wire
to control the 300 °C temperature needed to facilitate desorption
of the VOCs that have been collected.
3.1.2 Separation Module The type and configuration of the stainless steel capillary columns
used in the sample separation module are dependent on the
intended air-monitoring application. For fast separation and detec-
tion of VOCs, a Carbowax-coated column and a cyanopropyl phe-
nyl silicone-coated column are connected in series.
3.1.3 Detection Module The MIP used to coat the prongs of the TF sensors is designed to
provide selective detection of mono-aromatic and alkyl hydrocar-
bon VOCs. For the detection of BTEX, a polystyrene-based MIP
is synthesized by polymerizing divinylbenzene. A mixture of ethyl-
benzene and xylene is used as the porogenic solvent.
The MIP coating on the TFs is formed using the following
procedure:
1. The TFs are immersed in toluene containing 10 % (v/v) phen-
yltrimethoxysilane for 24 h to form a hydrophobic surface layer.
2. TFs are then rinsed with toluene and dried with clean air.
3. Next, the TFs are dipped in 20 mM dodecanethiol in isopro-
panol for 60 min to make the quartz surfaces and the elec-
trodes of the TFs hydrophobic.
4. The TFs are dip-coated with the MIP (see Note 2).
5. The TFs are mounted onto the sensor cartridge.
3.1.4 Fluidic The various components in this module are connected to those in
and Thermal Control the previously described modules using flexible PTFE tubing and
Module tube fittings (see Note 3). Air samples are drawn into the monitor-
ing system via a small vane pump (see Note 4). A three-way valve
(V1, Fig. 1) controls whether the air is drawn through a particle
filter (during detection mode) or a zero filter (during calibration
mode). The interference filter absorbs acid vapors from the sampled
206 Cheng Chen et al.
air to avoid interfering with the tuning fork sensors. The pump and
valves, as well as the heater for the preconcentrator, are powered
and controlled through the electronics module described below.
3.1.5 Electronics Module Custom designed and built circuitry is used to control all the mod-
ules within the integrated device. Figure 2 shows a block diagram
for the operation of the detection and wireless communication
modules. The circuitry includes a precision digital counter to mea-
sure the responses of the TFs in the sensor array, and a Bluetooth
chip for the wireless communication with the smartphone that is
utilized as the user interface.
1. Each TF in the sensor array is driven with an oscillator driver
chip and then multiplexed sequentially to the clock input of a
microcontroller (MCU1, Fig. 2). Microcontroller 1 uses the
clock signal to generate a pulse with duration of 32,768 clock
cycles. This pulse is sent to the inputs of the analog-to-digital
converter (ADC) of a second microcontroller (MCU2).
2. The input signal is compared against the reference clock cycle
with a much higher frequency (in the several-MHz range) that
is generated by MCU2. This allows the shift in the resonant
frequency of a TF sensor to be measured with very high resolu-
tion (a few mHz).
MCU1
ADC/
GPIO
MUX
Computation UART BT
RX TX
TX RX
Sensor
Array
MCU2
Fig. 2 A block diagram that illustrates the operational scheme for the sample detection and wireless commu-
nication modules. Microcontrollers (MCUs) handle conversion of the sensor data from analog into digital for-
mat, as well as control of components in the fluidic and thermal control module. The Bluetooth (BT) chip
communicates with its counterpart within the smartphone used as the user interface to the detection system
A Wearable Sensing System for Assessment of Exposures… 207
Fig. 3 (a) Photograph of the enclosure holding all the components of the integrated detection system, with the
smartphone utilized as the user interface on top. (b) Photograph of the smartphone screen showing a separa-
tion chromatogram of a BTEX mixture detected by the system
3.1.6 User Interface A smartphone running the Windows operating system (Motorola
Q9h) is used as the interface to the integrated device (Fig. 3a). A
custom application written using Java programming language is
loaded onto the phone to control the electronics in the integrated
device. After receiving the sensor data, the smartphone also pro-
vides the functions of additional processing, displaying, and stor-
age of the data.
3.2 Device Operation Operation of the device, as described below, is fully automated.
Initiation of a measuring cycle as described below is accomplished
by pushing a button on the smartphone.
3.2.1 Preconcentration 1. The pump turns on and the valves open to the configuration to
draw an air sample through the particle filter into the precon-
centrator. The duration of this step is adjusted to account for
the anticipated VOC concentration. The use of a longer pre-
concentration period usually produces larger signal peaks for a
given analyte concentration in the sampled air, due to the lon-
ger time for accumulation and interaction of the analyte with
the carbon black (see Note 5).
208 Cheng Chen et al.
2. Valves are then switched to draw air cleaned by the zero filter
to flow through the separation column and over the TF sen-
sors for 1 min. This allows the sensors to establish a baseline
reading.
3.2.3 Sample Injection 1. The valves are switched to flow filtered clean air through the
hot preconcentrator to transport the VOC vapors to the sepa-
ration column.
2. This injection stage has a 15-s duration.
3.2.4 Data Collection 1. As the components of the air sample are separated in the capil-
and Analysis lary column, they are transported to the TF sensors for
measurement.
2. Sensor responses are wirelessly sent to the smartphone where
the application software analyzes the data and displays the
measured results (Fig. 3b).
3.2.5 System Cleaning 1. Upon completion of the previous stage, the preconcentrator is
heated again.
2. Filtered clean air is flowed through the system to flush it clean
and to prepare it for the next measuring cycle.
3.3 Field Testing The wearable detection system was tested at the Shell Oil refinery
in Martinez, California. In this case, the system was configured
3.3.1 Exposure
with a long capillary column (19 m) to counter the high concen-
to Petrochemical Products
trations of hydrocarbon interferents present in the atmosphere in
in an Industry Setting
addition to the BTEX vapors targeted for measurement. The sys-
tem was operated as described in Subheading 3.2. As can be seen
in Fig. 4a, low levels of benzene vapor (5 ppb) can be clearly
resolved from n-hexane with a concentration 1,000 times higher
also present in the sampled air. The separation (elution) time here
was ~8 min. To detect all the individual components of BTEX,
longer elution times are needed (Fig. 4b). Note that all three iso-
mers of xylene can be resolved. An analyte with low vapor pressure
or high interaction with the coating of the capillary column will
result in a longer elution time. For a given analyte, slower elution
times are also encountered when longer capillary columns are used
to obtain higher separation resolution of complex samples with
multiple components, as in the case here for gasoline vapors.
3.3.2 Mapping BTEX The environment at a number of locations with varying amounts
Exposure to Local Activities of traffic and different activities in the metropolitan area of Phoenix,
and Environmental Arizona were tested for BTEX using the wearable detection
Conditions system. The system was operated as described in Subheading 3.2.
A Wearable Sensing System for Assessment of Exposures… 209
a b
9 benzene 35
30 benzene
toluene m-xylene
25
6
20
ethyl-
n-hexane 15 benzene p-xylene
3 o-xylene
10
5
0 0
5 6 7 8 10 20 30 40 50 60
Time (min) Time (min)
Fig. 4 Separation and detection of air samples containing BTEX vapors and related interferents. (a) Detection
of benzene at a concentration of 5 ppb, after the separation of n-hexane interferents with a concentration of
5 ppm. (b) The separation chromatogram for all the other components of the BTEX vapors sampled. Note that
all three isomers of xylene are resolved
4 Notes
1- School
> 5 ppb
B = 0 ppb E = 0 ppb
T = 0 ppb X = 0 ppb 7- Casino
2- Mall - Parking
B = 0 ppb E = 0 ppb
T = 10 ppb X = 0 ppb
B = 2.4 ppb E= 0 ppb
T = 0 ppb X= 0 ppb
6- Gas Station
3- Airport
B = 5 ppb E = 0 ppb
T = 0 ppb X = 0 ppb
B = 1.5 ppb E = 0 ppb
T = 0 ppb X = 0 ppb 4- School 5- Highway Traffic
Fig. 5 Correlation of BTEX exposures to sampling locations (within the metropolitan area of Phoenix, Arizona)
on Google Map, making use of GPS-based locationing capability in the smartphone. High levels of benzene and
toluene are found at a gas station and inside a gambling casino, respectively
2. The mass of the MIP coatings ranged from 0.5 to 3 μg, resulting
in a variation of within 20 % for the sensitivities of the TFs.
3. It is important to minimize dead volumes associated with the
connections between all the components involved in gas
handling.
4. An air flow rate of 8 mL/min is found to be optimal for the
current system configuration.
5. For a BTEX mixture with a concentration of 20 ppb for each
component, a preconcentration time of 20 min is sufficient. In
the case of a component concentration of 10 ppm, there is no
need for preconcentration.
Acknowledgments
References
1. Gauderman WJ, Avol E, Lurmann F, Kuenzli 7. Ren M, Forzani ES, Tao N (2005) Chemical
N, Gilliland F, Peters J, McConnell R (2008) sensor based on microfabricated wristwatch
Childhood asthma and exposure to traffic and tuning forks. Anal Chem 77:2700–2707
nitrogen dioxide. Epidemiology 16:737–743 8. Tsow F, Forzani E, Rai A, Wang R, Tsui R,
2. Carrieri M, Tranfo G, Pigini D, Paci E, Mastroianni S, Knobbe C, Gandolfi AJ, Tao
Salamon F, Scapellato ML, Fracasso ME, NJ (2009) A wearable and wireless sensor sys-
Manno M, Bartolucci B (2010) Correlation tem for real-time monitoring of toxic environ-
between environmental and biological moni- mental volatile organic compounds. IEEE
toring of exposure to benzene in petrochemi- Sensors J 9:1734–1740
cal industry operators. Toxicol Lett 192: 9. Iglesias RA, Tsow F, Wang R, Forzani ES, Tao
17–21 N (2009) Hybrid separation and detection
3. Schweigkofler M, Niessner R (1999) device for analysis of benzene, toluene, ethyl-
Determination of siloxanes and VOC in land- benzene, and xylenes in complex samples. Anal
fill gas and sewage gas by canister sampling and Chem 81:8930–8935
GC-MS/AES analysis. Environ Sci Technol 10. Lieberzeit PA, Gazda-Miarecka S, Halikias K,
33:3680–3685 Schirk C, Kauling J, Dickert FL (2005)
4. RAE Systems (http://www.raesystems.com/ Imprinting as a versatile platform for sensitive
products/colorimetric-gas-detection-tubes) materials – nano-patterning of the polymer
5. Freedman AN (1980) The photoionization bulk and surfaces. Sens Actuators B
detector: theory, performance and application 111–112:259–2632
as a low-level monitor of oil vapour. J 11. Chen C, Tsow F, Driggs Campbell K, Iglesias
Chromatogr A 190:263–273 R, Forzani E, Tao N (2013) A wireless hybrid
6. Boussaad S, Tao NJ (2003) Polymer wire chemical sensor for detection of environmental
chemical sensor using a microfabricated tuning volatile organic compounds. IEEE Sensors J
fork. Nano Lett 3:1173–1176 13:1748–1755
Chapter 15
Abstract
A paper-based microfluidic device was used to quantitatively detect active enzyme analytes in samples at
mid to low femtomolar levels. The device uses a hydrophobic oligomer that controls the wetting proper-
ties of the paper within the device. When the target analyte is present within the sample, the oligomer
depolymerizes, thus switching the paper to hydrophilic, allowing for the sample to wick through the
device. Measuring the time for the sample to wick to a control region relative to an assay region within the
device results in sensitive, quantitative measurements of the target enzyme (e.g., alkaline phosphatase or
β-d-galactosidase). This device requires the use of only a timer for quantifying a target analyte, and thus
the platform may be appropriate for use in resource-limited environments, where access to expensive diag-
nostic equipment is limited. A smartphone with integrated application software (the software has yet to be
developed) could be used for timing the assay and for relating the time measurement to the quantitative
readout for the assay. In future versions of this assay, it should be possible to configure the smartphone to
start and stop the time-based measurement to further simplify the assay for the user.
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_15, © Springer Science+Business Media New York 2015
213
214 Gregory G. Lewis and Scott T. Phillips
Fig. 1 Depiction of a paper-based microfluidic device used for quantitatively detecting active enzyme analytes,
as well as the reagents used in the assays. (a) Picture of the paper-based microfluidic device. The device is
made from stacked layers of patterned paper held together with spray adhesive and then laminated. The
dimensions of the paper portion of the device are 20 mm × 10 mm × 1.8 mm, the black regions are hydropho-
bic wax and the white regions are hydrophilic paper. (b) Cross-section of the device shown in (a), along the
white dotted line. The left channel in (b) is the assay channel and the right channel is the reference channel.
(c) A hydrophobic oligomer is incorporated into the device (layer 4 in (b)) and provides an amplified signal for
detection events. The oligomer depolymerizes from head-to-tail in response to hydrogen peroxide that is gen-
erated from the detection event (d). (d) Reagents are incorporated into the device to provide selective detection
of the target enzyme analyte. The enzyme reacts with the reagent and produces glucose, which is converted
to hydrogen peroxide within the device
216 Gregory G. Lewis and Scott T. Phillips
into the channel and generate glucose, but hydrogen peroxide will
not be generated, therefore hydrogen peroxide will not be present
to react with the oligomer. Hence, the time required for the sam-
ple to pass through this control region (and carry the green color
to the top of the device) depends on the temperature and humidity
under which the assay is conducted, as well as on the viscosity of
the sample. These factors will affect sample distribution rates in the
left region as well, and therefore this control region normalizes the
output of the assay for the effects of these variables on sample dis-
tribution. This normalization is implemented by measuring the
time required for the right region to turn green relative to when
the left region turns green.
The assay is capable of detecting low to mid femtomolar con-
centrations of active enzymes in buffered solutions (Fig. 2) and low
picomolar concentrations in complex fluids, such as serum [5]. The
platform should be compatible with other classes of analytes as well.
Fig. 2 Calibration curve for alkaline phosphatase, which is a model enzyme that
we used to demonstrate typical results for a quantitative POC assay. The data
points are an average of three measurements and the error bars reflect the stan-
dard deviations of these averages. The inset shows an expanded view of the
bracketed region. The data for alkaline phosphatase are provided in black,
whereas results for samples containing catalase (instead of alkaline phospha-
tase) are provided in green. Similar results for β-d-galactosidase are provided in
blue. Catalase and β-d-galactosidase were used as control enzymes because
they belong to different classes of enzymes and do not react with the substrate
for alkaline phosphatase (i.e., glucose-6-phosphate) that is included in layers 4
and 5 of the device. Limits of detection are dependent on the length of the hydro-
phobic oligomer, but range from 128 fM to low nM
Quantitative Point-of-Care (POC) Assays Using Measurements of Time… 217
2 Materials
2.2 Depositing Assay 1. Hydrophobic oligomer (Fig. 1c) [6, 8] dissolved in organic
Reagents into Paper solvent (e.g., ethyl acetate or tetrahydrofuran) at fixed concen-
Microfluidic Devices trations (see Note 3). Concentrations of oligomer ranged from
0.1 to 100 mM depending on the oligomer used. These solu-
tions are made fresh prior to use (see Note 4). Oligomers that
vary in length can be selected depending on the desired sensi-
tivity for an assay: longer oligomers provide increased sensitiv-
ity while shorter oligomers allow tuning of the assay for a
desired clinical range for an analyte. The oligomers are not
currently commercially available, but they can be prepared
using the procedures described in ref. 6.
2. Capillary micropipettes (e.g., Drummond 0.25 μL disposable
micropipettes) for depositing 0.25 μL of oligomer solution
(see Note 5).
3. Synthetic food dyes (Assorted Food Colors & Egg Dye; Wal-
Mart brand) were used to give colorimetric responses and to
track the distribution of fluids within a device. The dyes were
used as 1:5 mixtures of dye to distilled water. The synthetic
food dyes contain the following components: RED 40 (diso-
dium salt of 6-hydroxy-5-[(2-methoxy-5-methyl-4-sulfophenyl)
azo]-2-naphthalenesulfonic acid.), BLUE 1 (disodium salt
218 Gregory G. Lewis and Scott T. Phillips
of ethyl [4-[p-[ethyl(m-sulfobenzyl)amino]-a-(o-sulfophenyl)
benzylidene]-2,5-cyclohexadien-1-ylidene](m-sulfobenzyl)
ammonium hydroxide plus p-sulfobenzyl and o-sulfobenzyl
salts), YELLOW 5 (trisodium salt of 4,5-dihydro-5-oxo-1-(4-
sulfophenyl)-4-[4-sulfophenylazo]-1H-pyrazole-3-carboxylic
acid), and GREEN (a 1:1 mixture of YELLOW 5 and BLUE 1).
4. Assay substrate [5, 9] (e.g., glucose 6-phosphate) in aqueous
solution at a constant concentration (20 mM) (see Note 6).
The assay substrate reacts with the target enzyme to generate
1 unit of glucose for each enzymatic reaction.
5. Buffer solutions (e.g., 40 mM HEPES buffer, pH 8.0) for
depositing assay substrates (see Note 7). The hydrophobic
oligomer responds to hydrogen peroxide more effectively at
pH 8 than in more acidic media. The buffer salts dried in the
device ensure that the sample is at this pH value.
6. Cofactor (e.g., magnesium chloride) for the target enzyme to
increase the activity of the target enzyme and improve the sen-
sitivity of the assay (see Note 8).
7. Immobilized enzymes (i.e., glucose oxidase and catalase) for
removal of glucose and hydrogen peroxide from the sample
and/or generation of hydrogen peroxide from glucose that is
generated in response to the target enzyme (see Note 9).
3 Methods
3.1 Patterning The hydrophilic paper is patterned with hydrophobic wax to form
Microfluidic Channels defined hydrophilic regions that control sample distribution within
into Paper the paper.
1. Patterns of hydrophobic regions for each layer of paper in the
3D microfluidic device are designed using CleWin Layout
Editor (see Note 10) [10], with different layers of the device
designed on different layers in CleWin so that the features in
each layer are aligned perfectly (see Note 11). The design is
saved as a post-script (.ps) file (see Note 12).
Quantitative Point-of-Care (POC) Assays Using Measurements of Time… 219
Fig. 3 Photographs showing how paper is prepared for making the devices
shown in Fig. 1. (a) Sheets of patterned paper. Individual layers are numbered
according to Fig. 1b. The individual sheets of patterned paper are outlined by the
white dotted lines. (b) Photograph depicting the method for depositing reagents
into the defined regions onto the paper. The example is green dye being depos-
ited on layer 2 using a pipette. (c) Photograph showing the procedure for deposit-
ing the hydrophobic oligomer (Fig. 1c) using a capillary micropipette
220 Gregory G. Lewis and Scott T. Phillips
Table 1 Reagents used, and their function within each layer of the device shown in Figs. 1 and 4
3.2 Depositing Assay reagents are incorporated into the layers of patterned paper
Reagents for Assays before assembling the microfluidic devices. The choice of reagents
and their purpose are summarized in Table 1. An expanded view of
each layer of the microfluidic device is depicted in Fig. 4.
1. Before the layers of the device are assembled, assay reagents
(e.g., glucose 6-phosphate, which is the substrate for alkaline
phosphatase) are deposited (3 μL) as aqueous solutions into
appropriate hydrophilic regions of paper within the device
(e.g., layers 4 and 5 in the example device shown in Fig. 1b)
(see Note 19). These solutions are allowed to dry under ambi-
ent conditions for 30 min before the device is assembled. This
layer provides selectivity for the target analyte and generates
hydrogen peroxide when the analyte is present.
2. The immobilized enzymes (e.g., glucose oxidase) are depos-
ited (3–8 μL) as aqueous solutions (in 40 mM HEPES, pH
8.0) in the appropriate hydrophilic regions of paper within the
device (e.g., layer 2 and 5 in the example device shown in
Fig. 1b) (see Note 20). These solutions are allowed to dry
under ambient conditions for 30 min before the device is
assembled. The enzymes in layer 2 remove chemical impurities
(e.g., glucose and hydrogen peroxide) from the sample prior
to the sample reaches the assay reagents.
3. The food coloring (1.0 μL) is deposited into the center of a
2.4-mm diameter hydrophilic region of patterned paper
(Fig. 3b) that will be designated as the signaling component
within the 3D microfluidic device (e.g., layer 2 in the exam-
ple device shown in Fig. 1b). The layer is allowed to dry
under ambient conditions for 30 min before it is assembled
Quantitative Point-of-Care (POC) Assays Using Measurements of Time… 221
Fig. 4 Expanded view of the device in Fig. 1, showing each layer separately. The
regions of hydrophobic wax are colored gray and the hydrophilic regions of paper
are white. The device shown is 10 mm wide × 20 mm long × 1.8 mm thick
Fig. 5 Photographs showing the steps involved in assembling the device. (a) Application of spray adhesive to
layer 5. (b) Assembling sheets of patterned paper with applied spray adhesive. Layer 3 is placed on top of layer
4 by aligning the edges of the sheets of paper. (c) The laminate sheet is taped onto a glass sheet prior to cutting
the laminate using the laser cutter. The sheet of laminate is then aligned with the top left corner of the laser
cutter. (d) The assembled sheets of devices are taped onto a glass sheet prior to cutting using the laser cutter.
(e) Photographs of the cut laminate and sheet of devices. The features of the laminate and the devices are
aligned in order to seal the top of the devices. A region of backing (~5 cm) is peeled back to allow for easy
alignment of features without the sheet of laminate sticking to the devices. After alignment, the un-backed
laminate is sealed onto the paper and the remainder of the backing is then removed. (f) The assembled device
is laminated to secure the layers together. Layer 1 of the device is already sealed with laminate, the laminate
sheet for the bottom of the devices has some of the backing removed (~5 cm) and is then aligned with the top
sheet of laminate. (g) Individual devices are then cut out of a sheet of laminated devices
224 Gregory G. Lewis and Scott T. Phillips
3.4 Conducting Performing the assay requires only that the user add the sample to
Quantitative Assays the paper-based microfluidic device and then record the time for
Using Paper-Based the right hydrophilic circle in Fig. 1a to turn green after the left
Devices hydrophilic circle in Fig. 1a turns green.
1. The sample is added to the central circular region on the top of
the device (i.e., the sample addition region in Fig. 1) (see Note 27).
A minimum sample volume of 30 μL is required for the assay
to function properly, although any sample volume above the
minimum will allow for proper quantification of the target ana-
lyte (see Note 28).
2. Once the left-hand region of the device changes color, time is
measured until the right-hand region of the device changes color.
3. The quantity of target enzyme is determined by comparing the
measured time to a calibration curve (see Note 29).
4. The calculated quantity of target enzyme is adjusted for
temperature using the correction factors described in ref. 5
(see Note 30).
4 Notes
15. When printing from the .pdf file, make sure that the page
scaling menu is set to “none” and “auto-rotate and center” is
not checked.
16. From the main menu on the printer: move the arrow to
“Paper Tray Setup” and press OK. This action brings up the
“Paper Tray Setup” menu. Move the arrow to “Tray 1 Paper”
and press OK. This action will bring up the “Tray 1 Paper” menu
that states the current setup for tray 1. Move the arrow to
“Change Setup…” and press OK. This action brings up the
“Tray 1 Paper Size” menu. Move the arrow to “New Custom
Size” and press OK. This action brings up the “Short Edge”
menu. Move the arrow to “Change…” and press OK. Move
the arrow up or down until you reach 200 mm (the units are
set to mm and cannot be changed). Press OK. This action
will bring up the “Long Edge” menu. Move the arrow to
“Change…” and press OK. Move the arrow up or down
until 200 mm is reached, and press OK; the “Tray 1 Paper
Type” menu will appear. Move the arrow to “Card Stock”
and press OK. This action will return you to the “Paper Tray
Setup” menu. Move the arrow to “Exit” and press OK. This
action returns to the main menu.
17. A piece of aluminum foil is used to cover the shelf in the oven
while heating to provide a clean surface. The printed chroma-
tography paper is placed, wax side up, on the aluminum foil.
18. The paper needs to stay in the oven for between 1 min 30 s and
1 min 45 s. If the paper is heated longer than 2 min, the wax
will bleed laterally, and the sample will not wick through the
layer. If the paper is heated for less than 1 min 30 s, then the
wax will not fully penetrate the paper and separated hydro-
philic regions of paper may become connected.
19. The concentration of the assay reagents was 20 mM. The con-
centration can be adjusted as needed to improve the sensitivity
of the assay. The immobilized enzymes are stabilized on paper
and can be stored overnight at room temperature before
assembly.
20. The immobilized glucose oxidase and immobilized catalase are
spotted individually and dried [15, 16].
21. Hydrophobic oligomers where n ≥ 5 (Fig. 1c) are not soluble
in ethyl acetate (the solvent used for oligomers n < 5); instead
they are dissolved in THF [5, 6]. The symbol n refers to the
number of repeating units in the oligomer. The concentration
of the hydrophobic oligomer solution directly affects the sen-
sitivity of the assay. The oligomers can be prepared as outlined
in refs. 6, 8.
228 Gregory G. Lewis and Scott T. Phillips
References
1. Yager P, Domingo GJ, Gerdes J (2008) Point- 9. Mohapatra H, Phillips ST (2013) Reagents and
of-care diagnostics for global health. Annu Rev assay strategies for quantifying active enzyme
Biomed Eng 10:107–144 analytes using a personal glucose meter. Chem
2. Yager P, Edwards T, Fu E, Helton K, Nelson K, Commun 49:6134–6136
Tam MR, Weigl BH (2006) Microfluidic diag- 10. Carrilho E, Martinez AW, Whitesides GM
nostic technologies for global public health. (2009) Understanding wax printing: a simple
Nature 442:412–418 micropatterning process for paper-based micro-
3. Urdea M, Penny LA, Olmsted SS, Giovanni fluidics. Anal Chem 81:7091–7095
MY, Kaspar P, Shepherd A, Wilson P, Dahl CA, 11. Noh H, Phillips ST (2010) Fluidic “timers” for
Buchsbaum S, Moeller G, Hay Burgess DC paper-based microfluidic devices. Anal Chem
(2006) Requirements for high impact diagnos- 82:8071–8078
tics in the developing world. Nature 444:73–79 12. Lewis GG, DiTucci MJ, Baker MS, Phillips ST
4. Peeling RW, Holmes KK, Mabey D, Ronald A (2012) High throughput method for prototyp-
(2006) Rapid tests for sexually transmitted ing three-dimensional, paper-based microflu-
infections (STIs): the way forward. Sex Transm idic devices. Lab Chip 12:2630–2633
Infect 82:v1–v6 13. Liu H, Xiang Y, Lu Y, Crooks RM (2012)
5. Lewis GG, Robbins JS, Phillips ST (2013) Aptamer-based origami paper analytical device
Point-of-care assay platform for quantifying for electrochemical detection of adenosine.
active enzymes to femtomolar levels using mea- Angew Chem Int Ed 51:6925–6928
surements of time as the readout. Anal Chem 14. Xiang Y, Lu Y (2011) Using personal glucose
85:10432–10439 meters and functional DNA sensors to quan-
6. Lewis GG, Robbins JS, Phillips ST (2013) Phase- tify a variety of analytical targets. Nat Chem 3:
switching depolymerizable poly(carbamate) 697–703
oligomers for signal amplification in quantita- 15. Mateo C, Palomo JM, Fernandez-Lorente G,
tive time-based assays. Macromolecules 46: Guisan JM, Fernandez-Lafuente R (2007)
5177–5183 Improvement of enzyme activity, stability and
7. Lewis GG, DiTucci MJ, Phillips ST (2012) selectivity via immobilization techniques.
Quantifying analytes in paper-based microfluidic Enzyme Microb Technol 40:1451–1463
devices without using external electronic read- 16. Khan MS, Li X, Shen W, Garnier G (2010)
ers. Angew Chem Int Ed 51:12707–12710 Thermal stability of bioactive enzymatic papers.
8. Robbins JS, Schmid KM, Phillips ST (2013) Coll Surf B 75:239–246
Effect of aromaticity on the rate of azaquinone 17. Phillips ST, Lewis GG (2013) Advances in
methide-mediated release of benzylic phenols. materials that enable quantitative point-of-care
J Org Chem 78:3159–3169 assays. MRS Bull 38:315–319
Chapter 16
Abstract
We describe here a compact smartphone-based fluorescence detector for mHealth. A key element to
achieving high sensitivity using low sensitivity phone cameras is a capillary array, which increases sensitivity
by 100×. The capillary array was combined with a white LED illumination system to enable wide spectra
fluorescent excitation in the range of 450–740 nm. The detector utilizes an orthographic projection sys-
tem to form parallel light projection images from the capillaries at a close distance via an object-space tel-
ecentric lens configuration that reduces the total lens-to-object distance while maintaining uniformity in
measurement between capillaries. To further increase the limit of detection (LOD), a computational image
processing approach was employed to decrease the level of noise. This enables an additional 5–10× decrease
in LOD. This smartphone-based detector was used to measure serial dilutions of fluorescein with a LOD
of 1 nM with image stacking and 10 nM without image stacking, similar to the LOD obtained with a com-
mercial plate reader. Moreover, the capillary array required a sample volume of less than 10 μl, which is an
order of magnitude less than the 100 μl required for the plate reader.
As fluorescence detection is widely used in sensitive biomedical assays, the approach described here
has the potential to increase mHealth clinical utility, especially for telemedicine and for resource-poor set-
tings in global health applications.
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_16, © Springer Science+Business Media New York 2015
231
232 Joshua Balsam et al.
2 Materials
2.1 Smartphone- 1. Samsung Galaxy SII cell phone (Samsung Electronics Co.)
Based Detector with a built-in lens with a focal ratio of f/2.6 and a 4 mm focal
length or similar phone.
(a) Alternative imager is a generic webcam.
2. Any simple lens with a diameter larger than the capillary array
to be imaged.
(a) Lenses with shorter focal ratios result in a more compact
device. In this instance, plano-convex lenses with diame-
ters of 45 or 20 mm were used (150 mm and 10 mm focal
lengths, respectively).
3. Green emission filter HQ535/50 M (Chroma Technology
Corp Rockingham, VT).
4. Blue excitation filter D486/20× (Chroma Technology Corp,
Rockingham, VT).
5. Illumination: LED illumination box containing red, green,
blue, and white LEDs was custom built by Luminous Film
(Shreveport, Louisiana, www.luminousfilm.com/led.htm).
(a) As an alternative an LED flashlight can be used (see Note 1).
(b) As an another alternative to LED illumination, low cost
lasers equipped with line generator can be used (from vari-
ous ebay vendors).
6. Fluorescein (Sigma-Aldrich, St. Louis, MO).
7. For flashlight configuration two 1 cm × 1 cm mirrors.
8. Capillaries, heparinized borosilicate glass capillaries
(Drummond Scientific, Broomall, PA).
(a) The length of the heparinized capillaries used is 32 mm
with outer diameter of 0.8 mm, the inner diameter of
0.65 mm and with glass thickness of around 50 μm.
3 Methods
3.1 Fabrication The detector and the assay plate were fabricated by laser machining
Methods which enable rapid fabrication and is ideal for prototyping.
1. Micromachining: The detector and the fluidics were made of
3.2 or 1 mm black acrylic, micromachined with a computer
controlled 65 W Epilog Legend CO2 laser system (see Note 3).
2. Bonding: The polycarbonate bottom was bonded to the acrylic
with double sided pressure-sensitive adhesive transfer tape (see
Note 4) which was bonded to the micromachined layer (so
there is no contact between the adhesive to the fluids).
Assembled devices were bonded together and were processed
with a hot roll laminating machine to eliminate air bubbles and
for uniform bonding.
a b
i
ii
df
iii
iv
vi
vii
c i d Smartphone
ii
iii
viii
iv
v
Flashlight
vi
vii Filter
Fig. 1 Smartphone based fluorescence detector for mHealth. (a) A schematic of the fluorescent sensor and (b) a
photograph of the device. The main components are: (i) camera phone, (ii ) emission filter, (iii ) secondary lens, (iv )
alignment fixture, (v ) capillary tube array, (vi ) two spaced excitation filters, and (vii ) multiwavelength LED light
box. In (a), df is the distance between the capillaries and the camera lens. (c) Flashlight configuration with (i )
camera phone, (ii ) emission filter, (iii ) secondary lens, (iv ) alignment fixture, (v ) capillary tube array, (vi ) excitation
filters and a diffuser (vii ) light path with two mirrors (viii ) LED flashlight, and (d) a photograph of the flashlight
based device
panel. The top of the box consists of a diffusion panel (milky white
plastic panel), which assures uniformity of the light and uniform
illumination of the sample chip [25]. The dimensions of the diffu-
sion panel are 8.5 × 5.5 cm. The LED box is powered by 12 V DC.
Alternatively LED flashlight (Fig. 1c VIII) with a diffusion
panel can be used as a low-cost illuminator (see Note 3) with two
mirrors to focus the light to the capillary array (Fig. 1 VII).
3.4 Optical System To image the three-dimensional capillary array, orthographic pro-
jection optics using a telecentric lens configuration to represent the
three-dimensional array in two dimensions was used. The main
challenge of three-dimensional imaging is uneven distribution of
the entire light source (Fig. 2a). Fluorescence measurement of a
capillary array is shown in Fig. 2b, in which the center capillaries
236 Joshua Balsam et al.
Fig. 2 The light distribution of orthographic projection optics. (a) An ImageJ 3D analysis of the planar illumina-
tion source is shown as seen with the single lens system. (b) An image of an array with 16 capillary tubes using
the single lens optics with the corresponding 3D visualization (c). (d) An ImageJ 3D analysis of imaging with
telecentric lens with the corresponding array of 16 capillary tubes (e) and the 3D visualization (f), demonstrat-
ing a significantly more uniform illumination field
Smartphone-Based Fluorescence Detector for mHealth 237
3.5 Waveguide While it is possible to use plates for assay detection, the waveguide
Capillary Array capillary array [3, 5, 24] shown schematically in Fig. 1a-V (actual
photo shown in Fig. 1b) was used for fluorescence amplification. In
this array, the excitation light-wave energy propagating through the
capillary walls can interact directly with and excite the fluorophore
molecules (via evanescent waves), which in turn emit light that can
be detected at the end of the capillary by an imaging detector. To
orient all the capillary channels towards the camera image sensor
simultaneously, two four-by-four arrays of holes in 3.2 mm thick
plates of black acrylic were fabricated which hold the capillaries in a
parallel configuration (see Note 8). The length of the capillaries
used is 32 mm with inner diameter of 0.8 mm. When loading a
capillary in the array with a sample, it is important to avoid air bub-
bles that can reduce the fluorescence signal (see Note 9). For the
fluorescence detection used in this work, light is emitted by a LED
passed through the excitation filters (Fig. 1a-V), carried through
the capillaries (Fig. 1a-V), collected by a telecentric lens (Fig. 1aIII)
which forms a parallel projection of the capillary array through the
emission filter (Fig. 1-II), and is then finally captured by the camera
(Fig. 1a-I). The resulting orthographic projection optical system
allows for a more compact device (with a phone-to-capillary dis-
tance of 32 mm as shown in Fig. 1b) which enhances portability for
mHealth. In previous designs that utilized conventional lens sys-
tems (such as the stock lens that came with the camera), a larger
distance (~150 mm) between the imaging device and the capillaries
was required in order to obtain images with uniform brightness
across the capillary array (see Note 10).
238 Joshua Balsam et al.
3.6 Image Capturing Images can be captured by mobile phone camera or by webcam.
Regardless of the device, the signals from the images can be ana-
lyzed by the following procedure:
1. Mobile phone imaging: Images were captured with the expo-
sure time for the cell phone camera set to its maximum of
1/15 of a second and the camera gain set to its maximum of
800 ISO. The images obtained are in 8-bit color JPEG format,
maximum JPEG quality was used (level 100, compression ratio
2.6:1). At this level of compression, only extremely minor arti-
facts exist. Each color image is essentially three different mono-
chrome images, each one representing a color channel (red,
green, and blue) and each pixel of each image having a value
between 0 and 255. Because the signal of interest for fluores-
cein is in the green spectrum, the green channel alone is ana-
lyzed and the red and blue channels are discarded.
To reduce background noise in the final image and improve
the signal-to-noise ratio (SNR), an average dark frame was sub-
tracted from each image to be analyzed. A dark frame is an
image with identical exposure time and gain settings as the
original image, but taken with no light reaching the camera
sensor (see Note 11 for how best to block light from reaching
the sensor). When several of these dark frames are averaged
together, the resulting image represents the average back-
ground noise generated by the camera sensor. This averaging
was done in ImageJ using many (>30) dark frames with the
mean value for each pixel calculated and combined to form the
final dark frame. The green channel was extracted from this
mean dark frame image and subtracted from the green channel
of the corresponding original image containing the signal of
interest. This process has been described in greater depth in
previous work [23, 26].
2. Webcam imaging: The incoming video stream from the
Webcam is enhanced using a software-based video processing
amplifier built into the CamApp software (available in most
webcam interface software). Exposure time and gain settings
are set to their maximum unless this setting causes over-expo-
sure (in the case of very high fluorescent signal, in which case
exposure time should be reduced to remove over-exposure).
The video processing amplifier can also apply adjustments to
the video stream to change brightness and contrast. These
should be chosen such that no pixels are recorded as having a
luminance value of zero or maximum (for an 8-bit image this
corresponds to values of 0 or 255). Because all pixels are gen-
erating a signal at the hardware level (due either to photons or
to on-chip sources of noise), if any pixels in the final recorded
image have zero signal it means that signal is being lost.
The goal of the brightness and contrast adjustment is to force
the range of the recorded pixel values to be as large as possible to
Smartphone-Based Fluorescence Detector for mHealth 239
3.7 Computational The images can be analyzed using any standard image processing
Image Enhancement software. For our system, we chose the freeware ImageJ developed
to Improve Detection at NIH (see Note 12). It enables image stacking, 2D linear image
Sensitivity analysis of the intensity of each spot in a rows or columns (Fig. 3a),
as well as 3D (Fig. 2b) spatial analysis (2D of the spatial position of
the spots and the third dimension the intensity of the spots), to
provide an enhanced visual representation of the image brightness,
which is shown in Fig. 3b. For spot analysis, the mean intensity
value of every assay spot was exported to an Excel spreadsheet and
to the scientific data analysis and graphing software Sigmaplot,
which was used to plot the data. Several analyses were conducted,
including subtracting baseline noise level and calculating the
Signal-to-Noise ratio (S/N). A computational image enhancement
of images captured from webcam video [23] was used to increase
the sensitivity of mHealth detection. The schematic of image stack-
ing for enhanced imaging is shown in Fig. 3-I. In video mode, the
webcam captures n individual frames each with underlying signal
of interest (marked with white circles) and noise. In image stack-
ing, the pixels of the n individual frames are averaged together
(stacked image), which reduces the standard deviation of this back-
ground noise, and its mean value can be subtracted from each
pixel, resulting in an enhanced signal to noise ratio.
3.8 Factors Although mobile device cameras (e.g., smartphones and web-
Contributing to System cams equipped with CMOS) each inherently have less sensitivity
Sensitivity compared with CCDs, photomultipliers, or avalanche photodi-
odes, the sensitivity of the mobile device camera was increased to
the level of a photomultiplier-based detector through the use of
combination of factors:
1. The use video mode combine with image stacking described
above increases sensitivity.
2. The quality of filters is very critical; using high quality narrow
band filters at the Emission/Excitation wavelengths will reduce
noise and improve detection.
3. While camera phone lenses are not interchangeable, with many
webcams it is possible to change the lens (we used an f/1.2
lens [23]) to maximize the amount of light transmitted to the
sensor (see Note 13).
240 Joshua Balsam et al.
II 1 2 3 4 5 6 IV 1 2 3 4 5 6
I
1 A VI
B
2 C 100
3 D
E
4 F
10
SNR
5 III V
1
n-1
n 0.1
0.001 1 1000
Stacked Concentration (nM)
image
Fig. 3 Computational image enhancement of images captured in video mode (using a webcam detector). A sche-
matic of image stacking for enhanced imaging is shown in (I). In video mode, the n individual frames each with
underlying signal of interest (marked with white circles), and interfering noise. In image stacking, each pixel is
averaged throughout the n frames, which reduces the standard deviation of this background noise and its mean
value can be subtracted from each pixel, resulting in an enhanced signal-to-noise ratio (SNR). To demonstrate
this, a 36 well plate was loaded with six concentrations of fluorescein (column 1–6), each in six replicas (rows
A–F). The signals of the wells were detected by the CMOS webcam operating in a still single frame mode (II) with
the corresponding ImageJ 3D analysis in (III). The plate analyzed in video mode and the image enhanced by image
stacking (IV) with the corresponding ImageJ 3D image in (V). The SNRs for both modes were plotted (VI). Triangles
are data points for video captured stacked images and circles are single image mode with no stacking. The LODs,
the mean plus three times standard deviation of a control (water), is marked as a dashed line. The fluorescein
concentrations used were (column #1–6): 10,000, 1,000, 100, 10, 1 nM, and control (water)
4 Notes
Fig. 4 Characteristics of cell-phone images. Two images captured with the same cell-phone camera with the
aperture blocked. Both images are identically stretched in ImageJ to reveal the faint detail they contain. (a) Using
maximum gain and maximum EV compensation, (b) using auto-gain and auto-EV setting. In (a) 70 % of pixels
have a value of 0 (no data recorded). Images such as this may be suitable for enhancement via image stacking.
In (b), 98 % of pixels have a value of zero. Images such as these are not likely to benefit from image stacking
244 Joshua Balsam et al.
References
Abstract
There is a new potential to address needs for medical diagnostics in Point-of-Care (PoC) applications using
mHealth (Mobile computing, medical sensors, and communications technologies for health care), a
mHealth based lab test will require a LOC to perform clinical analysis. In this work, we describe the design
of a simple Lab-on-a-chip (LOC) platform for mHealth medical diagnostics. The LOC utilizes a passive
capillary valve with no moving parts for fluid control using channels with very low aspect ratios cross sec-
tions (i.e., channel width ≫ height) achieved through transitions in the channel geometry via that arrest
capillary flow. Using a CO2 laser in raster engraving mode, we have designed and fabricated an eight-
channel LOC for fluorescence signal detection fabricated by engraving and combining just two polymer
layers. Each of the LOC channels is capable of mixing two reagents (e.g., enzyme and substrate) for vari-
ous assays. For mHealth detection, we used a mobile CCD detector equipped with LED multispectral
illumination in the red, green, blue, and white range. This technology enables the development of low-cost
LOC platforms for mHealth whose fabrication is compatible with standard industrial plastic fabrication
processes to enable mass production of mHealth diagnostic devices, which may broaden the use of LOCs
in PoC applications, especially in global health settings.
Key words LOC, Microfabrication, Microfluidics, Valve, Laminated object manufacturing, Laser
engraving, CCD, LED, Fluorescence
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_17, © Springer Science+Business Media New York 2015
247
248 Joshua Balsam et al.
2 Material
2.3 Data Analysis 1. Image analysis software: ImageJ, free NIH software, (http://
rsb.info.nih.gov/ij/download.htm) (see Note 1).
2. Data analysis software: Excel (Microsoft, Redmond, WA) and
Sigma plot (Sigma plot, Ashburn, VA).
3 Methods
3.1 General LOM Corel Draw computer aided drawing (CAD) software was used to
Approach design the patterns and features of the device. Before cutting, layers
of PC and PMMA were coated with 3 M 9770 adhesive transfer
double-sided tape. The cutting and engraving was done with a CO2
laser cutter as described in previous work [7–9] designed to work
with thin sheets of polymer film, such as acrylic, polycarbonate, and
polyester. The layers were assembled and bonded by successive lam-
ination with adhesive [3] to produce a 3D device [1–5].
Laser cutting: Depending on the material being cut, different
power levels and speed settings must be used to ensure a clean cut (see
Note 2). The appropriate settings are arrived at by trial and error. It
is good practice to use the lowest necessary power for cutting to
minimize burning of the material and localized heating of the work-
piece. The power settings for the vector mode used for fabrication
depend on the cutting objective. In most cases, vectors cuts are
designed to be through cuts which require higher power and slower
beam speeds. It is essential to clean debris from the cuts after laser
cutting as well to avoid blocking the channels by polymer and adhe-
sive debris. After cutting, the edge of the PC, PMMA and double-
sided tape needs to be cleaned using a fine brush and any debris in the
fluid channels should be removed using a needle or by sonication.
Laser Engraving: To reduce the number of layers, we have
sought to utilize a new variant on the laser cutting approach where
both surfaces of the PMMA can be engraved to include the func-
tional elements of the LOC. The technical challenge in this approach
is appropriate control of the Z-axis of the features, which is how
deep the features are engraved. This was achieved by empirically
determining the relationship between the power of the laser and the
depth of the engraving for a given polymer. In addition, the features
must satisfy the clearance requirements of the engraving process
(i.e., undercuts are not possible as with processes like etching).
When engraving both sides of the PMMA, both sides must
align precisely so fluids can flow from the top side wells to the bot-
tom side fluidics (see Note 3). This precise alignment was
achieved by:
1. Engraving the top side.
2. Flipping the layer and placing it in a custom frame built to hold
the machined layer in a precise position. The frame is designed
so that the PMMA layer will snap-fit into it for repeatable
Two-Layer Lab-on-a-Chip (LOC) with Passive Capillary Valves for mHealth Medical… 251
a b
1
2
3
4
5
6
7
8
Fig. 1 Functional elements of multichannel LOC system. (a) A schematic of an eight-channel microfluidic LOC plat-
form where the reagents reservoirs (1) and (2) are joined via a V-junction to the joining channel (3). The mixed
reagents are monitored in the detection wells (4) followed by a mixing trap (5). The eight-channel negative pressure
distribution manifold (6) is connected to a waste chamber (7) to the outlet, which is connected to a pump or syringe
(8). (b) A clear LOC device connected to a syringe loaded with food coloring to enhance the LOC features
3. With a ruler (or a roller), press the tape against the polymer
surface and slowly unwind the tape and press it against the
acrylic surface with the ruler to prevent air bubbles.
4. Cut the tape.
5. To create a polymer surface with adhesive on both sides, repeat
for the other side of the polymer.
Assembly: For LOM, the liner is removed from the adhesive on
each layer. Each layer is then aligned using guide holes machined
in all layers. A simple plate with two pins is used to align the layers
(Fig. 1) which are first joined together with gentle pressure.
3.2 Design Figure 1a shows the functional elements of the LOC system which
and Fabrication are similar to our previous design of a six layer microfluidic device
of Two-Layer LOC developed for botulinum neurotoxin A (BoNT-A) activity analysis
[9]. Each of the channels is designed to mix two different reagents
(e.g., enzyme and substrate) in order to generate a signal for detec-
tion. As shown in Fig. 1a, each channel is based on a V-junction
design [9] that joins the two reagents reservoirs (1, 2) passing into
a 0.5 mm joining channel (3). The mixing and incubation of
reagents is performed and monitored in detection wells (4) (see
Note 5). At the end of the channel, a trap chamber (5) prevents
diffusion among the connected channels during incubation. All
the eight channels are connected to a negative pressure distribu-
tion splitter (6), which leads to a waste chamber buffer (7).
Two-Layer Lab-on-a-Chip (LOC) with Passive Capillary Valves for mHealth Medical… 253
Fig. 2 Outline of layers and the passive valve design for the two-layer LOC platform. Outline of the layers for
the two-layer LOC platform. Layers are numbered in order from the top layer down. Both layers are 3.2 mm
PMMA. Layer I has top side (I-T) and bottom side engraving (I-B). The second layer has top side engraving only
(II). Image III is the combined device shown from the top and image IV is the side view of the LOC showing the
fluid flow between the layers. The passive capillary valve with a sharp transition in cross-sectional geometry
is used to arrest capillary flow
3.3 Flow Control One critical issue with microfludics is control of capillary flow (e.g.,
Using Passive during filling wells), which is mainly a function of channels diam-
Capillary Valves eter, length, and fluid viscosity (i.e., Hagen–Poiseuille’s Equation).
Fluid control in a LOC can be achieved by using a pump and valves
with moving parts, which complicates fabrication. In multilayer
LOCs, a 6-layer design that we have previously developed was used
[9] to create channels with high aspect ratios which can naturally
limit uncontrolled “spontaneous” flow [9].
To reduce the number of layers, we incorporated a new passive
capillary valve design, shown in Fig. 2-III with all layers superim-
posed. A side view is shown in Fig. 2-IV with the PMMA layer
engraving shown from the sides, the top (I-T), and the bottom
(I-B) and the top engraved layer II. To prevent uncontrolled flow
during filling of the reagent, there is no direct flow between sample
reservoirs and detection wells, with flow instead via channels in
layer II. In addition, the passive capillary valve shown in the circled
area in Fig. 2-IV marked with an arrow contains a sharp transition
in cross-sectional geometry to restrict capillary flow. Fluid must
transition from layer II up to a channel in I-B. This step change in
channel geometry creates a point where the fluid contact angle is
undefined and additional work (in the form of increased pressure)
is required to overcome this. The result is a passive capillary valve
which minimizes flow during filling of the reagent wells.
Figure 2-IV demonstrates the challenges of engraving both sides
of the PMMA, both sides have to be precisely aligned so fluids can
flow from the top side wells to the bottom side fluidics. This precise
alignment was achieved by first engraving the top side and then flip-
ping the layer and engraving the bottom side using a custom frame
built to hold the machined layer in a precise position. The frame was
designed so that the PMMA layer would snap-fit into it for reliable
and repeatable alignment. The frame is then aligned to built-in
guides on the laser cutter stage. Another technical challenge is the
control of the Z-axis, which is how deep the features are engraved.
As previously discussed, this was achieved by empirically varying the
power of the laser to determine the relationship to the depth of
engraving for a given polymer. By combining engraving and lamina-
tion, we have been able to reduce the number of layers required to
achieve microfluidic flow in a LOC platform from 6 to 2, substan-
tially simplifying the fabrication of a LOC while maintaining preci-
sion. The reduced number of layers also reduces the number of
interfaces, which significantly enhances the reliability of these devices
as these interfaces can potentially weaken and disrupt the flow in the
LOC eventually rendering it inoperable.
Two-Layer Lab-on-a-Chip (LOC) with Passive Capillary Valves for mHealth Medical… 255
1 2 3 4 5 6 7 8
a b c
1
2
3
4
5
6
Fig. 3 Fluorescein mixing and detection using the LOC platform. (a) The main elements of CCD based fluorescent
detector: CCD camera (1) equipped with a manual zoom 4–12 mm, f/1.2 C-mount lens (2) and a green pass band
emission filter (3). The eight-channel LOC (4) is placed above the blue band pass excitation filter (5) which is
mounted on the top of the multiwavelength LED illuminator (6). (b) The image of the two-layer LOC after mixing,
the florescent dyes in the detection wells are marked with circle and (c) the 3D ImageJ analysis of the wells
3.4 Fluorescence The use of the LOC for mHealth requires an appropriate detection
Detector technique, where optical detection is most commonly used. The
basic optical configuration of such mHealth fluorescence detector
[10–12] is shown schematically in Fig. 3a. The main elements of
this CCD based fluorescent detector are: CCD camera (1) equipped
with a manual zoom 4–12 mm to capture the image, f/1.2
C-mount lens (2) and a green pass band emission filter (3) which
block the excitation light (see Note 5). The eight-channel LOC
described above (4) is placed above the blue band pass excitation
filter (5) which is mounted on the top of the multiwavelength
LED illuminator (6) used for the excitation of the fluorophores
(see Note 6). An example of the image of the two-layer LOC with
a florescent dye in the detection well (marked with circle) is shown
in Fig. 3b and in Fig. 3c the 3D ImageJ analysis of the wells.
3.5 LOC Fluorescein The LOCs were tested for mixing and detection of fluorescein, a
Mixing and Detection medically relevant and widely used fluorescent tracer. To measure
the mixing uniformity of the eight-channel device, one well in each
channel was loaded with ~10 μl fluorescein and the other with
10 μl water (see Note 7). After applying low pressure at the outlet
port using a syringe, the fluids are drawn into the mixing wells. We
were able to control the flow with the simple passive capillary valve
(Fig. 2-IV) which prevented uncontrolled flow and filling of the
channels.
Fluorescein in the mixing wells was detected by measuring the
signal from the wells excited at 480 nm and detected at 523 nm by
256 Joshua Balsam et al.
the CCD detector (see Note 8). The schematics of the CCD detec-
tor [13] are shown in Fig. 3a, with the main elements consisting of
an SXVF-M7 CCD camera (1) equipped with a Tamron manual
zoom CCTV 4–12 mm, f1.2 C-mount lens (2) that has a green
pass band emission filter (3) mounted on the end of the lens. The
eight-channel LOC (4) is placed above the blue band pass excita-
tion filter (5) which is mounted on the top of a multiwavelength
LED illuminator (6). The image of the two-layer LOC after mix-
ing is shown in Fig. 3b and the 3D ImageJ analysis of the wells is
shown in Fig. 3c.
For the two-layer design (Fig. 3b) the average signal is 196
ADU and the standard deviation of the measurements of 25
ADU, which is ~13 %. For the three-layer device (not shown),
the average signal is 86 ADU which is lower than the signal of
the two-layer design, most likely due to replacing the thin poly-
carbonate film with the approximately order of magnitude
thicker PMMA (~1/3 of it is the engraved channels and wells).
The standard deviation of the measurements was 16 ADU,
which is ~19 % of the average. The issue of uniformity can be
attributed to the use of the CO2 laser in engraving raster mode.
When laser pulsing for engraving, heating of the plastic surface
is less uniform than during raster cutting. This nonuniformity
introduces surface roughness that can affect the uniformity of
the measurement. For cheap, mass production of the prototype,
replication processes using a metal mold master or injection
molding could improve surface quality and therefore increase
the uniformity of the measurements. Therefore, these results
suggest that the trade-off for simplicity, using two-layer fabrica-
tion, is a lower signal and lower uniformity among channels.
However, the lower signal may not be a major issue because it
can be compensated for by increasing exposure time, provided
the background noise does not increase with time.
A critical issue in microfluidic LOC platforms is cross talk (see
Note 9), which occurs when there is mixing or diffusion between
channels, especially in assays requiring long incubation times. Such
cross talk reduces measurement reliability, decreases sensitivity, and
limits the usefulness of the LOC. To overcome this problem we
use traps (Fig. 1a-5) at the end of channels to separate measuring
wells (Fig. 1a-4) from the connected channels (Fig. 1a-6). To mea-
sure the design’s effectiveness, sample and reagent reservoirs of
alternating channels were loaded with either water or Fluorescein.
The solutions were drawn to fill the detection wells and the traps,
but not the connected channels. Measurements of the fluorescent
signals suggested that there was no significant cross talk between
channels loaded with fluorescein to the channels loaded with water,
even after 2 h of incubation, suggesting that this design can elimi-
nate cross talk or diffusion between channels.
Two-Layer Lab-on-a-Chip (LOC) with Passive Capillary Valves for mHealth Medical… 257
4 Notes
References
1. Irawan R, Tjin SC, Yager P, Zhang D (2005) 8. Sapsford KE, Francis J, Sun S, Kostov Y,
Cross-talk problem on a fluorescence multi- Rasooly A (2009) Miniaturized 96-well ELISA
channel microfluidic chip system. Biomed chips for staphylococcal enterotoxin B detec-
Microdevices 7(3):205–211 tion using portable colorimetric detector. Anal
2. Schilling EA, Kamholz AE, Yager P (2002) Bioanal Chem 394(2):499–505
Cell lysis and protein extraction in a microflu- 9. Sun S, Ossandon M, Kostov Y, Rasooly A
idic device with detection by a fluorogenic (2009) Lab-on-a-chip for botulinum neuro-
enzyme assay. Anal Chem 74(8):1798–1804 toxin a (BoNT-A) activity analysis. Lab Chip
3. Munson MS, Hasenbank MS, Fu E, Yager P 9(22):3275–3281
(2004) Suppression of non-specific adsorption 10. Balsam J, Bruck HA, Rasooly A (2013)
using sheath flow. Lab Chip 4(5):438–445 Capillary array waveguide amplified fluores-
4. Rossier JS, Schwarz A, Reymond F, Ferrigno cence detector for mHealth. Sens Actuators B
R, Bianchi F, Girault HH (1999) Microchannel 186:711–717
networks for electrophoretic separations. 11. Balsam J, Bruck HA, Rasooly A (2013)
Electrophoresis 20(4–5):727–731 Orthographic projection capillary array fluo-
5. Rossier J, Reymond F, Michel PE (2002) rescent sensor for mHealth. Methods 63:276
Polymer microfluidic chips for electrochemical 12. Balsam J, Rasooly R, Bruck HA, Rasooly A
and biochemical analyses. Electrophoresis (2013) Thousand-fold fluorescent signal
23(6):858–867 amplification for mHealth diagnostics. Biosens
6. Pompano RR, Platt CE, Karymov MA, Ismagilov Bioelectron 51C:1–7
RF (2012) Control of initiation, rate, and rout- 13. Sun S, Francis J, Sapsford KE, Kostov Y,
ing of spontaneous capillary-driven flow of liquid Rasooly A (2010) Multi-wavelength Spatial
droplets through microfluidic channels on slip- LED illumination based detector for in vitro
chip. Langmuir 28(3):1931–1941 detection of Botulinum Neurotoxin A Activity.
7. Sapsford KE, Sun S, Francis J, Sharma S, Sens Actuators B 146(1–8):297–306
Kostov Y, Rasooly A (2008) A fluorescence 14. Hawkins KR, Yager P (2003) Nonlinear
detection platform using spatial electrolumi- decrease of background fluorescence in poly-
nescent excitation for measuring botulinum mer thin-films - a survey of materials and how
neurotoxin A activity. Biosens Bioelectron they can complicate fluorescence detection in
24(4):618–625 microTAS. Lab Chip 3(4):248–252
Chapter 18
Abstract
Many modern spectrometric instruments use diode arrays, charge-coupled arrays, or CMOS cameras for
detection and measurement. As portable or point-of-use instruments are desirable, one would expect that
instruments using the cameras in cellular telephones and tablet computers would be the basis of numerous
instruments. However, no mass market for such devices has yet developed. The difficulties in using mega-
pixel CMOS cameras for scientific measurements are discussed, and promising avenues for instrument
development reviewed. Inexpensive alternatives to use of the built-in camera are also mentioned, as the
long-term question is whether it is better to overcome the constraints of CMOS cameras or to bypass them.
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_18, © Springer Science+Business Media New York 2015
259
260 Alexander Scheeline
2 Materials
Fig. 1 Spectra of typical LED light sources. Three narrowband photodiodes, emitting blue, green, and red, can
be set to various relative intensities which the human eye perceives as any of a continuum of colors. Chemical
species respond only to the actual emitted wavelengths; there is almost no light from narrowband LEDs
between 580 and 610 nm. A fourth “True white” LED has no gaps in the spectrum from 430 to 730 nm.
Redrawn from images by superbrightleds.com
CMOS/Cell Phone Spectrometry 261
255 255
Byte Value Image 2
0 0
0 255 0 255
Byte Value Image 1 Byte Value Image 1
Fig. 2 Effect of gain setting on blue channel response of MightEx 3 Mpix CMOS camera. (a) Gain set to 1×. (b)
Gain set to 2×. Note gain saturation at less than full scale and noisy value for saturation in (a), while signal fills
entire scale in (b)
CMOS/Cell Phone Spectrometry 263
3 Methods
Fig. 3 An open-air spectrometer made of inexpensive components (CR2032 battery, RL-W5020 LED, 500 line
per mm transmission grating, Motorola Droid RAZR Max running Android 4.1.2, 6 Mpix camera, normal expo-
sure, printed baseplate designed for classroom use.). Inset (a) shows the layout with component labels and
overlays of the optical path. Dispersion and saturation depend on diffraction order and exposure. Inset (b) shows
an overexposed image when the grating/camera distance is 0.12 m and the grating normal/camera observation
angle is approximately 30°. While no sample cuvette is present in Inset (a), the position in which it might be
placed is indicated. Inset (a) has been heavily retouched to remove distractions. Note that room light, allowing
the setup to be photographed, is an example of stray light if one is trying to perform spectroscopy
3.2 Spectrometry Whitesides, at Harvard, has been pursuing the use of cell phones
with Cellular for carrying out telemedicine [12, 13]. As an early adopter of this
Telephones technology, the group faced programming challenges and a rapidly
evolving platform; while current cellcams feature multiple mega-
pixels, the cameras available at the time of the cited work were 1
megapixel or smaller. Further, lighting conditions are difficult to
control, so interpretation of colors from raw images can be mis-
leading. The group advocated transmission of pictures to a central
base or authority for interpretation using tools such as PhotoShop®
to normalize response and extract chemically, environmentally, or
medically important information. With the explosive growth of
phone-born computing power, it is no longer obvious that central
processing is required. The extent to which systems can be hard-
ened against user limitations without real-time oversight by experts
is also unknowable as yet.
An early example of a spectrometer, developed by Mitch Nelson,
using a consumer-grade CCD camera is still accessible on the web
[14]. It employs a 35 mm diagonal holographic transmission grat-
ing, an approach that has come into favor due to the astonishingly
low cost of such dispersion. Also in common with other cellcam and
webcam-based systems, intensity calibration is not addressed.
At about the same time as Nelson’s development, a sleeker
device of similar layout was published by Field [15]. Billed as “high
resolution,” it is not clear how such a term should be defined. In
conversation with the author, Field noted that he did wavelength
calibration by aligning components so that undispersed light (zero
order) is centered in the instrument focal plane. Plus and minus
first order for specific wavelengths show up symmetrically on oppo-
site sides of zero order. Because fluorescent lamps emit strongly at
the mercury 546.1 nm green atomic emission line, pointing the
input slit of the instrument at a fluorescent light generates the req-
uisite light. Silence in the writeup about response linearity and off-
set leads one to believe that intensity measurements were accepted
at face value, without considering dark current or relative response
as a function of wavelength. The quantum efficiency of a detector
(number of measureable charges generated per incident photon) is
always wavelength-dependent. Thus, absent a traceable standard,
only relative measurements between similar sources is possible.
266 Alexander Scheeline
of the dispersed LED light in Fig. 3b. This highlights the need to
embed software control in an “app” if serious work is to be done
on cell phones; the default exposure tends to saturation at least
somewhere in the image. Because of this unregulated default per-
formance, further detail is avoided here. No medical use should be
made of an instrument of this design. Nevertheless, one of the first
students to employ this “toy instrument” combined the gratings in
such a way that a novel stacked diffraction grating arrangement,
potentially useful for absorption, fluorescence, and reflectance
spectrometry, was devised. A patent for this grating arrangement is
now pending [23].
Other efforts to get inexpensive spectrometry into classrooms
has exploited the wide availability of cellcams and webcams.
A lightbox or computer screen can provide reasonably uniform back-
lighting for an array of cuvettes so that an entire working curve can
be obtained in a single photographic frame [24]. Spectral resolution
can either be from the RGB image plane resolution of the camera or
from placing a bandpass filter in front of the camera. The authors
used PhotoShop® to extract intensity data, and kept absorbance below
1 to ensure adequate digitization resolution. Published data showed
little evidence of stray light(see Note 5). In a related approach, an
array of LEDs backlit a multiwell plate. The RGB response of a web-
cam measured absorbance in each of the wells [25].
Fig. 4 Two cell cams look at a hot stove. (a) Samsung Omnia Windows CE phone; camera has no near infrared
filter. Note purple tone to heating element. (b) Motorola Droid phone with near infrared filter. Color of an orange
plastic jar top included for comparison; the top in (b) as well as the burner color there is approximately the
same as the author’s perception. Not only does this illustrate the need for proper filtering to avoid interference
from the near-infrared, but also the difficulty of accurately measuring color from camera images
Fig. 5 Images from two figures in ref. [36]. (a) Layout of fiber optics and webcam detector. (b) Zero order (right
side) and first order (left side) for (a) 465, (b) 591, and (c) 639 nm for light through the five fiber optics. Note
comatic blur, field curvature, and offset. When using a minimum of additional optics with consumer cameras,
software must compensate for non-ideal imaging. Images copyright by the Society for Applied Spectroscopy.
Used with permission
4 Notes
5. Stray light is any light reaching the detector that has not
followed a path that corresponds to the physics and chemistry
expected for an experiment. Room light, light from junctions
on integrated circuits acting as weak LEDs, and light scattered
from dust that sprays around the interior of an instrument are
examples of stray light.
6. Webcam and cellcam detection can be extended to the X-ray
region using a 5 μm aluminum foil transducer to convert X-rays to
avalanches of “dark counts” in nearby pixels [56]. The CCD or
CMOS transducer must be adjacent to the foil, so the foil must
be placed inside the camera housing. Backscatter from nearby elec-
tronics or scattering from housing components may blur images.
7. What resolution is feasible with a cellcam? Lens focal length is
4–8 mm. Readily available gratings have 300–3,600 lines per
mm, so an example at 1,200 lines per mm is useful. First, if the
light directed at the grating is collimated, using the relation-
ship in Note 3 results in dispersion of 0.06 to 0.13 nm/pixel
for 1.5 μm pixels, ignoring aberrations (imperfect imaging)
and the blur from the (de)magnified image of the entrance
aperture. Realistically, with a 100 μm diameter fiber optic as
light source and a 100 mm focal length collimation lens or
mirror, the source image diameter would be between 8/100
and 4/100 × 100 μm in diameter, or 3–5 pixels, corresponding
to 0.2–0.8 nm resolution. This is consistent with the author’s
measurements using a 50 μm pinhole entrance aperture,
200 groove/mm grating, 25 mm focal length collimating
optic, and 8 to 12 mm focal length zoom lens on the MightEx
camera mentioned elsewhere in this chapter.
Acknowledgments
References
1. Rieke G (2003) Detection of light: from the log/displayproduct.cfm?productID=1490 ,
ultraviolet to the submillimeter, 2 edn. (downloaded 12/27/2009)
Cambridge University Press, Cambridge 4. Rainbow Symphony Inc (2009) Rainbow
2. Su J-C, Song S-F, Chen H-S (2011) Symphony Store Holographic Gratings. http://
Chromaticity stability of phosphor-converted www.rainbowsymphonystore.com/scienanded1.
white light-emitting diodes with an optical fil- html, (downloaded 12/27/2009)
ter. Appl Opt 50:177–182 5. Coates J (2010) New miniaturized spectral
3. Edmund Scientific diffraction grating page, measurement platforms covering the visible to
http://www.edmundoptics.com/onlinecata- the mid-IR, http://depts.washington.edu/
274 Alexander Scheeline
metric detection of biomarkers in sweat and 44. Ozcan A (2013) Ozcan Research Group
saliva. Lab Chip 13:3232–3238 Refereed Journal Publications, http://inno-
33. Hoving W (2013) Environmental monitoring/ vate.ee.ucla.edu/refereed-journal-
spectroscopy: crowd-sourced measurements publications.html, (downloaded 10/09/2013)
offer a unique view of pollution’s effects. 45. Titlow JP (2011) Why user-customized prod-
BioOptics World, vol 6. http://www.biooptic- ucts are the future of business (for real this
sworld.com/ar ticles/print/volume-6/ time), http://readwrite.com/2011/04/15/
issue-5/features/environmental-monitoring- user-customized-products-future-of-business#
spectroscopy-crowd-sourced-measurements- awesm=~okg94zZ2kkPR4l, (downloaded
offer-a-unique-view-of-pollution-s-effects.html 10/14/2013)
34. University L (2013) Measuring particulates 46. Xiang Y, Lu Y (2011) Using personal glucose
with your smartphone, http://www.research. meters and functional DNA sensors to quantify a
leiden.edu/news/measuring-particulates- variety of analytical targets. Nat Chem 3:697–703
with-your-smartphone.html, (downloaded 47. Lillehoj PB, Huang M-C, Truong N, Ho C-M
10/10/2013) (2013) Rapid electrochemical detection on a
35. White JD, Scholten RE (2012) Compact dif- mobile phone. Lab Chip 13:2950–2955
fraction grating laser wavemeter with sub- 48. Ingle JD, Crouch SR (1972) Evaluation of
picometer accuracy and picowatt sensitivity precision of quantitative molecular absorption
using a webcam imaging sensor. Rev Sci spectrometric measurements. Anal Chem
Instrum 83:113104 44:1375–1386
36. Subriddetchkajorn S, Intaravanne Y (2012) 49. Rothman LD, Crouch SR, Ingle JD (1975)
Home-made N-channel fiber-optic spectrome- Theoretical and experimental investigation of
ter from a web camera. Appl Spectrosc factors affecting precision in molecular absorp-
66:1156–1162 tion spectrophotometry. Anal Chem
37. Lima MB, Andarde SE, Barreto IS, Almeida 47:1226–1233
LF, Araujo MCU (2013) A digital image-based 50. Ingle JD, Crouch SR (1988) Spectrochemical
micro flow batch analyzer. Microchem J analysis. Benjamin, New York
106:238–243 51. Skoog DA, Holler FJ, Crouch SR (2007)
38. Balsam J, Bruck HA, Kostov Y, Rasooly A Principles of instrumental analysis. Thomson
(2012) Image stacking approach to increase Higher Education, Belmont, CA
sensitivity of fluorescence detection using a low 52. Inc. HS (2013) VS7000-PDA Miniature PDA
cost complementary metal-oxide- Spectrometer, http://www.horiba.com/fil-
semiconductor (CMOS) webcam. Sens Actuat eadmin/uploads/Scientific/Documents/
B 171–172:141–147 OEM/VS-7000-PDA-2013.pdf, (downloaded
39. Horlick G (1975) Reduction of quantization 12/24/2013)
effects by time averaging with random added 53. Hsu W-H, Sainz MA, Coleman DM (1989) An
noise. Anal Chem 47:352–354 aberration-corrected time- and spatially-
40. Belchamber RM, Horlick G (1981) Use of resolved spectrometer for studies of transient
added random noise to improve bit-resolution discharges. Spectrochim Acta 44B:109–121
in digital signal averaging. Talanta 28:547–548 54. Miller DL, Scheeline A (1993) A computer
41. Tseng D, Mudanyali O, Oztoprak C, Isikman program for the collection, reduction, and
SO, Sencan I, Yaglidere O et al (2010) Lensfree analysis of echelle spectra. Spectrochim Acta
microscopy on a cell-phone. Lab Chip 48B:E1053–E1062
10:1787–1792 55. Sadler DA, Littlejohn D, Perkins CV (1995)
42. Zhu H, Yaglidere O, Su T, Tseng D, Ozcan A An automatic wavelength calibration proce-
(2010) Cost-effective and compact wide-field dure for use with an optical spectrometer and
fluorescent imaging on a cell-phone. Lab Chip array detector. J Anal Atom Spec
11:315–322 10:253–257
43. Mudanyali O, Dimitrov S, Sikora U, 56. Lane DW (2012) X-ray imaging and spectros-
Padmanabhan S, Navruz I, Ozcan A (2012) copy using low cost COTS CMOS sensors.
Integrated rapid-diagnostic-test reader plat- Nucl Instrum Methods Phys Res B Beam
form on a cellphone. Lab Chip 12:2678–2686 Interact Mater Atoms 284:29–32
Chapter 19
Abstract
The development of simple, inexpensive paper-based sensors for medical diagnostics and other applications
is now an important emerging area in the field of biosensors; however, the electronic instrument or reader
used to interrogate such sensors adds significantly to the cost of the analysis. In this chapter we describe the
design and construction of novel, low-cost disposable electrochemiluminescent (ECL) sensors based on
screen printed carbon electrodes and paper-based microfluidics. Moreover, a method to interrogate these
sensors using only a mobile phone is articulated. This is realized by exploiting the audio output of the device
to achieve electrochemical control, while using the camera to detect the resulting light emitted during the
ECL reaction. The combination of cell phone technology with low-cost paper microfluidic sensors dramati-
cally reduces the cost of sensing and has the potential to enhance health-care outcomes by exploiting the
functionality, connectivity, and close to worldwide penetration of mobile phone technology.
Key words Paper microfluidics, Low-cost sensing, Mobile phone-based sensing, Electro-
chemiluminescence
1 Introduction
The growth in mobile phone usage since the late 1990s has been phe-
nomenal. Moreover, their capability has grown dramatically in that
time. Even more remarkable is the projection that virtually all of the
inhabitants of this planet (including those in the developing world) will
have access to such a device within the next 5–10 years [1]. We believe
that paper-based microfluidic sensors combined with the power and
ubiquity of mobile phones can be harnessed to facilitate a revolutionary
approach to medical diagnostics in the developing world.
Microfluidic paper-based analytical devices (or μ-PADs) have
the significant advantage of not requiring any external means of
fluid transport which occurs via capillary action. Also, they require
only small sample volumes, the paper may filter or otherwise
separate the components of the sample, they are easy to store and
transport and are readily disposed of safely via incineration. While
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_19, © Springer Science+Business Media New York 2015
277
278 Jacqui L. Delaney and Conor F. Hogan
2 Materials
2.1 Chemicals 1. All solutions were made up using deionized water with a resis-
and Materials tivity of at least 18 MΩ/cm.
2. 0.1 M phosphate buffer solutions (pH 7.5 unless otherwise
stated), were prepared using sodium phosphate dibasic
(≥99 %, Sigma-Aldrich) and sodium phosphate monobasic
(≥99 %, Sigma-Aldrich).
Mobile Phone Based Electrochemiluminescence Detection… 279
2.2 Instruments 1. The pH of the phosphate buffers was tested using a MEP
Instruments, Metrohm 827 pH Lab Meter and an MEP
Instruments’ Metrohm 6.0228.010 electrode.
2. Cyclic voltammetry was performed using a CH Instruments
(Texas) potentiostat (660B or 620C).
3. Acquisition and analysis of voltage and ECL intensity signals
was performed using an eDAQ e-corder 401 model ED401.
4. Low-cost screen printed electrodes (SPEs) were manufactured by
Zensor R&D (Taiwan) (Zensor TE100 screen printed voltam-
metric electrodes, Cat no. ET077). SPEs contain a 3 mm diam-
eter working electrode and an arc shaped auxiliary electrode
(both made of graphite carbon powder) and a Ag/AgCl pellet
reference electrode all on a 50 × 13 mm plastic substrate (Fig. 2c).
5. ECL optimization experiments were performed using an Eco
Chemie, μ-Autolab type II potentiostat and an Electron Tubes
Ltd. (model 98285B) photomultiplier tube (PMT) in a
custom-built light-tight Faraday cage. A high voltage power
supply was used biased at 500 V (unless otherwise stated).
280 Jacqui L. Delaney and Conor F. Hogan
3 Methods
3.1 Fabrication We will describe the two main approaches to the fabrication of the
of Paper Microfluidic paper microfluidic substrates used in this work. The first is the
Sensors method of Li et al. [10] where the fluidic channel is defined by a
hydrophobic barrier printed into the paper using inkjet printing.
The second referred to here as the “cut and seal” method, is where
the paper fluidic element is cut or punched to the desired shape
Mobile Phone Based Electrochemiluminescence Detection… 281
Fig. 1 (a) is a CAD drawing of the sensor holder used to interface the sensor with the mobile phone camera
and exclude ambient light. The photographs in (b) without the phone and (c) with the phone show how the
holder is used. From ref. 9
Fig. 2 The fabrication and operation of the paper-based microfluidic ECL sensor. Paper microfluidics were
produced in bulk using a conventional inkjet printer (a). Once the fluidics were loaded and dried they were
individually cut to size (b). The paper microfluidics were then aligned and fixed onto the face of the SPE via
lamination (c). A drop of analyte was introduced via a small incision at the bottom of the channel. Once the
paper was fully wetted, the sensor was placed in front of the camera of the mobile phone. A potential was
applied and the resulting ECL emission was captured on the mobile phone (d). (From ref. 8)
3.1.2 Fabrication 1. The paper microfluidics were cut to shape using a simple hole
of “Cut-and-Seal” Paper punch with a 6 mm diameter from Whatman filter paper (No.
Microfluidic Sensors 4). In this case the laminate itself defined the perimeter of the
fluidic zone rather than a hydrophobic chemical agent.
2. The 6 mm empty discs were loaded with 7 μL of Ru(bpy)32+
and left to air dry for approximately 60 min.
3. Once dry, the discs were placed in a warm oven (≈50 °C) for
approximately 15 min to ensure all moisture was removed.
4. The dry Ru(bpy)32+ loaded discs were placed in GBC A5 Peel
n Stick Pouches which have an adhesive back.
Mobile Phone Based Electrochemiluminescence Detection… 283
1 2 3 4
Fig. 3 Steps involved in fabrication of the paper microfluidic sensors. (1) The paper fluidic element (6 mm
Ru(bpy)32+ loaded disc) is laminated into an adhesive backed laminating pouch with a dummy disc under the
paper fluidic. (2) The adhesive back of the laminating pouch is exposed. (3) The dummy disc is removed to
expose the Ru(bpy)32+ loaded disc. (4) The laminated paper microfluidic element is adhered to the screen
printed electrode with the detection zone aligned with the working electrode [9]
3.2 Detection The sensors described above may be interrogated in two ways:
and Analyses Firstly, a potentiostat or other external voltage source may be used
to initiate ECL emission which is then detected and quantified
using the camera of a mobile phone to photograph the lumines-
cence. Secondly, the mobile phone itself can be used to provide the
electrochemical control while the ECL is simultaneously moni-
tored using the camera in video mode.
3.2.1 Mobile Phone 1. To ensure even, consistent pressure was applied to all sensors a
as Detector with External perspex “clamp” was made. Perspex was chosen because it is a
Potentiostatic Control sturdy, transparent plastic material. The clamp consisted of two
pieces of perspex (10 mm thick) held together by four screws
(one in each corner). The sensor was placed in between the
pieces of perspex and the screws were tightened (Fig. 4).
284 Jacqui L. Delaney and Conor F. Hogan
Fig. 4 An example of how a perspex clamp was used. Pressure is applied evenly
over the sensor
2. Once the sensor was in the clamp a small incision was made at
the bottom of the paper microfluidic channel where the ana-
lyte solution would be introduced into the sensor.
3. A drop of the sample was applied to the incision whereupon it
was transported to the detection zone by capillary action
(<10 s).
4. Once the detection zone was fully wetted, the sensor, still in
the perspex clamp was connected to the potentiostat and
aligned under the PMT (for optimization studies) or in front
of the mobile phone camera (for proof-of-concept detection).
Example cyclic voltammograms and ECL profiles produced
using the sensors for optimization work, are shown in Fig. 5.
5. Using DBAE as a model analyte, an LOD of 0.9 μM was
obtained using a conventional light detector (PMT), while the
LOD for NADH was determined to be 72 μM.
6. For mobile phone detection the camera was positioned 10 cm
away from the sensor in a darkened room.
7. The settings on the mobile phone were changed to “fire-
works” and a timer was started for 10 s. When the timer hit
the 3 s mark the potentiostat was started to initiate the ECL
reaction.
8. The potential step used by the potentiostat was 0 to 1.25 V for
2 s. The bright red emission was then captured in a photo-
graph from the mobile phone. (See Fig. 6b).
Mobile Phone Based Electrochemiluminescence Detection… 285
Fig. 5 Typical sensor responses for (a) 5 mM DBAE and (b) 5 mM NADH. The
voltametric (current) responses are dashed blue, and the ECL emission profiles
are in red. Both experiments were run at a scan rate of 0.2 V/s and were versus
a Ag/AgCl reference electrode. (From ref. 8)
Fig. 6 (a) Calibration curve between 0.5 and 20 mM for DBAE using paper microfluidic ECL sensor with mobile
camera phone as the detector. The magnitude of the ECL signal is proportional to the intensity of the red pixels
in the digital image. (b) Digital photographic images of ECL emission from the paper fluidic sensor obtained for
various concentrations of DBAE using camera phone [8]
3.2.2 Both Potentiostatic 1. In order to test the voltage obtainable from the mobile phone
Control and Detection (in this case a Samsung Galaxy S (i9000)), a lead connected to
Performed by Mobile a stereo 3.5 mm audio jack on the one end and two alligator
Phone clips (that were once ear phones) on the other was plugged in
Mobile Phone Based Electrochemiluminescence Detection… 287
Fig. 8 Arrangement used for mobile phone based ECL sensing. The audio jack
supplies the potential to the paper microfluidic sensor, while the resultant emis-
sion is detected by the camera. Both the excitation and detection processes are
controlled by a software application. The black plastic holder positions the sen-
sor adjacent to the camera and blocks ambient light. (From ref. 9)
References
Abstract
iStethoscope Pro is the first piece of software (an “App”) produced for iOS devices, which enabled users
to exploit their smartphones, music players, or tablets as stethoscopes. The software exploits the built-in
microphone (and supports externally added microphones) and performs real-time amplification and filter-
ing to enable heart sounds to be heard with high fidelity. The software also enables the heart sounds to be
recorded, analyzed using a spectrogram, and to be transmitted to others via e-mail. This chapter describes
the motivation, functionality, and results from this work.
Key words Stethoscope, Digital auscultation, Audio processing, iPhone app, iStethoscope Pro
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_20, © Springer Science+Business Media New York 2015
293
294 Peter J. Bentley
2 Materials
3 Methods
3.1 Launching The iStethoscope Pro app should first be located on the App Store,
the App downloaded to an appropriate device, and launched. On first
launching the user will be prompted to activate the app by going to
296 Peter J. Bentley
Settings and agreeing to the terms of use. These terms make the
user aware that the device may interfere with implanted medical
equipment such as pacemakers and defibrillators and that it should
not be used near people with such implanted equipment. Once acti-
vated, the app can be launched and will run normally from then on.
On launching, the app shows an initial “splash page” which
contains a link to the help system. After 3 seconds the main screen
is shown, see Fig. 1. The user may return to the splash page by
pressing “i” at the bottom of the main page at any time.
Fig. 1 Example screenshots for iStethoscope Pro. (a) The main screen showing the mode button with text indicating
the current audio mode, the stethoscope image, the gain control, and the “i” information button. (b) The playback
screen showing a spectrogram of the sound. (c) Some of the user controllable Settings for the program
iStethoscope: A Demonstration of the Use of Mobile Devices for Auscultation 297
yes
yes
no
yes
convert audio queue into AIF format file; convert spectrogram into
bitmap image; create blank email with both files as attachments;
enable user to modify, send or cancel email
When the user is content with the audio quality they can quickly
remove the device from the body and shake it sharply, or touch the
stethoscope image. This initiates playback mode.
3.3 Audio Playback In playback mode a spectrogram of the audio is shown and the last
7 or 8 s is played continuously. The spectrogram plots the audio
using a Fast Fourier Transform [8], see Fig. 4. The x-axis is time,
the y-axis is frequency, brighter colors indicate volume. Although
the S1 and S2 components are visually easy to pick out because of
the higher frequencies, most of the key information is found in the
lower frequencies, at the bottom of the image.
When in this mode the user may press the mail symbol. This
triggers the creation of an audio file containing the audio being
played back, and a bitmap image of the spectrogram. The two files
are attached to a blank e-mail and the user is then able to send the
data to another computer (or save the data in their own e-mail out-
box for later). Figure 5 illustrates an example of normal and abnor-
mal heart sounds as plotted by Audacity on a desktop computer.
3.4 Crowd In the Settings for the app, the user may choose to activate a “med-
Sourcing Data ical trial” setting. This overrides all other settings, and forces spe-
cific filtering, amplification and AVC settings that were chosen
through experimentation by Nordehn’s group [4]. After ausculta-
tion, if the user chooses to send an e-mail containing their heart
sounds, the resulting mail is automatically addressed to a computer
at the University of Minnesota Duluth, which anonymizes the data
and stores the audio in an archive for later processing.
iStethoscope: A Demonstration of the Use of Mobile Devices for Auscultation 301
Fig. 5 Data analysis spectrogram plots by Audacity software (sounds have been gathered using a low-pass
filter at 400 Hz). Left: normal heart sound. Right: Aortic regurgitation. The difference in the sounds is immedi-
ately clear to a clinical expert accustomed to reading such spectrogram displays
4 Notes
4.2 Assessment The main aim of this work was to demonstrate the use of mobile
of Success phones for auscultation and by doing so, increase awareness of
healthy hearts worldwide. To measure success it is necessary to
examine the uptake and use of the app. So far the reception of the
app has been remarkable, receiving substantial attention world-
wide. Two UK broadsheet articles (Independent and Guardian
[10, 11]) triggered significant media attention at the beginning of
September 2010. The author was then interviewed for over 20
radio shows, and many television programs and news channels
worldwide. The story was picked up by most national papers in the
UK and worldwide, in addition to magazines, leaflets and online
media. In 1 week the iPhone app had been downloaded a further
200,000 times. Out of the 250,000 programs available for these
devices, this became the number one bestselling app in the App
Store. It is hard to estimate the number of people worldwide that
have been made aware of this work, and through it, highlighted
the importance of healthy hearts, but given the extensive world-
wide television, radio, and print coverage, the number is likely to
be several hundred million.
iStethoscope: A Demonstration of the Use of Mobile Devices for Auscultation 303
The story has been highly positive for all those with an interest
in technology and health. Feedback includes comments from doc-
tors who say the use of the program in training will save lives.
Others suggest that now heart sounds can be captured from
patients in remote regions of the world and e-mailed to the experts
in the cities. The publicity has also led to many new possibilities for
further funding, trials of the technology remote areas of the world
including India, China, and Peru, and licensing of the technology
by medical companies. One user described how the app helped to
save the life of her unborn child as it suffered complications.
The iPhone application has been a huge success, with two to
three million downloads from over 100 countries, and a resulting
large database of useful heart sounds. The response from the medi-
cal profession and public has been overwhelmingly positive; indeed
an article has been published independently by cardiologists in
USA, detailing the use of the app for heart sound collection [4].
References
1. WHO (2011) Global status report on non- PASCAL heart challenge workshop, AISTATS
communicable diseases 2010. World Health 2012, La Palma, 25 Mar 2012
Organization, Geneva 7. Gomes EF, Bentley PJ, Coimbra M, Pereira E,
2. WHO (2011) Global atlas on cardiovascular Deng Y (2013) Classifying heart sounds:
disease prevention and control. World Health approaches and results for the PASCAL
Organization, Geneva Challenge. In Proc. 6th International
3. Mathers CD, Loncar D (2006) Projections of Conference on Health Informatics, HealthInf
global mortality and burden of disease from 2013, Barcelona, Spain, Feb 2013
2002 to 2030. PLoS Med 3(11):e442 8. Bracewell RN (1986) The Fourier transform
4. Palm D, Burns S, Pasupathy T et al (2010) and its applications, vol 31999. McGraw-Hill,
Artificial neural network analysis of heart New York
sounds captured from an acoustic stethoscope 9. Bentley P, Nordehn G, Coimbra M, Mannor S
and emailed using iStethoscopePro. J Med (2011) The PASCAL classifying heart sounds
Device 4(2):027531–027531-1. challenge. http://www.peterjbentley.com/
doi:10.1115/1.3443737 heartchallenge/index.html
5. Audacity. Available under the terms of the 10. Laurance J (2009) 59p stethoscope on iphone
GNU General Public License (GPL) as pub- proves a hit. Article in Independent Newspaper,
lished by the Free Software Foundation: 14 Dec 2009. The Independent (London)
http://audacity.sourceforge.net/ 11. Hill A (2010) iPhone set to replace the stetho-
6. Deng Y, Bentley PJ (2012) A robust heart scope. The Guardian (London), Monday 30
sound segmentation and classification algo- Aug 2010.
rithm using wavelet decomposition and 12. Bentley P. iStethoscope Prop Help. http://
spectrogram. Extended abstract in the first www.peterjbentley.com/istethoscopepro.html
Chapter 21
Abstract
iPhysioMeter is a new smartphone application (“App”) for the Apple iPhone and iPod touch that allows
photoplethysmography (PPG) to be implemented without the need for any additional devices. The resulting
signal, the photoplethysmogram, allows the calculation of basic but valuable and frequently used physiological
indices such as heart rate (HR) and pulse volume (PV). The design of iPhysioMeter has very much been
influenced by a consideration of usability, as is immediately evident from ones first experience with it. However,
its apparent simplicity in use should not disguise the need for correct operation, which otherwise might lead
to collection of invalid or inaccurate data. There are several unexpected pitfalls that might not only produce
inaccurate values, but, under some circumstances, could also damage the device or present a hazard to the user
or subject. We therefore describe here, firstly, the core technology that makes it possible to perform PPG and
to calculate HR and normalized PV (NPV) from the photoplethysmogram using only a smartphone, secondly,
the correct and optimum methods and procedures for using iPhysioMeter that will help to ensure safety and
the derivation of valid data under real operational conditions. We hope that these descriptions will help facili-
tate any activities related to physiological measurement when using only a smartphone.
Key words Smartphone, Physiological indices, Photoplethysmography, Heart rate, Pulse volume,
iPhysioMeter, CMOS Camera
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_21, © Springer Science+Business Media New York 2015
305
306 Kenta Matsumura et al.
Fig. 1 (a) An illustration of photoplethysmography performed using only a smartphone. (b) Schematic drawing
illustrating a section of the measuring site. The white light emitted by the LED flash propagates into the finger,
being scattered and absorbed by blood and other hard and soft tissues, and finally reaches the camera. The
light intensity captured by the camera varies in accordance with arterial blood volume pulsations; that is, pulse
wave. The longer wavelength has deeper penetration into the tissue, thereby probing deeper regions. (c) An
example of an actual image captured by the CMOS camera from the finger. The light intensity change involved
in arterial pulsations could be measured using any of the three light colors, that is red (R), green (G), or blue
(B), available from the CMOS camera. The color discrimination is achieved by the camera
use smartphone PPG measurement tool (Fig. 2). The main features
of iPhysioMeter are: (1) It shows good agreement with results of
HR and NPV from dedicated laboratory devices [21, 22]; (2) It is
available worldwide for free at iTunes App Store by Apple, Inc.; (3)
It allows green light measurement, which has consistently shown
its superiority over red and blue PPG in terms of the relative free-
dom from motion artifacts and high signal/noise ratio [22–25];
(4) It is designed to measure not only the time-related of measure
of HR (to be precise, pulse rate) but also the amplitude related
index NPV (to be precise, cutaneous PV) (see Note 1); (5) It is
equipped with an on-line beat-by-beat HR and NPV auto analysis
function with automatic outlier detection; (6) It is provided with a
data management function that enables users to display, name, and
delete a set of beat-by-beat data, and to send these data in a comma-
separated value (CSV) format via e-mail; (7) It is designed to mea-
sure three-axis g-forces, which is useful for judging whether there
308 Kenta Matsumura et al.
Fig. 2 An example of a measurement procedure in progress, with both clean pulsations and motion artifacts in
the “Measure” window. (A) Measurement meter, with colored segments ranging between 0 (empty) and 13
(full). 0 (empty) = measurement off, 1 = preparing for measurement (during camera calibration or when finger
is not ready), 2–12 = measurement in progress, 13 (full) = completion of the one-shot measurement or begin-
ning of continuous measurement. (B) The mean heart rate (HR) over the preceding (approximately) 10 s during
measurement, the mean HR over the whole period when the measurement is finished; beats per minute (bpm).
(C) The mean logarithmically transformed (ln) normalized pulse volume (NPV) over the preceding (approxi-
mately) 10 s, the mean (ln) NPV over the whole period when the measurement is finished; arbitrary unit (a.u.).
(D) Measured pulse wave. (E) Real-time color image of the finger-tip captured by the camera. (F) The result of
real-time analysis of the pulse wave. Red (the second brightest in gray scale) circle = peak, Blue (the darkest
in gray scale) circle = bottom, Yellow (the brightest in gray scale) circle (peak) = outlier of HR, Yellow circle (bot-
tom) = outlier of NPV. (G) Elapsed time; seconds (s) and sampling rate; frames per second (fps). (H) Start/Stop
button. Quoted from ref. 29 with some modification
2 Materials
3 Methods
3.1 Measuring PPG 1. Red, green, and blue PPG signals can be measured simultane-
from the ously from the CMOS camera (image sensor). In brief, the
CMOS Camera CMOS camera captures each light by using a set of very small
red-, green-, and blue-optical filters located with each pixel.
2. At the beginning of the measuring sequence, the CMOS cam-
era is set to video capture mode with 8 bits depth for red,
green, and blue light, with 192 × 144 pixels resolution.
3. The sampling frequency is set to 30 frames per second (fps)
(see Note 5).
4. The white balance, focus, and exposure (sensitivity to light)
mode of the CMOS camera are set to “Locked,” “Locked,”
and “ContinuousAutoExposure,” respectively [26].
5. Once actual measurement sequence has started after the LED
flash set to “on,” an image captured by the CMOS camera is
displayed on the measurement screen of iPhysioMeter
(Fig. 2E). Although an image captured by the CMOS camera
is square (Fig. 1c), here it is clipped in a circle and displayed.
All captured images are averaged among all of the 192 × 144 pix-
els to produce one red, green, and blue value per each frame.
6. During measurement, it is most important is to set the light sen-
sitivity of the CMOS camera to an appropriate value that enables
it to measure PPG stably. If sensitivity is too high or too low, it
makes it impossible to make PPG measurements. The image
shown in Fig. 1c was captured with an appropriate sensitivity.
310 Kenta Matsumura et al.
3.2 Detecting Red and blue signals derived at each frame are used to judge whether
a Finger the finger is on the camera or not. The actual measurement of PPG
will be conducted only when iPhysioMeter detects the finger on the
camera. iPhysioMeter deems the finger to be on the camera if the
red signal is over 180 (minimum = 0 and maximum = 255, due to
each color being 8-bit depth) and the blue signal is below 180.
A pair of these criteria varies depending on the selections of “Finger
Detection” in “Settings” (Fig. 3b). The criteria for “Strict,”
“Moderate,” “Loose,” and “N/A” are 200, 200, 180, and −1 (thus
always “on”) for red and 50, 120, 180, and 256 (thus always “on”)
for blue, respectively. When “N/A” is chosen, the analysis will be
conducted regardless of the finger-camera positioning.
3.3 Calculating HR 1. iPhone 4s and later models have enough CPU power to pro-
and NPV cess the PPG signals from all the three light color at once.
However, green light PPG has superior features as described in
the Introduction. Figure 4 shows a typical example of simulta-
neous recordings of red, green, and blue light PPG signals and
three-axis accelerations (see Note 7) with (Right) and without
(Left) intentionally produced motion artifact. The chart clearly
illustrates the higher tolerance to motion artifact in green light
PPG compared to red and blue light PPG. So only the data
from green PPG are subsequently processed.
2. The PPG signal is smoothed by using a first order IIR-type low
pass filter to extract the DC component of the PPG signal, and
then the AC component is separated from the DC component
by using the same type of high pass filter. Figure 5 shows
frequency characteristics of the filters used in iPhysioMeter.
Although there is no strict rule in filter design, the main band-
width of the PPG power spectrum is known to be less than 5 Hz
iPhysioMeter: A Smartphone Photoplethysmograph for Measuring Various… 311
Fig. 3 Links between “Environment” windows. (a) Environment window. You can change the “environment”
that is the set of measured data and the settings. This makes it easy to manage concurrent experiments and/
or measurements of multiple individuals. The “>” button is checked for the currently selected environment.
You can select another environment by tapping its name only when measurement is not ongoing. Measurement
data are stored in the currently selected environment. It may take sometime to change to or from the environ-
ment that has a large volume of data. (b) System and operation settings. This window opens when tapping a
“>” button in a row (a). Although there are many settings, often-used settings are: (1) Sound: the pulse detec-
tion sound (OFF, POP, Morse). (2) “Complete” Vibration: the vibration when measurement stops (ON/OFF). The
setting is invalid for the iPod touch fifth generation. (3) Operation Mode: you can select either “One-Shot,”
measurement automatically stops when the measurement meter (see Fig. 2A) becomes full or “Continuous,”
measurement stops only when you press the Stop button. Quoted from ref. 29 with some modification
in ordinary practice [27, 28]. This finding holds true for PPG
signals derived using iPhysioMeter. Figure 6 shows a typical
example of the power spectra of red, green, and blue light PPG
raw (non-filtered) signals derived at rest using iPhysioMeter.
3. For all of the stored green light PPG data, 300 ms (9 frames at
30 fps) after their capture, it is determined whether or not
there is a peak (systolic) point of the AC waveform. This is car-
ried out using a local maximum search algorithm (Fig. 7).
Specifically, if the value at this time point is a maximum among
the values over the period from 333.33 ms (10 frames at
30 fps) before this point to 233.33 ms (7 frames at 30 fps) after
this point, then the point is judged as the peak. Only after a
peak has been detected is the search for a foot (diastolic) point
begun. The foot point corresponding to this peak point is
312 Kenta Matsumura et al.
1.2
Red PPG
(a.u.)
−1.2
1.5
Green PPG
(a.u.)
−1.5
0.5
Blue PPG
(a.u.)
−0.5
0.5
x-axis A
(G)
−0.5
0.5
y-axis A
(G)
−0.5
−0.5
z-axis A
(G)
-1
−1.5
90 95 100 105 110 115
Time (s)
Fig. 4 A typical example of simultaneous recordings of the alternating-current (AC) components of red, green,
and blue photoplethysmogram (PPG)s and three-axis acceleration (A)s. The left side and the right side of the
chart correspond to in the absence and presence of motion artifact, respectively. Red and blue closed circles
represent the peak and foot points identified by a beat-by-beat auto analysis algorithm equipped with iPhysi-
oMeter. Open circles identify outlier points, that may have arisen from misidentification and/or omission of an
expected preceding point, are also judged by this algorithm. Quoted from ref. 22 with some modification. a.u.
arbitrary units
−3
DC filter
−9
AC filter
−12
0.1 1 10
Frequency (Hz)
3.4 Using 1. iPhysioMeter has many system and operation settings, but all of
iPhysioMeter these are set automatically according to the device on which
iPhysioMeter is run. Therefore it is not necessary to set
314 Kenta Matsumura et al.
Red PPG
0.2
HR
0.15
Power (a.u.)
2nd harmonics
0.1
of HR
0.05
3nd harmonics
of HR
0
0 5 10 15
Hz
Green PPG
HR
2
1.5
Power (a.u.)
2nd harmonics
1 of HR
0
0 5 10 15
Hz
Blue PPG HR
0.15
Power (a.u.)
0.1
2nd harmonics
0.05
of HR
0
0 5 10 15
Hz
Corresponding R-R
intervals of ECG
Fig. 6 A typical example of power spectra of red, green, and blue light photoplethysmography (PPG) raw (non-
filtered) signals. Dotted line around 1.5 Hz represents mean heart rate (HR) expressed in Hz calculated from
corresponding R-R intervals of electrocardiogram (ECG). Data points are 512 and sampling frequency (Hz) are
512 and 30, respectively. Quoted from ref. 22 with some modification. a.u. arbitrary units
iPhysioMeter: A Smartphone Photoplethysmograph for Measuring Various… 315
AC signal (a.u.)
0.4
0 Foot
−0.4
333 ms 233 ms
−0.8
91 91.5 92
Tims (s)
Fig. 7 Schematic drawing illustrating the peak and foot detection algorithm
implemented in iPhysioMeter. AC alternating current
Model
4s 5 iPod 5s
−2
−3 L
lnNPV (a.u.)
M
−4
S
−5
−6
−7
use the older iOS products and versions that are proven to be
stable but have enough power to run iPhysioMeter.
2. Whenever possible, it is recommended that you stop any back-
ground Apps before running iPhysioMeter.
3.5 Preparation 1. Firmly hold the iPhone or iPod touch in which iPhysioMeter
for Measurement was installed, so that the tip of the index finger completely cov-
ers both the LED flash and the CMOS camera lens (Fig. 1a).
As described earlier, the PPG signal is calculated by averaging
over the whole area of the captured image. This means that if
changes in background light levels are captured, there may not
be a correct measurement. In addition, it is important to note
that the background light should be constant during the mea-
surements if possible. If the background light level is too
bright, such as under direct sunlight, it might be impossible to
conduct the PPG measurement due to saturation of the cam-
era. Although almost all background green and blue light is
absorbed by the biological tissue [30], red light can be trans-
mitted through the finger and then exert an adverse influence
on the adjustment of the exposure of the CMOS camera.
2. It is important to keep the measuring site as constant as possi-
ble in every measurement, not only across the multiple mea-
surements in one person but also across multiple individuals.
Although the HR calculation is virtually free from this kind of
problem, the NPV is influenced by the measuring site.
3. It is also important to keep the finger—iPhone (or iPod touch)
contact pressure as constant as possible. Fluctuations in con-
tact pressure are one of the major sources of error in the NPV
measurements [21, 22]. So it is recommended to hold the
iPhone firmly, although excessive contact pressure will occlude
arterioles, and it will then be impossible to record the
photoplethysmogram.
4. To obtain reliable measurements of NPV, it is recommended
to hold the iPhone or iPod touch at heart level. Positioning
the device below or above heart level will affect the NPV value.
The HR calculation is virtually free from this kind of
problem.
5. Having dealt with all of the above matters, run iPhysioMeter
by tapping its icon on the home screen (or if already launched,
tap “Play” button), then the measurement will start automati-
cally (Fig. 2).
6. Ensure that all preparations for using iPhysioMeter are made
before the measurement process commences. Of particular
importance is to start the measurement after both the LED
flash and camera lens are covered with the index finger com-
pletely unless “Exposure Adj (adjustment)”is set to “iOS
iPhysioMeter: A Smartphone Photoplethysmograph for Measuring Various… 317
3.6 During 1. Keep the finger and the body as still as possible. Measurement is
Measurement possible under a certain level of motion artifacts because iPhysi-
oMeter benefits by the use of the green light PPG [22–25], but
keeping the finger and the body still produces better results.
2. If the finger is extremely cold, the PPG signal might not be
obtained. In such a case, PPG signals cannot be obtained even
with conventional PPG instrumentation. The only option in
this situation is to wait for re-warming of the finger.
3. The measurement will pause if iPhysioMeter detects the finger
off the camera unless “Finger Detection” in “Settings” is set to
“N/A” (Fig. 3b). Note that when resuming from this pause,
camera calibration will not be carried out when “Exposure
Adj” in “Settings” is set to “Fixed.”
4. Make sure that the PPG measurement is running correctly.
With correct operation the PPG waveform and the results of
on-line beat-by-beat analysis to give HR and NPV should be
displayed together (Fig. 2).
5. During the operation of iPhysioMeter within an experimental
procedure, it is beneficial to cover the iPhone or iPod touch
and measurement site with a black cloth to prevent the partici-
pants from looking at the display. Although this practice also
prevents the experimenters from looking at the display, the
iPhone or iPod touch’s screen can be mirrored to an external
LCD monitor by using a display adapter that is released from
Apple Inc. or third party organizations (e.g., [21]).
6. As described above, outlier judgment by iPhysioMeter is per-
formed on the assumption that HR and NPV are relatively
constant over 10 s periods, and is affected by the “Outlier
Check” selection in “Settings” (Fig. 3b). So this judgment is
not an absolute judgment, and should only be used as a guide.
7. The measurement stops automatically when the measurement
meter reaches full if “Operation Mode” in “Settings” is set to
“One-Shot” (Fig. 3b). You can also stop the measurement at
any time by tapping the “Stop” button (Fig. 2).
8. If “Operation Mode” in “Settings” is set to “Continuous”
(Fig. 3b), you can make continuous measurements indefinitely,
or at least until the battery is fully discharged. However, it is
very important to pay special attention to the inevitable tem-
perature increase in the LED flash. If the measurement period
318 Kenta Matsumura et al.
3.7 After 1. Name the measurement that was automatically saved to iPhone
the Measurement Is or iPod touch’s main storage when measurement finishes
Finished (Fig. 9e). A “measurement” is defined as all of the data col-
lected during the period from the “play” button being tapped
to the end of the measurement.
2. Although you do not have to name the measurement, it is highly
recommended to do this in order to reduce mistakes in data
management. In addition, because HR and NPV related mea-
sures can be affected by many factors such as posture, mental
state, time after eating, smoking, exercising or taking medicine,
last day sleep, and air temperature, consider including these sta-
tus items to the name in case these variables are not controlled.
In fact, in our experiments, we make every effort to control
these factors to the maximum extent (e.g., [4, 21, 22, 31]).
Fig. 9 (continued) (c) or tapping “Open…” button in (b) or (d). You can name or rename the measurement,
check, e-mail, and erase each stored dataset on this screen. Data contents include the beat-by-beat data that
can be checked and sent via e-mail. They are (1) Time: the time (s) from the start of the measurement. (2) gTime:
the time (s) from when the start button was tapped. There is an approximately 1.3 s latency between Time and
gTime. (3) HR: the beat-by-beat value automatically calculated. (4) lnNPV: the beat-by-beat value automatically
calculated. (5) AC: AC component of the pulse wave. (6) DC: DC component of the pulse wave. (7) A_x, A_y, and
A_z: three-axis g-forces when the peak was detected. (8) HR_Out: checked when HR is judged as an outlier. (9)
NPV_Out: checked when lnNPV is judged as an outlier. Quoted from ref. 29 with some modification
iPhysioMeter: A Smartphone Photoplethysmograph for Measuring Various… 319
Fig. 9 Links among “Graph” windows. (a) Graph window. You can zoom in or zoom out, or scroll the graph, and
can select either heart rate (HR) or normalized pulse volume (NPV) graphs by tapping the right-bottom button
in the display. (b) Popup Summary. When tapping the graph in (a), the popup summary of the nearest place
data appears. The summary includes the following information: serial number; date when the start button was
tapped; name; measurement duration (s); the number of segment, which increases by detaching the finger;
the number of beats; the mean HR and NPV during the whole measuring period; frames per second (fps). (c) A
list of the whole measured data. This data list opens when tapping “Data>>” button in (a). The serial number,
the mean HR and lnNPV during the whole measuring period, the number of beats, and name are also shown.
(d) Popup summary. When tapping the row in (c), the popup summary of the selected data appears. The con-
tents are same as (b). (e) Data management menu. This window opens when tapping a “>” button in a row
320 Kenta Matsumura et al.
3.8 Other Matters 1. There are likely to be individual differences in the LED flash
Considered Significant and CMOS camera even if the same models of particular
devices are used (for example, two iPhone 4Ss). So if you use
iPhysioMeter in your experiment, use only one device (for
example, your iPhone 4s) for the whole study. If that really is
impossible, then devices should be randomized (for example,
your iPhone 4S and your colleague’s iPhone 4S) across partici-
pants. Mixing usage of models (for example, your iPhone 4S
and colleague’s iPhone 5) should be avoided wherever possi-
ble. These considerations also hold true with respect to the
iOS version being used. Having said that, the HR calculation
is virtually free from this kind of problem.
2. The sampling rate of iPhysioMeter is set to 30 fps (see Note 5).
At this sampling speed, beat-by-beat HR can have in principle
approximately ±2 and ±6 bpm error at 60 and 100 bpm,
respectively (Fig. 10). One of the best practices to overcome
this problem is to average HR values over 20-s or 60-s, for
example, and get one averaged value. This method has been
shown to yield quite accurate values for HR as well as NPV as
compared with those derived from standard laboratory elec-
200
Ȉ = 13.6 bpm
150
Heart Rate (bpm)
Ȉ = 2.1 bpm
50
0
0 500 1000 1500 2000
100 2
y = 1.006 x − 0.288 Bias = −0.12, SD = 0.36
r = 0.9991
HR by iPhone 4s (bpm)
Difference (bpm)
1
80
60
−1
0 −2
0 60 80 100 40 60 80 100
HR by ECG (bpm) Average (bpm)
−6 −5 −4 −3 0 2
0 Bias = 0.01, SD = 0.43
y = 0.936 x − 0.279
lnNPV by iPhone 4s (a.u.)
r = 0.791 1
Difference(a.u.)
−3
0
−4
−1
−5
−2
−6 −6 −5 −4 −3
Fig. 11 Scatter plots (Left) and their Bland-Altman plots (Right; [35]) of heart rate (HR; Upper) and logarithmically
transformed normalized pulse volume (lnNPV; Lower) between iPhysioMeter and standard laboratory instru-
ments. Solid lines and dashed lines represent linear fit and its 95 % confidential interval (CI)s of geometric mean
regression (GMR; [36]), respectively. Quoted from ref. 22 with some modification. ECG electrocardiograph, PPG
photoplethysmograph, NIR=near-infrared, bpm=beats per minute, a.u. arbitrary units, CI confidential interval
4 Notes
2 2
0 0
0 2 4 6
0 2 4 6
LF/HF by ECG LF/HF by ECG
Green PPG
6
y = 0.734 x − 0.214
r = .909
LF/HF by Green Light
0
0 2 4 6
LF/HF by ECG
Blue PPG
6
y = 0.626 x − 0.408
r = .615
LF/HF by Blue Light
0
0 2 4 6
LF/HF by ECG
Fig. 12 Scatter plots of low to high frequencies ratio (LF/HF) between iPhysioMeter and electrocardiograph (ECG; Left)
and standard laboratory near-infrared (NIR) photoplethysmography (PPG) and ECG (Right). Solid lines and dashed lines
represent linear fit and its 95 % confidential interval (CI)s of geometric mean regression (GMR; [36]), respectively
324 Kenta Matsumura et al.
References
1. Parker KH (2009) A brief history of arterial wave responses to stress following alcohol intake in
mechanics. Med Biol Eng Comput 47:111–118. social drinkers who are at risk of hazardous
doi:10.1007/s11517-009-0440-5 drinking. Biol Psychol 93:9–16. doi:10.1016/j.
2. Challoner AVJ (1979) Photoelectric plethys- biopsycho.2012.12.009
mography for estimating cutaneous blood flow. 8. Yamakoshi T, Matsumura K, Hanaki S, Rolfe
In: Rolfe P (ed) Noninvasive physiological P (2013) Cardiovascular hemodynamic
measurements, vol 1. Academic, London, effects of Red Bull(R) Energy Drink
pp 125–151 during prolonged, simulated, monotonous
3. Matsumura K, Yamakoshi T, Yamakoshi Y, driving. SpringerPlus 2:215. doi:10.1186/
Rolfe P (2011) The effect of competition on 2193-1801-2-215
heart rate during kart driving: a field study. 9. Yamakoshi T, Matsumura K, Rolfe P, Hanaki S,
BMC Res Notes 4:342. Ikarashi A, Lee J, Yamakoshi K (2014) Potential
doi:10.1186/1756-0500-4-342 for health screening using long-term cardiovas-
4. Matsumura K, Yamakoshi T, Noguchi H, Rolfe cular parameters measured by finger volume-
P, Matsuoka Y (2012) Fish consumption and oscillometry: pilot comparative evaluation in
cardiovascular response during mental stress. regular and sleep-deprived activities. IEEE J
BMC Res Notes 5:288. Biomed Health Inform 18:28–35.
doi:10.1186/1756-0500-5-288 doi:10.1109/JBHI.2013.2274460
5. McNally RJ, Lasko NB, Clancy SA, Macklin 10. Allen J (2007) Photoplethysmography and its
ML, Pitman RK, Orr SP (2004) application in clinical physiological measure-
Psychophysiological responding during script- ment. Physiol Meas 28:R1–R39.
driven imagery in people reporting abduction doi:10.1088/0967-3334/28/3/R01
by space aliens. Psychol Sci 15:493–497. 11. Pitman RK, Orr SP, Forgue DF, de Jong JB,
doi:10.1111/j.0956-7976.2004.00707.x Claiborn JM (1987) Psychophysiologic assess-
6. al’Absi M, Nakajima M, Hooker S, Wittmers L, ment of posttraumatic stress disorder imagery
Cragin T (2012) Exposure to acute stress is in Vietnam combat veterans. Arch Gen
associated with attenuated sweet taste. Psychiatry 44:970–975
Psychophysiology 49:96–103. 12. Kuvin JT, Patel AR, Sliney KA, Pandian NG,
doi:10.1111/j.1469-8986.2011.01289.x Sheffy J, Schnall RP, Karas RH, Udelson JE
7. Nakajima M, Kumar S, Wittmers L, Scott MS, (2003) Assessment of peripheral vascular endo-
al’Absi M (2013) Psychophysiological thelial function with finger arterial pulse wave
iPhysioMeter: A Smartphone Photoplethysmograph for Measuring Various… 325
34. Giardino ND, Lehrer PM, Edelberg R (2002) methods of clinical measurement. Lancet
Comparison of finger plethysmograph to ECG 1:307–310. doi:10.1016/
in the measurement of heart rate variability. S0140-6736(86)90837-8
Psychophysiology 39:246–253. doi:10.1017/ 36. Ludbrook J (1997) Comparing methods of
S0048577202990049 measurements. Clin Exp Pharmacol Physiol
35. Bland JM, Altman DG (1986) Statistical meth- 24:193–203. doi:10.1111/j.1440-1681.1997.
ods for assessing agreement between two tb01807.x
Chapter 22
Abstract
With the ubiquity of smartphones and the rising technology of 3D printing, novel devices can be
developed that leverage the “computer in your pocket” and rapid prototyping technologies toward
scientific, medical, engineering, and creative purposes. This paper describes such a device: a simple
3D-printed extension for Apple’s iPhone that allows the sound from an off-the-shelf acoustic stetho-
scope to be recorded using the phone’s built-in microphone. The attachment’s digital 3D files can be
easily shared, modified for similar phones and devices capable of recording audio, and in combination
with 3D printing technology allow for fabrication of a durable device without need for an entire fac-
tory of expensive and specialized machining tools. It is hoped that by releasing this device as an open
source set of printable files that can be downloaded and reproduced cheaply, others can make use of
these developments where access to cost-prohibitive, specialized medical instruments are not avail-
able. Coupled with specialized smartphone software (“apps”), more sophisticated and automated
diagnostics may also be possible on-site.
Key words iPhone, Stethoscope, Audio recording, 3D printing, STL, Shapeways, Smartphone, App,
Sound, Selective laser sintering, Fused deposition modeling
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_22, © Springer Science+Business Media New York 2015
327
328 Jeff Thompson
Fig. 2 A CAD rendering of the 3D printed attachment, which slides tightly onto the bottom of the iPhone. Note
how the sound from the stethoscope’s tube is funneled directly into the phone’s built-in microphone. Shapes
like this are difficult to manufacture, making 3D printing an ideal solution in such cases
2 Materials
3 Methods
3.1 Printing Shapeways, a 3D printing service based in the USA and the
the Attachment Using Netherlands, provides very high-quality printing in a variety of
Shapeways’ 3D materials.
Printing Service 1. Visit the following link to order a 3D-printed version of the
stethoscope attachment: http://shpws.me/oQUX.
2. Choose a material color from the “Material Options” drop-
down menu—default is white (see Note 2).
3. Click the “Buy Now” button to order the printed model in the
material specified (see Note 3); at time of writing, commercial
printing of the attachment was approximately $30 USD.
3.3 Prepare A single length of flexible PVC tubing is needed to connect the chest-
Stethoscope Tubing piece to the 3D printed attachment. This can be repurposed from an
existing stethoscope or bought in bulk and cut to size (see Note 1).
1. Cut one section of stethoscope tubing using a sharp knife or
scissors. If using the tubing from an existing stethoscope, cut
below the fork to form a single tube. Length is variable but less
than 15″ (38 cm) can make it difficult to maneuver; very long
tubing will likely cause a reduction in the fidelity of the audio.
3.5 Test and Record 1. Turn on the phone and launch an audio recording app like
Voice Recorder (see Note 7).
2. Set your app’s settings to highest quality possible, if available.
Higher sample rate and bit depth will result in higher-fidelity
recordings. Avoid recording in compressed formats like MP3 if
non-compressed formats such as WAV or AIFF are available.
3. Press record and test the stethoscope. Depending on your
app’s sensitivity settings, loud sounds such as tapping the
stethoscope head should be very clearly picked up as sharp
spikes by the microphone (see Note 8).
3.6 Audio Processing While the recordings from the stethoscope may be used without any
and Analysis processing, some filtering will make the isolation of certain sounds
easier, especially for automated analysis.
1. A variety of adult chest sounds can be heard with a stethoscope
and can be identified by their frequency ranges. These include
“low heart sounds (including first, second, and third heart
sounds) from 20 to 115 Hz; medium/high heart sounds
(including systolic and diastolic murmurs) from 200 to
660 Hz; vesicular (normal) breathing from 150 to 1,000 Hz;
bronchial breathing from 240 to 1,000 Hz; and crepitations
(crackles) greater than or equal to 750 Hz” (see ref. 1).
332 Jeff Thompson
Fig. 4 The waveform from an audio recording of a human heartbeat with no filter applied. While the heartbeat
is visible, there is significant noise in the recording
Fig. 5 The same recording of a human heartbeat as Fig. 4 with a low-pass filter applied. The heartbeat is much
more clearly visible, especially to the untrained eye
Fig. 6 Frequency analysis of a recording of a human heartbeat with no filter applied. While the noise floor for
the iPhone’s built-in microphone is rather high, the low-frequency heartbeat is still clearly visible
4 Notes
References
Abstract
Smartphones and portable media devices are both equipped with sensor components, such as accelerom-
eters. A software application enables these devices to function as a robust wireless accelerometer platform.
The recorded accelerometer waveform can be transmitted wireless as an e-mail attachment through con-
nectivity to the Internet. The implication of such devices as a wireless accelerometer platform is the experi-
mental and post-processing locations can be placed anywhere in the world. Gait was quantified by mounting
a smartphone or portable media device proximal to the lateral malleolus of the ankle joint. Attributes of
the gait cycle were quantified with a considerable accuracy and reliability. The patellar tendon reflex
response was quantified by using the device in tandem with a potential energy impact pendulum to evoke
the patellar tendon reflex. The acceleration waveform maximum acceleration feature of the reflex response
displayed considerable accuracy and reliability. By mounting the smartphone or portable media device to
the dorsum of the hand through a glove, Parkinson’s disease hand tremor was quantified and contrasted
with significance to a non-Parkinson’s disease steady hand control. With the methods advocated in this
chapter, any aspect of human movement may be quantified through smartphones or portable media
devices and post-processed anywhere in the world. These wearable devices are anticipated to substantially
impact the biomedical and healthcare industry.
Key words Portable media device, iPod, Smartphone, iPhone, Wireless accelerometer, Wearable device,
Quantification, Patellar tendon reflex, Reflex response, Gait, Gait analysis, Parkinson’s disease, Tremor
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_23, © Springer Science+Business Media New York 2015
335
336 Robert LeMoyne and Timothy Mastroianni
1.1 Smartphones/ One of the most evolved biomedical applications for wearable and
Portable Media wireless devices pertains to gait analysis [6, 7]. During the 1950s
Devices for Gait the accelerometer applications were recommended for the role of
Quantification quantifying human movement. Industries associated with the
accelerometer technology space, such as the automotive industry,
Use of Smartphones and Portable Media Devices for Quantifying Human Movement… 337
1.2 Smartphones/ The tendon reflex is a feature that is inherently integrated with gait
Portable Media cycle [13]. The wireless quantified reflex device methodology was
Devices for Tendon developed by LeMoyne et al. [13, 50]. The intrinsic attribute of the
Reflex Response wireless quantified reflex device is the use of wireless accelerometer
Quantification nodes, with one node secured by elastic band proximal to the ankle
joint and eventually another node mounted to the swing arm of the
impact pendulum. A potential energy impact pendulum mounted
to a reflex hammer was incorporated to precisely evoke the patellar
Use of Smartphones and Portable Media Devices for Quantifying Human Movement… 341
2 Materials
3 Methods
3
2.5
2
1.5
1
0.5
0
0 2 4 6 8 10
Time (seconds)
Fig. 2 Gait acceleration waveform from smartphone (iPhone) application [2]. Reproduced from 2010
with permission from IEEE
Table 1
Smartphone (iPhone) quantified gait parameters [2]
3.2 Implementation Figure 3 illustrates the wireless quantified reflex system that applies
of a Portable Media the portable media device as a wireless accelerometer platform. The
Device anatomical mounting strategy is also exemplified in Fig. 3 with the
and Smartphone portable media device mounted proximal to the ankle and secured
for Quantification by the elastic band of a sock. The potential energy reflex input sys-
of Tendon Reflex tem demonstrates the capacity to accurately and reliably target the
Response patellar tendon with precise and discrete settings of potential energy.
For the test and evaluation experiment of the wireless reflex quanti-
fication system with a portable media device, a potential energy set-
ting of 30° relative to gravity vector was selected [4].
The goal of the portable media device wireless accelerometer
platform for reflex quantification was to provide a simplified alter-
native for quantifying the patellar tendon reflex response.
348 Robert LeMoyne and Timothy Mastroianni
Fig. 4 Portable media device (iPod) wireless accelerometer reflex quantification system acceleration waveform
of the reflex response [4]. Reproduced from 2012 with permission from IEEE
Use of Smartphones and Portable Media Devices for Quantifying Human Movement… 349
Table 2
Portable media device (iPod) wireless accelerometer device maximum reflex response (30 trials) [4]
3.3 Implementation Figure 5 illustrates the functional simplicity for using the smart-
of a Smartphone phone application to quantify Parkinson’s disease hand tremor.
for Quantifying The smartphone is secured to the dorsum of the hand by a glove.
Parkinson’s Disease The experiment and post-processing were conducted in remote
Tremor locations. The experimental test and evaluation of the application
involved a subject with Parkinson’s disease and a subject without
Parkinson’s disease representing a control. The setting may be
selected at the convenience of the subject. The recorded acceler-
ometer signal for both subjects was conveyed by wireless
transmission through e-mail. Post-processing was conducted thou-
sands of miles remote to the experimentation site [1].
1.2
1.1
1
0.9
0.8
0.7
0.6
0 2 4 6 8 10 12
Time (seconds)
Fig. 6 Acceleration waveform of static hand condition for subject without Parkinson’s disease [1]. Reproduced
from 2010 with permission from IEEE
1.2
1.1
1
0.9
0.8
0.7
0.6
0 2 4 6 8 10 12
Time (seconds)
Fig. 7 Acceleration waveform of hand tremor attributes for subject with Parkinson’s disease [1]. Reproduced
from 2010 with permission from IEEE
Frequency response of subject without Parkinson's disease tremor using Blackman window
10
6
Amplitude
0
0 5 10 15 20 25 30 35 40 45 50
Frequency (Hz)
Fig. 8 Frequency response with Blackman window for the subject without Parkinson’s disease tremor [1].
Reproduced from 2010 with permission from IEEE
Frequency response of subject with Parkinson's disease tremor using Blackman window
10
6
Amplitude
0
0 5 10 15 20 25 30 35 40 45 50
Frequency (Hz)
Fig. 9 Frequency response with Blackman window for the subject with Parkinson’s disease tremor [1].
Reproduced from 2010 with permission from IEEE
4 Notes
Acknowledgement
References
1. LeMoyne R, Mastroianni T, Cozza M, Coroian applications in biofeedback gait rehabilitation
C, Grundfest W (2010) Implementation of an concepts and movement disorder characteriza-
iPhone for characterizing Parkinson’s disease tion. In: Barros de Mello CA (ed) Biomedical
tremor through a wireless accelerometer appli- engineering. Vienna, Austria, Intech, Ch 10
cation. In Proceedings of the 32nd Annual 8. Culhane KM, O’Connor M, Lyons D, Lyons GM
International Conference of the IEEE EMBS, (2005) Accelerometers in rehabilitation medicine
pp 4954–4958 for older adults. Age Ageing 34:556–560
2. LeMoyne R, Mastroianni T, Cozza M, Coroian 9. Jovanov E, Milenkovic A, Otto C, de Groen
C, Grundfest W (2010) Implementation of an PC (2005) A wireless body area network of
iPhone as a wireless accelerometer for quantify- intelligent motion sensors for computer
ing gait characteristics. In Proceedings of the assisted physical rehabilitation. J Neuroeng
32nd Annual International Conference of the Rehabil 2:6
IEEE EMBS, pp 3847–3851 10. Saremi K, Marehbian J, Yan X, Regnaux JP,
3. LeMoyne R, Mastroianni T, Grundfest W Elashoff R, Bussel B, Dobkin BH (2006)
(2011) Wireless accelerometer iPod applica- Reliability and validity of bilateral thigh and
tion for quantifying gait characteristics. In foot accelerometry measures of walking in
Proceedings of the 33rd Annual International healthy and hemiparetic subjects. Neurorehabil
Conference of the IEEE EMBS, Neural Repair 20:297–305
pp 7904–7907 11. Kavanagh JJ, Morrison S, James DA, Barrett R
4. LeMoyne R, Mastroianni T, Grundfest W, (2006) Reliability of segmental accelerations
(2012) Quantified reflex strategy using an iPod measured using a new wireless gait analysis sys-
as a wireless accelerometer application. In tem. J Biomech 39:2863–2872
Proceedings of 34th International Conference 12. Kavanagh J, Barrett R, Morrison S (2006) The
of the IEEE EMBS, pp 2476–2479 role of the neck and trunk in facilitating head
5. LeMoyne R, Mastroianni T, Grundfest W, stability during walking. Exp Brain Res
Nishikawa K (2013) Implementation of an 172:454–463
iPhone wireless accelerometer application for 13. LeMoyne R, Mastroianni T, Coroian C,
the quantification of reflex response. In Grundfest W (2011) Tendon reflex and strate-
Proceedings of 35th International Conference gies for quantification, with novel methods
of the IEEE EMBS, pp 4658–4661 incorporating wireless accelerometer reflex
6. Patel S, Park H, Bonato P, Chan L, Rodgers M quantification devices, a perspective review. J
(2012) A review of wearable sensors and sys- Mech Med Biol 11:471–513
tems with application in rehabilitation. J 14. Lee JA, Cho SH, Lee JW, Lee KH, Yang HK
Neuroeng Rehabil 9:21 (2007) Wearable accelerometer system for
7. LeMoyne R, Coroian C, Mastroianni T, measuring the temporal parameters of gait. In
Opalinski P, Cozza M, Grundfest W (2009) Proceedings of the 29th Annual International
The merits of artificial proprioception, with Conference of the IEEE EMBS, pp 483–486
356 Robert LeMoyne and Timothy Mastroianni
15. Lee JA, Cho SH, Lee YJ, Yang HK, Lee JW 27. Kavanagh JJ (2009) Lower trunk motion and
(2010) Portable activity monitoring system for speed-dependence during walking. J Neuroeng
temporal parameters of gait cycles. J Med Syst Rehabil 6:9
34:959–966 28. Lai DT, Charry E, Begg R, Palaniswami M
16. Bamberg SJ, Benbasat AY, Scarborough DM, (2008) A prototype wireless inertial-sensing
Krebs DE, Paradiso JA (2008) Gait analysis device for measuring toe clearance. In Proceedings
using a shoe-integrated wireless sensor system. of the 30th Annual International Conference of
IEEE Trans Inf Technol Biomed 12:413–423 the IEEE EMBS, pp 4899–4902
17. LeMoyne R, Coroian C, Mastroianni T, 29. Reininga IH, Stevens M, Wagenmakers R,
Grundfest W (2009) Wireless accelerometer Bulstra SK, Groothoff JW, Zijlstra W (2012)
assessment of gait for quantified disparity of Subjects with hip osteoarthritis show distinc-
hemiparetic locomotion. J Mech Med Biol tive patterns of trunk movements during gait-a
9:329–343 body-fixed-sensor based analysis. J Neuroeng
18. LeMoyne R, Coroian C, Mastroianni T (2009) Rehabil 9:3
Wireless accelerometer system for quantifying 30. Tura A, Raggi M, Rocchi L, Cutti AG, Chiari L
gait. In Proceedings of the IEEE/ICME (2010) Gait symmetry and regularity in trans-
International Conference on Complex Medical femoral amputees assessed by trunk accelera-
Engineering (CME), pp 1–4 tions. J Neuroeng Rehabil 7:4
19. LeMoyne R, Coroian C, Mastroianni T, 31. Tura A, Rocchi L, Raggi M, Cutti AG, Chiari L
Grundfest W (2008) Virtual proprioception. J (2012) Recommended number of strides for
Mech Med Biol 8:317–338 automatic assessment of gait symmetry and
20. LeMoyne R, Coroian C, Mastroianni T, Wu W, regularity in above-knee amputees by means of
Grundfest W, Kaiser W (2008) Virtual proprio- accelerometry and autocorrelation analysis.
ception with real-time step detection and pro- J Neuroeng Rehabil 9:11
cessing. In Proceedings of the 30th Annual 32. Hsu CC, Chen JH (2011) A novel sensor-
International Conference of the IEEE EMBS, assisted RFID-based indoor tracking system
pp 4238–4241 for the elderly living alone. Sensors
21. Mizuike C, Ohgi S, Morita S (2009) Analysis 11:10094–10113
of stroke patient walking dynamics using a tri- 33. Yeoh WS, Pek I, Yong YH, Chen X, Waluyo AB
axial accelerometer. Gait Posture 30:60–64 (2008) Ambulatory monitoring of human pos-
22. Guo Y, Wu D, Liu G, Zhao G, Huang B, Wang ture and walking speed using wearable acceler-
L (2012) A low-cost body inertial-sensing net- ometer sensors. In Proceedings of the 30th
work for practical gait discrimination of hemi- Annual International Conference of the IEEE
plegia patients. Telemed J E Health EMBS, pp 5184–5187
18:748–754 34. Choquette S, Hamel M, Boissy P (2008)
23. Prajapati SK, Gage WH, Brooks D, Black SE, Accelerometer-based wireless body area net-
McIlroy WE (2011) A novel approach to work to estimate intensity of therapy in post-
ambulatory monitoring: investigation into the acute rehabilitation. J Neuroeng Rehabil 5:20
quantity and control of everyday walking in 35. Hurkmans HL, Ribbers GM, Streur-
patients with subacute stroke. Neurorehabil Kranenburg MF, Stam HJ, van den Berg-
Neural Repair 25:6–14 Emons RJ (2011) Energy expenditure in
24. Bugané F, Benedetti MG, Casadio G, Attala S, chronic stroke patients playing Wii Sports: a
Biagi F, Manca M, Leardini A (2012) pilot study. J Neuroeng Rehabil 8:38
Estimation of spatial-temporal gait parameters 36. Huang H, Wolf SL, He J (2006) Recent devel-
in level walking based on a single accelerome- opments in biofeedback for neuromotor reha-
ter: validation on normal subjects by standard bilitation. J Neuroeng Rehabil 3:11
gait analysis. Comput Methods Programs 37. McGregor SJ, Armstrong WJ, Yaggie JA,
Biomed 108:129–137 Bollt EM, Parshad R, Bailey JJ, Johnson SM,
25. Watanabe T, Saito H, Koike E, Nitta K (2011) Goin AM, Kelly SR (2011) Lower extremity
A preliminary test of measurement of joint fatigue increases complexity of postural con-
angles and stride length with wireless inertial trol during a single-legged stance. J Neuroeng
sensors for wearable gait evaluation system. Rehabil 8:43
Comput Intell Neurosci 2011: 975193 38. Dozza M, Chiari L, Chan B, Rocchi L, Horak
26. Djurić-Jovičić MD, Jovičić NS, Popović DB FB, Cappello A (2005) Influence of a portable
(2011) Kinematics of gait: new method for audio-biofeedback device on structural
angle estimation based on accelerometers. properties of postural sway. J Neuroeng
Sensors 11:10571–10585 Rehabil 2:13
Use of Smartphones and Portable Media Devices for Quantifying Human Movement… 357
39. Lee BC, Kim J, Chen S, Sienko KH (2012) 50. LeMoyne R (2010) Wireless quantified reflex
Cell phone based balance trainer. J Neuroeng device, Ph.D. dissertation, UCLA, Biomed.
Rehabil 9:10 Eng. IDP
40. Djurić-Jovičić MD, Jovičić NS, Popović DB, 51. LeMoyne R, Coroian C, Mastroianni T,
Djordjević AR (2012) Nonlinear optimization Grundfest W (2008) Quantified deep tendon
for drift removal in estimation of gait kinemat- reflex device for response and latency, third
ics based on accelerometers. J Biomech generation. J Mech Med Biol 8:491–506
45:2849–2854 52. LeMoyne R, Mastroianni T, Kale H, Luna J,
41. Weenk D, van Beijnum BJ, Baten CT, Hermens Stewart J, Elliot S, Bryan F, Coroian C,
HJ, Veltink PH (2013) Automatic identifica- Grundfest W (2011) Fourth generation wire-
tion of inertial sensor placement on human less reflex quantification system for acquiring
body segments during walking. J Neuroeng tendon reflex response and latency. J Mech
Rehabil 10:31 Med Biol 11:31–54
42. LeMoyne R, Mastroianni T, Cozza M, Coroian 53. LeMoyne R, Coroian C, Mastroianni T, (2009)
C (2010) Quantification of gait characteristics Wireless accelerometer reflex quantification
through a functional iPhone wireless acceler- system characterizing response and latency. In
ometer application mounted to the spine. In Proceedings of 31st Annual International
Proceedings of ASME 2010 5th Frontiers in Conference of the IEEE EMBS,
Biomedical Devices Conference (BioMed), pp 5283–5286
pp 87–88 54. LeMoyne R, Jafari R (2006) Quantified deep
43. LeMoyne R, Mastroianni T, Cozza M, Coroian tendon reflex device, second generation. In
C (2010) iPhone wireless accelerometer appli- Proceedings of 15th International Conference
cation for acquiring quantified gait attributes. on Mechanics in Medicine and Biology
In Proceedings of ASME 2010 5th Frontiers in ICMMB-15 2006, 6–8 December 2006,
Biomedical Devices Conference (BioMed), Furama Riverfront, Singapore
pp 19–20 55. LeMoyne R, Dabiri F, Jafari R (2008)
44. LeMoyne R, Mastroianni T (2012) iWalk, a gait Quantified deep tendon reflex device, second
analysis device incorporating an iPod wireless generation. J Mech Med Biol 8:75–85
accelerometer application. In Proceedings of 56. LeMoyne R, Jafari R, Jea D (2005) Fully quan-
42nd Society for Neuroscience Annual Meeting, tified evaluation of myotatic stretch reflex. In
New Orleans, LA, October 13–17, 2012 Proceedings of 35th Society for Neuroscience
45. Nishiguchi S, Yamada M, Nagai K, Mori S, Annual Meeting, Washington, DC, November
Kajiwara Y, Sonoda T, Yoshimura K, Yoshitomi 12–16, 2005
H, Ito H, Okamoto K, Ito T, Muto S, Ishihara 57. LeMoyne R., Dabiri F., Coroian C.,
T, Aoyama T (2012) Reliability and validity of Mastroianni T., Grundfest W. (2007)
gait analysis by android-based smartphone. Quantified deep tendon reflex device for assess-
Telemed J E Health 18:292–296 ing response and latency. In Proceedings of
46. Yamada M, Aoyama T, Mori S, Nishiguchi S, 37th Society for Neuroscience Annual Meeting,
Okamoto K, Ito T, Muto S, Ishihara T, San Diego, CA, November 3–7, 2007
Yoshitomi H, Ito H (2012) Objective assess- 58. LeMoyne R., Coroian C., Mastroianni T.,
ment of abnormal gait in patients with rheuma- Cozza M., Grundfest W. (2010) Quantification
toid arthritis using a smartphone. Rheumatol of reflex response through an iPhone wireless
Int 32:3869–3874 accelerometer application. In Proceedings of
47. Mellone S, Tacconi C, Chiari L (2012) Validity 40th Society for Neuroscience Annual Meeting,
of a smartphone-based instrumented timed up San Diego, CA, November 13–17, 2010
and go. Gait Posture 36:163–165 59. LeMoyne R., Mastroianni T. (2011) Reflex
48. Palmerini L, Mellone S, Rocchi L, Chiari L response quantification using an iPod wireless
(2011) Dimensionality reduction for the quan- accelerometer application. In Proceedings of
titative evaluation of a smartphone-based timed 41st Society for Neuroscience Annual Meeting,
up and go test. In Proceedings of the 33rd Washington, DC, November 12–16, 2011
Annual International Conference of the IEEE 60. Seeley RR, Stephens TD, Tate P (2003)
EMBS, pp 7179–7182 Anatomy and physiology. McGraw-Hill,
49. Wagner R, Ganz A (2012) PAGAS: portable New York, NY
and accurate gait analysis system. In 61. Kandel ER, Schwartz JH, Jessell TM (2000)
Proceedings of the 34th Annual International Principles of neural science. McGraw-Hill,
Conference of the IEEE EMBS, pp 280–283 New York, NY
358 Robert LeMoyne and Timothy Mastroianni
62. Diamond MC, Scheibel AB, Elson LM (1985) mated, at-home assessment of mobility among
The human brain coloring book. Harper patients with Parkinson disease, using a body-
Perennial, New York, NY worn accelerometer. Neurorehabil Neural
63. Bickley LS, Szilagyi PG (2003) Bates’ guide to Repair 25:810–818
physical examination and history taking. 74. LeMoyne R, Coroian C, Mastroianni T (2009)
Lippincott Williams and Wilkins, New York, NY Quantification of Parkinson’s disease character-
64. Nolte J, Sundsten JW (2002) The human istics using wireless accelerometers. In
brain: an introduction to its functional anat- Proceedings of the IEEE/ICME International
omy. Mosby, St. Louis, MO Conference on Complex Medical Engineering
65. Volkmann J, Moro E, Pahwa R (2006) Basic (CME), pp 1–5
algorithms for the programming of deep brain 75. Giuffrida JP, Riley DE, Maddux BN, Heldman
stimulation in Parkinson’s disease. Mov Disord DA (2009) Clinically deployable Kinesia tech-
21:S284–S289 nology for automated tremor assessment. Mov
66. Rouse AG, Stanslaski SR, Cong P, Jensen RM, Disord 24:723–730
Afshar P, Ullestad D, Gupta R, Molnar GF, 76. Cancela J, Pansera M, Arredondo MT, Estrada
Moran DW, Denison TJ (2011) A chronic gen- JJ, Pastorino M, Pastor-Sanz L, Villalar JL
eralized bi-directional brain-machine interface. (2010) A comprehensive motor symptom
J Neural Eng 8:1–36 monitoring and management system: the bra-
67. Obwegeser AA, Uitti RJ, Witte RJ, Lucas JA, dykinesia case. In Proceedings of the 32nd
Turk MF, Wharen RE Jr (2001) Quantitative Annual International Conference of the IEEE
and qualitative outcome measures after tha- EMBS, pp 1008–1011
lamic deep brain stimulation to treat disabling 77. Pastorino M, Cancela J, Arredondo MT,
tremors. Neurosurgery 48:274–281 Pansera M, Pastor-Sanz L, Villagra F, Pastor
68. Kumru H, Summerfield C, Valldeoriola F, MA, Martin JA (2011) Assessment of bradyki-
Valls-Solé J (2004) Effects of subthalamic nesia in Parkinson’s disease patients through a
nucleus stimulation on characteristics of EMG multi-parametric system. In Proceedings of the
activity underlying reaction time in Parkinson’s 33rd Annual International Conference of the
disease. Mov Disord 19:94–100 IEEE EMBS, pp 1810–1813
69. Keijsers NL, Horstink MW, Gielen SC (2006) 78. Cancela J, Pastorino M, Arredondo MT,
Ambulatory motor assessment in Parkinson’s Pansera M, Pastor-Sanz L, Villagra F, Pastor
disease. Mov Disord 21:34–44 MA, Gonzalez AP (2011) Gait assessment in
70. Keijsers NL, Horstink MW, van Hilten JJ, Hoff Parkinson’s disease patients through a network
JI, Gielen CC (2000) Detection and assess- of wearable accelerometers in unsupervised
ment of the severity of levodopa-induced dys- environments. In Proceedings of the 33rd
kinesia in patients with Parkinson’s disease by Annual International Conference of the IEEE
neural networks. Mov Disord 15:1104–1111 EMBS, pp 2233–2236
71. Gurevich TY, Shabtai H, Korczyn AD, Simon 79. www.apple.com
ES, Giladi N (2006) Effect of rivastigmine on 80. Kostikis N, Hristu-Varsakelis D, Arnaoutoglou
tremor in patients with Parkinson’s disease and M, Kotsavasiloglou C, Baloyiannis S (2011)
dementia. Mov Disord 21:1663–1666 Towards remote evaluation of movement dis-
72. Schrag A, Schelosky L, Scholz U, Poewe W orders via smartphones. In Proceedings of the
(1999) Reduction of Parkinsonian signs in 33rd Annual International Conference of the
patients with Parkinson’s disease by dopami- IEEE EMBS, pp 5240–5243
nergic versus anticholinergic single-dose chal- 81. LeMoyne R, Mastroianni T, Grundfest W
lenges. Mov Disord 14:252–255 (2013) Wireless accelerometer configuration
73. Weiss A, Sharifi S, Plotnik M, van Vugt JP, for monitoring Parkinson’s disease hand
Giladi N, Hausdorff JM (2011) Toward auto- tremor. Adv Parkinson Dis 2:62–67
Chapter 24
Abstract
Tremor is the most common movement disorder. However; characterizing it in large populations is not easily
accomplished since current methodologies are not adapted to large-scale field studies. To overcome this chal-
lenge, a smartphone application was developed as a stand-alone platform to assess tremor. The current book
chapter details the steps taken to validate this mobile application. Data recorded with the smartphone was
analyzed online and offline as well as compared to laboratory equipment and a clinical scale. This allowed for
the identification of the tremor properties that could reliably be characterized with the smartphone as well as
the limits of the hardware. It also allowed for the identification of tasks that could be performed with the
smartphone when tremor was being assessed. Finally, we confirmed the clinical relevance of the results
provided by the smartphone application.
Key words Recording, Assessment, Monitoring, Parkinson, Essential tremor, Multiple sclerosis,
Mobile, Health, Stand-alone, Telephone
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_24, © Springer Science+Business Media New York 2015
359
360 Benoit Carignan et al.
2 Materials
3 Methods
3.1 Getting Access 1. Next are general guidelines to get access to the smartphone’s
to the Smartphone’s hardware and software. Specific codes and functions differ
Hardware between platforms such as Blackberry®, iPhone®, Android™,
and Software or Windows Phone® as well as between software versions such
as Blackberry® 7 and Blackberry® 10 which would make it
futile for us to provide the code to access the hardware and
Measuring Tremor with a Smartphone 361
3.2 Selecting Tasks 1. Tremor can be observed during rest, holding a limb against
gravity (postural), during movement (kinetic or action), and
during precision tasks (intention). If an application is aimed at
patients with Parkinson’s disease, the evaluation of tremor may
be directed towards a resting position as it is the predominant
type of tremor observed in this disease. On the other hand,
patients with Essential tremor mainly exhibit postural and
kinetic tremor. As such, the tasks selected must have clinical
significance and the application must be validated for each task
for which it may be used to assess tremor (see Note 8). Since
our application was not targeted at a specific patient popula-
tion but rather at any pathological tremor, we opted to have
patients perform rest, postural, kinetic, and intention tasks. Of
course, this meant that we had to validate our application for
all the proposed tasks (see below).
3.3 Creating 1. During the first steps of validation of the smartphone, the user
the Interface interface should be as simple as possible to facilitate and expe-
dite testing. Then, depending on the results, testing procedures
may be changed to add complexity and bring the application
closer to a finished product. Here is a flowchart (Fig. 1) of a
simple interface that could be used for validation.
(a) Participant identification was entered in the smartphone
application using the tactile keyboard on the screen. We
opted for a numerical identification system to preserve par-
ticipant anonymity and coded for a three-digit number as
we knew we were testing less than 1,000 participants. We
also entered the trial number next to the patient identifica-
tion number. We coded for a 2 digit number as we knew
each participant would perform less than 100 trials. As
such, each trial was identified by the participant identifica-
Measuring Tremor with a Smartphone 363
3.4 Tremor 1. Next are some specific points that we want to emphasize when
Assessment using laboratory equipment to validate a smartphone applica-
with Laboratory tion since an extensive body of literature already exists on how
Equipment to use this type of equipment for tremor assessment.
(a) The laboratory accelerometer needs to be fixed to the smart-
phone, preferably to the back where it will not interfere with
the interface. Attention must be paid to align the axes of both
accelerometers in order to facilitate the interpretation of the
results. Since we used a uniaxial accelerometer, we aligned the
axis of the laboratory accelerometer with the axis of the
smartphone’s accelerometer that we thought would experi-
ence the tremor with the highest amplitude.
(b) The gain of the laboratory accelerometer should be set in
accordance with the type of tremor being recorded. Since we
recorded tremor ranging from less than 1 mm to more than
several centimeters, we adjusted the gain for each participant
and recorded it to take it into consideration during analysis.
(c) The length of the recording should be longer than the
smartphone recording time. Indeed, the laboratory record-
ing should be started before the smartphone recording
and should end after it in order to capture the entire smart-
phone trial. We started the recording of the laboratory
accelerometer approximately 2 s before pressing on the
start button of the smartphone application and the record-
ing ended approximately 2–3 s after the smartphone appli-
cation had stopped recording. As such, the length of the
recording of the smartphone application was 10 s while it
was 15 s for the laboratory accelerometer.
(d) Files containing data of each trial were identified in accor-
dance with files recorded on the smartphone SD card; using
the participant identification and trial numbers. As such,
we could easily match the trials that were recorded
simultaneously.
3.5 Time Series 1. Since both systems cannot be started exactly at the same time,
Analysis ( see Note 15) the time series needed to be synchronized. We opted to use the
vibration produced by the smartphone at the end of each trial
to synchronize the recording systems. The vibration frequency
produced by the Blackberry® Storm™ 9530 was about 140 Hz.
Then, after removing the signal below 100 Hz (which largely
removed human movement), it was easy to locate the begin-
ning of the vibration on the laboratory accelerometer. Once
Measuring Tremor with a Smartphone 365
the end of the trial was located, the beginning was found using
the length of the recording. Note that the sampling rate of the
laboratory accelerometer needs to be set adequately in order to
record the vibrations produced by the smartphone.
2. Spatial analysis.
(a) The following analyses were performed on the time series
recorded by the accelerometer of both recording systems.
The mean of the signal was subtracted from each time series
to remove the earth’s acceleration component (see Note 16).
The triaxial accelerometer of the smartphone provided
three different time series (x, y, and z). These time series
were analyzed separately. They could have also been ana-
lyzed together by calculating the resultant of the three axes.
(b) Tremor amplitude: A root mean square was performed on
the time series. First, each data point of the time series was
squared. Then, the mean of all squared values was calcu-
lated. Finally, the square root of this mean was calculated.
(c) Tremor regularity: First, the signal needed to be normalized,
i.e., each value of the time series was divided by the standard
deviation of the entire time series (note that the signal aver-
age was previously set to zero). Second, the time series was
separated into 1 s epoch. The amplitude of each epoch was
calculated using a root mean square (see above). Finally, the
standard deviation of the amplitude of each epoch was con-
sidered as the measure of regularity of the signal.
3. Spectral analysis.
(a) The following analyses were performed on the power spec-
trum of the signal recorded by the accelerometer of both
systems. The power spectrum was obtained using a fast-
Fourier transform method as it is the adequate method for
this type of signal [21]. Note that values under 1 Hz and
over 20 Hz were set to zero in order to remove non-
tremor related movement such as drift [11].
(b) Tremor power distribution: This represents the percentage
of power contained within a selected frequency band. We
chose the 3–7 Hz frequency band due to its clinical rele-
vance (i.e., it is where most of the power lies when examin-
ing pathological tremors such as in Parkinson’s disease and
Essential tremor). Power distribution was calculated by
dividing the area under the curve within the selected band
by the area under the entire curve.
(c) Tremor median power frequency (MPF): This corresponds
to the frequency where half of total power (area under the
curve) was located on each side. It was calculated by initi-
ating an incremental loop where the area under the curve
before a given index was compared to the area under the
curve above.
366 Benoit Carignan et al.
3.6 Validation 1. The first step of the validation was to assess the adequacy of the
code for the online analyses specifically written for the smart-
phone. To do this, tremor characteristics calculated online with
the smartphone were compared with results obtained using
traditional analyses performed offline on the signal recorded by
the smartphone. Hundreds of trials of simulated tremor were
performed with a lot of variability between them (amplitude
and frequency) and in all tasks that were to be validated. One
of the experimenter produced the simulated tremor as we felt
that this would make the validation more ecological than hav-
ing a device produce different types of oscillations. The statisti-
cal approach taken to compare the results was to perform a
Pearson correlation, Bland–Altman plot and concordance cor-
relation coefficient. After this, characteristics reaching the
threshold of acceptance (set a priori) were used in the next step
of validation. Correlation between results were expected to be
very high (r > 0.95, [20]) since any error would come only
from mathematical or programming issues and not from reso-
lution or protocol issues. Characteristics that did not reach the
threshold of acceptance were ignored in the following steps.
Of course, it may have also been possible to improve their
respective programming. Nonetheless, the results obtained
during this step made us put aside tremor power peak as it did
not meet our threshold [20].
2. The second step of the validation process was to assess the capac-
ity of the smartphone to reproduce results obtained with the
already validated laboratory devices. Therefore, results com-
puted online with the smartphone were compared with those
arising from the laboratory equipment. Figure 2 shows examples
of tremor of different amplitude recorded with the smartphone
Fig. 2 Example of tremor traces recorded with the smartphone and the accelerometer with their corresponding
power spectrum. Top pane: example of a moderate amplitude tremor. Middle pane: example of a high ampli-
tude tremor. Bottom pane: example of a low amplitude tremor (physiological tremor). (a) Example of a tremor
trace recorded with the smartphone, (b) example of the tremor trace from the same trial as in (a) but recorded
with the accelerometer, (c) power spectrum of the tremor trace recorded with the smartphone which was
calculated with the algorithms implemented within the smartphone, (d) power spectrum of the tremor trace
recorded with the smartphone which was calculated offline using our laboratory software, (e) power spectrum
of the tremor trace recorded with the accelerometer which was calculated offline using our laboratory soft-
ware. Figure as originally published in Daneault, J. F., B. Carignan, C.É. Codère, A.F. Sadikot, and C. Duval
(2012). “Using a smart phone as a standalone platform for detection and monitoring of pathological tremors.”
Front Hum Neurosci 6: 357. doi: 10.3389/fnhum.2012.00357
368 Benoit Carignan et al.
4 Notes
References
1. Bain P, Brin M, Deuschl G, Elble R, Jankovic J, 7. Iwadate Y, Saeki N, Namba H, Odaki M, Oka
Findley L, Koller WC, Pahwa R (2000) Criteria N, Yamaura A (1989) Post-traumatic intention
for the diagnosis of essential tremor. Neurology tremor–clinical features and CT findings.
54(11 Suppl 4):S7 Neurosurg Rev 12(Suppl 1):500–507
2. Louis ED (2000) Essential tremor. Arch 8. Morrison S, Mills P, Barrett R (2006)
Neurol 57(10):1522–1524 Differences in multiple segment tremor dynam-
3. Jankovic J (2008) Parkinson’s disease: clinical ics between young and elderly persons. J
features and diagnosis. J Neurol Neurosurg Gerontol A Biol Sci Med Sci 61(9):982–990
Psychiatry 79(4):368–376 9. Morrison S, Newell KM (2000) Postural and
4. Alusi SH, Worthington J, Glickman S, Bain PG resting tremor in the upper limb. Clin
(2001) A study of tremor in multiple sclerosis. Neurophysiol 111(4):651–663
Brain 124(Pt 4):720–730 10. Carignan B, Daneault JF, Duval C (2009)
5. Pittock SJ, McClelland RL, Mayr WT, The amplitude of physiological tremor can
Rodriguez M, Matsumoto JY (2004) be voluntarily modulated. Exp Brain Res
Prevalence of tremor in multiple sclerosis 194(2):309–316
and associated disability in the Olmsted 11. Carignan B, Daneault JF, Duval C (2010)
County population. Mov Disord 19(12): Quantifying the importance of high frequency
1482–1485 components on the amplitude of physiological
6. Andrew J, Fowler CJ, Harrison MJ (1982) tremor. Exp Brain Res 202(2):299–306
Tremor after head injury and its treatment by 12. Carignan B, Daneault JF, Duval C (2011) The
stereotaxic surgery. J Neurol Neurosurg organization of upper limb physiological
Psychiatry 45(9):815–819 tremor. Eur J Appl Physiol 112:1269
374 Benoit Carignan et al.
13. Daneault JF, Carignan B, Duval C (2010) New actigraph for long-term tremor record-
Bilateral effect of a unilateral voluntary modula- ing. Mov Disord 21(8):1136–1143
tion of physiological tremor. Clin Neurophysiol 18. (2010) Mobile computing platforms such as
121(5):734–743 the iPhone are beginning to make inroads into
14. Daneault JF, Carignan B, Duval C (2011) the laboratory-serious prospect or fairy tale?
Finger tremor can be voluntarily reduced Nat Methods 7(2): 87
during a tracking task. Brain Res 1370: 19. Wolf JA, Moreau JF, Akilov O, Patton T, English
164–174 JC, Ho J, Ferris LK (2013) Diagnostic inaccu-
15. Daneault JF, Carignan B, Rahimi F, Sadikot racy of smartphone applications for melanoma
AF, Duval C (2013) Postural tremors. In: detection. JAMA Dermatol 149(4):422–426
Grimaldi G, Manto M (eds) Mechanisms and 20. Daneault JF, Carignan B, Codere CE, Sadikot
emerging therapies in tremor disorders. AF, Duval C (2012) Using a smart phone as a
Springer, New York, NY, pp 133–150 standalone platform for detection and moni-
16. Yang MH, Sheu YH, Shih YH, Young MS toring of pathological tremors. Front Hum
(2003) Portable tremor monitor system for Neurosci 6:357
real-time full-wave monitoring and analysis. 21. McNames J (2013) Signal processing. In:
Rev Sci Instrum 74(3):1303–1309 Grimaldi G, Manto M (eds) Mechanisms and
17. Van Someren EJ, Pticek MD, Speelman JD, emerging therapies in tremor disorders.
Schuurman PR, Esselink R, Swaab DF (2006) Springer, New York, NY, pp 371–389
Chapter 25
Abstract
The purpose of this chapter is to introduce novice and intermediate EEG researchers to a convenient and
user-friendly EEG system from NeuroSky, Inc. In our recent study we were interested in changes in the
frontal cortical EEG activity of healthy adults as a function of accommodative stress during performance
of a sustained attention task. We used a commercially available low-cost wireless EEG device from NeuroSky
(MindSet), which has a single active Fp1 dry electrode capable of recording research-grade EEG coupled
with powerful noise-filtering and data software support. The convenience and ease-of-use of MindSet is
further enhanced with validated eSense meters of Attention and Meditation. In this chapter we also pro-
vide additional data analytic support for EEG power spectrum using SPSS syntax commonly used in many
biobehavioral sciences.
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_25, © Springer Science+Business Media New York 2015
375
376 Dmitri V. Poltavski
Aplha waves
Beta waves
Gama waves
Delta waves
Theta waves
Dorsolateral Prefrontal
Cortex
Orbitofrontal
The rationale for the study was the finding that many of the adverse
symptoms expressed by individuals with accommodative and ver-
gence problems of the eyes have also been reported for individuals
diagnosed with attention problems [8, 9]. Granet et al. [9] went
further to suggest that medications used to treat ADHD may actu-
ally aggravate certain oculomotor conditions such as convergence
insufficiency without improving symptoms of inattentiveness. The
function of stimulant medication according to the neuropharma-
cological model of ADHD is to decrease the “noise” in the PFC
due to insufficient dopamine (DA) availability by increasing extra-
cellular DA in this region [10], which in behavioral studies is asso-
ciated with improved attentiveness [11] and in EEG studies with
frontal attenuation of power in EEG bands linked to inattention
such as theta and alpha [12]. In our study we hypothesized that
the disruption of visual processing alone may be sufficient to
account for symptoms of inattentiveness in the absence of changes
in the frontal cortical activity. For our study we used a commer-
cially available low-cost wireless EEG device from NeuroSky, which
is a single, dry-electrode EEG system (MindSet) with built-in noise
filtering and data software support.
2 Materials
Fig. 4 Twenty-one electrode locations of the International 10–20 system for EEG
(electroencephalography) recording
380 Dmitri V. Poltavski
2.2 Data Analytic In our study we used SPSS 19.0 (also known as IBM SPSS since
Software 2009) statistical package that is among the most widely used pro-
grams for statistical analysis in social and behavioral sciences. More
recent SPSS base versions (latest version 22.0) and add-on mod-
ules provide a fairly versatile array of tools for inferential statistical
analyses. In addition to statistical analysis, data management of
The Use of Single-Electrode Wireless EEG in Biobehavioral Investigations 381
3 Methods
3.1 Settings Support software for the MindSet/ThinkGear comes as part of the
NeuroSky MindSet Research Tools (MRT) in two varieties:
1 Included with the MindSet is the NeuroView software, which
makes it easy to connect, graph, view, and record MindSet data
in real-time. NeuroView is designed to be appropriate for nov-
ice to intermediate EEG researchers wishing to view and record
EEG data in real-time. The recorded data can be easily exported
to other third-party applications (e.g., Excel, SPSS) for down-
stream data analysis and processing as it is saved in the comma-
separated value (csv) file format.
2 MRT also includes the more advanced NeuroSkyLab MATLAB
module, which adds the ability to define custom MATLAB
scripts and functions for customized processing and analysis of
MindSet data. It is targeted at the more advanced EEG
researcher who is familiar with the MATLAB environment.
For those comfortable with MATLAB scripting, NeuroSkyLab
provides much more powerful capabilities than NeuroView in
terms of customization, real-time data viewing, and real-time
analysis.
In our 2012 study [7] we used the MindView software and
performed all subsequent scripting and analysis in SPSS 19.0. This
was a preferred method of data processing as we had multiple physi-
ological and behavioral recording channels. The use of SPSS allowed
us to aggregate all subject data in one file for multivariate analyses.
Once the Neuroview is opened, the Bluetooth connection
with the ThinkGear headset is established and the headset is placed
on the subject, the status bar at the bottom of the Neuroview win-
dow will display a value for the signal quality. A Poor Signal Quality
value of 0 indicates a relatively clean signal, while higher values
indicate progressively poorer signals. A value of 200 has a special
meaning, namely, that the electrodes of the ThinkGear module/
382 Dmitri V. Poltavski
headset are not contacting the skin of the head at all. Poor signal
may be caused by a number of different things. In order of severity,
they are:
(a) Sensor, ground, or reference electrodes not being on a per-
son’s head (i.e., when nobody is wearing the ThinkGear).
(b) Poor contact of the sensor, ground, or reference electrodes to
a person’s skin (i.e., hair in the way, or headset which does not
properly fit a person’s head, or headset not properly placed on
the head).
(c) Excessive motion of the wearer (i.e., moving head or body
excessively, jostling the headset).
(d) Excessive environmental electrostatic noise (some environ-
ments have strong electric signals or static electricity buildup
in the person wearing the sensor).
(e) Excessive non-EEG biometric noise (i.e., EMG, EKG/ECG,
EOG, etc.)
To minimize interference with the signal it is thus recom-
mended to, first of all, ensure a good contact of dry electrodes with
the person’s earlobe and frontal (forehead) area by
(a) Wiping out contact areas of the electrodes and the subject’s
head with an alcohol wipe.
(b) Removing all hair and metal objects (e.g., ear rings) from the
target site.
(c) Making sure that the adjustable head-band is properly sized: snug
over the head without being too loose or too tight, and is posi-
tioned over the central line of the head (C3, Cz, C4; see Fig. 4).
It is also recommended to allow the subject to sit quietly and
relatively motionless before a good EEG signal is obtained. Thirdly,
acquisition of the initial EEG signal of good quality (i.e., value of
zero on the meter) may be improved by turning off all other elec-
tronic equipment not necessary for signal acquisition.
The optimal setting for recording is obtained when the “poor
signal” gauge at the bottom of the screen indicates “0” and the
e-sense meters show activity (when the signal quality is “200,” the
e-sense meters do not move. At the same time the raw EEG graph
may be showing fluctuations due to a mixture of extraneous noise
and EEG activity).
3.2 Recording Once acceptable signal quality is obtained (see Note 1) and EEG
recording is started (by pressing on the green “Start” button),
NeuroView has the capability of displaying ongoing EEG activity
in four different modes (by selecting each mode through the
“View” command): as raw EEG, power spectrum (μV2), and in
terms of eSense™ measures of attention and meditation (see Fig. 5
for an example).
The Use of Single-Electrode Wireless EEG in Biobehavioral Investigations 383
3.3 Analysis Once each subject’s CSV power spectrum file was imported into
SPSS, mean EEG band data was derived for each subject using the
“Descriptives” command in SPSS applied to the power spectrum
over the entire duration of each session (baseline and stress in our
The Use of Single-Electrode Wireless EEG in Biobehavioral Investigations 385
study). Mean values for each EEG power band were then entered
as columns into a combined subjects’ file, where each row repre-
sented one subject. The raw power spectrum file contains fre-
quency bins with increments of 0.25 Hz. Thus before we calculated
mean values for each subject in each power band of interest (alpha,
beta and theta), we combined frequency bins to represent a specific
window of interest using the following SPSS syntax (see Note 3):
*Computing power for frequencies of the THETA wave EEG band.
COMPUTE theta_4_7HZ=(@4 + @4.25 + @4.5 + @4.75 + @5 +
@5.25 + @5.5 + @5.75 + @6 + @6.25 + @6.5 + @6.75 + @7 +
@7.25 + @7.5 + @7.75) / 16.
VARIABLE LABELS theta_4_7HZ 'mean power within the theta
EEG band'.
EXECUTE.
*Computing power for frequencies of the Alpha wave EEG band.
COMPUTE alpha_8_12HZ=(@8 + @8.25 + @8.5 + @8.75 + @9 +
@9.25 + @9.5 + @9.75 + @10 + @10.25 + @10.5 + @10.75 + @11
+ @11.25 + @11.5 + @11.75) / 16.
VARIABLE LABELS alpha_8_12HZ 'mean power within the
alpha EEG band band'.
EXECUTE.
*Computing power for frequencies of the Beta wave EEG band.
COMPUTE beta_12_30HZ=(@12 + @12.25 + @12.5 + @12.75 +
@13 + @13.25 + @13.5 + @13.75 +@14 + @14.25 + @14.5 +
@14.75 + @15 + @15.25 + @15.5 + @15.75 + @16 + @16.25 +
@16.5 + @16.75 + @17 + @17.25 + @17.5 + @17.75 + @18 +
@18.25 + @18.5 + @18.75 + @19 + @19.25 + @19.5 + @19.75 +
@20 + @20.25 + @20.5 + @20.75 +@21 + @21.25 + @21.5 +
@21.75 + @22 + @22.25 + @22.5 + @22.75+@23 + @23.25 +
@23.5 + @23.75 + @24 + @24.25 + @24.5 + @24.75 + @25 +
@25.25 + @25.5 + @25.75 + @26 + @26.25 + @26.5 + @26.75 +
@27 + @27.25 + @27.5 + @27.75 + @28 + @28.25 + @28.5 +
@28.75+ @29 + @29.25 + @29.5 + @29.75) / 72.
VARIABLE LABELS beta_12_30HZ 'mean power within the
beta EEG band'.
EXECUTE.
*Computing amplitude for frequencies of the Gamma wave EEG
band.
COMPUTE gamma_30_60HZ=(@30+ @31 + @32 + @33 + @34
+ @35 + @36 + @37 + @38 + @39 + @40 + @41 + @42 + @43 + @44
+ @45 + @46 + @47 + @48 + @49 + @50 + @51 @52 + @53 + @54
+ @55 + @56 + @57 + @58 + @59+ @60 + @30.25+ @31.25 +
@32.25 + @33.25 + @34.25 + @35.25 + @36.25 + @37.25 [email protected]
+ @39.25 + @40.25 + @41.25 + @42.25 + @43.25 + @44.25 +
@45.25 + @46.25 + @47.25 + @48.25 + @49.25 + @50.25 +
386 Dmitri V. Poltavski
3.00
Ln power mV2
2.00
1.00
0.00
−1.00
−5.74 −5.23 −4.60 −4.60 −4.41 −4.18 −3.55 −3.37 −3.28 −2.38
accommodative lag (D) in the stress condition
Fig. 6 Relationship between frontal EEG bands and accommodative lag during completion of Conners CPT
in the stress condition
3.4 Results Consistent with our research hypothesis, the results of our study
showed that performance on a computerized test of sustained
attention (Conner’s CPT) can be compromised by adding a −2.00
D accommodative stimulus to the normal −2.50 D accommodative-
vergence stimulus demand. This increased −4.50D accommodative
demand resulted in a significantly larger accommodative lag and
significantly poorer performance on the CPT (slower reaction
time, greater standard error of hit reaction time, greater response
variability, poorer stimulus detectability, greater number of perse-
verations, and higher overall probability of clinical classification) in
the absence of any appreciable change in frontal lobe electrophysi-
ological activity. There was similarly no significant correlation
between the power of individual EEG bands and accommodative
lag. Figure 6 shows a fairly flat pattern of fluctuations of the three
bands across the range of accommodative lag responses. What was
particularly interesting was the finding that accommodative lag
alone could account for a significant proportion of cases (about
40 %) with a higher probability of clinical classification on the
Conner’s CPT (higher likelihood of ADHD symptoms) even after
controlling for individual band EEG activity. The results have
direct implications for management of drug-resistant forms of
ADHD by suggesting importance of bottom-up processes in sus-
tained attention.
388 Dmitri V. Poltavski
4 Notes
References
Abstract
The diagnosis of sleep disorders, highly prevalent in Western countries, typically involves sophisticated
procedures and equipment that are highly intrusive to the patient. The high processing capabilities and
storage capacity of current portable devices, together with a big range of available sensors, many of them
with wireless capabilities, create new opportunities and change the paradigms in sleep studies. In this
work, a smartphone based sleep monitoring system is presented along with the details of the hardware,
software and algorithm implementation. The aim of this system is to provide a way for subjects, with no
pre-diagnosed sleep disorders, to monitor their sleep habits, and on the initial screening of abnormal
sleep patterns.
Key words Smartphone, Sleep parameters, Home monitors, ECG, Tachogram, Accelerometer, Sleep
Diary, Dream Diary
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_26, © Springer Science+Business Media New York 2015
391
392 Alexandre Domingues
2 Materials
3 Methods
Anatomy of a Bitalino
Micro-Controller Unit
up to 1000Hz sampling rate
6 analog inputs (4@10-bit + 2@6-bit)
4 digital inputs, and 4 digital outputs
Electromyography (EMG) Electrocardiography ECG
measures the electrical activity tracks the electrical changes
produced by your muscles associated with your heartbeat
Light (LUX)
know the changes in ambient
light or other light sources
Bluetooth
a widespread standard
for short-range wireless connectivity Power
so that you can operate
your creations
Fig. 1 BITalino is a customizable biosignal acquisition system, based on the Arduino hardware, currently being
used by several researchers due to its simplicity and reliability. The standard board contains several amplifying
and signal accommodation components, allowing the acquisition of: Electromyography, Electrodermal Dermal
Activity, Light Intensity, Electrocardiography, and Accelerometry. The board also included an LED, a program-
mable Micro-controller Unit, Bluetooth Unit, and a Power Stage. The different components can be included in
the system according to the specific requirements of the intended application
394 Alexandre Domingues
Fig. 2 The Sleep Monitor uses six sections of the BITalino board: The Micro-
controller Unit, the Bluetooth, the Power Unit, the LED, the Accelerometer, and the
Electrocardiography Unit. The six sections were connected and configured in a
compact format
3.2 Sleep Parameter The sleep parameter estimation method, displayed in Fig. 4, is
Estimation Software composed by the preprocessing and feature extraction procedures,
and Algorithm followed by the classification stage, designed to reject the ambigu-
ous features, and regularization.
The multimodal dataset includes the ECG, from which the RR
signal is computed, and Accelerometer (ACT) data. The prepro-
cessing operations are required to reduce the movement artifacts,
normalize the data across different patients, and prepare it for fea-
ture extraction.
ECG filtering and QRS complex detection is performed
according to the methods described in [4], the tachogram is then
constructed from the detected R peaks, and downsampled to 2 Hz.
Magnitude normalization and DC component removal are applied
to both RR and ACT data according to:
a (n) − µ (n)
ă ( n ) =
σ (n)
Smartphone Based Monitoring System for Long-Term Sleep Assessment 395
Fig. 3 Precise placement of the three electrodes is not fundamental but should
be in the area shown in the figure
where a(n) is the original sample, μ(n) and σ(n) are the mean and
standard deviation of the data within the 5 min window centered
at the nth sample and ă ( n ) is the normalized sample.
After preprocessing, each dataset is divided in contiguous
epochs of T = 30 s.
Let wj = [RRj; ACTj] represent a T dimensional window, con-
taining the multimodal data from the jth epoch, where j ∈ [1, …, M]
and M the total number of epochs.
The extracted features and the extraction procedures are the
following:
1. RR features: The RR frequency domain features are computed
in the LF = [0.015 … 0.15]Hz and HF = [0.15 … 0.4]Hz
bands [3]. In order to extract these features from each RRj an
eight-order autoregressive model (AR) is fitted to the extended
window RRj* = [RRj − 3, …, RRj] (see Note 4) and a set of opti-
mal coefficients aRR and a residual, ERR, are obtained. The
power spectrum is computed from the estimated AR coeffi-
cients and the following features are extracted:
PMj: Magnitude of the high frequency pole of the filter impul-
sive response filter (IIR) described by the coefficients aRR.
PPj: Phase of the high frequency pole.
ERRj: Residual of the AR model fitted to RRj*.
TPj: Total power (LF + HF).
HFj: Power in the HF range.
LFj: Power on the LF range.
LF/HFj: Power ratio between the two frequency bands.
MHRj: Mean heart rate on the considered RRj.
396 Alexandre Domingues
Fig. 4 After the acquisition of RR and ACT data (5-min segment shown on top of
the image) the sleep parameter estimation algorithm consists of (1) Preprocessing,
where the noise is filtered and the signal normalized, (2) Feature extraction,
where the information regarding the states of interest is extracted, (3)
Classification by two binary classifiers, having a rejection option for outliers and/
or ambiguous samples, and (4) Regularization allowing a precise computation of
the parameters by taking into account the number of rejected samples and the
accuracy of the classifiers. The acquired signals consist of: RR: R-to-R interval
tachogram, computed from the ECG and ACT—Accelerometer data, a measure-
ment of the user’s activity
Fig. 5 The figure shows the 2D scatter plot of two features (Mean Heart Rate and LF/HF ratio) and the Support
Vector Classifier, trained with these two features, together with the rejection areas. The figure illustrates a
binary classification problem, where each sample is classified as Sleep, Wakefulness, or Rejected. The shown
data corresponds to real data, acquired with the described system, but for illustration purposes the description
of each sample is limited to two features: Mean Heart Rate and LF/HF ratio. Every sample falling in the area
above the Wakefulness rejection boundary will be classified as Wakefulness; every sample falling below the
Sleep rejection boundary will be classified as Sleep. Samples falling between the two boundaries will be
rejected. The “+” and “*” markers shown the true class of each sample. All the samples inside the rejection
areas are considered ambiguous from a classification point of view and thus discarded. The plotted data cor-
responds to real data, acquired with the described system
correspond to the mean heart rate and the LF/HF ratio during
Sleep and Wakefulness states.
An accurate computation of the sleep parameters implies a reli-
able estimation of the number of epochs (samples) during each sleep
state (Wakefulness/Sleep; REM/Non-REM). Since the output of
the classifiers is affected by an error and since a certain number of
samples are rejected during this classification, a regularization is
required to properly estimate the number of samples on each state.
The final step in the computation of the sleep parameters is
thus the regularization of the classifier output. The regularization
operation corrects the estimated number of Sleep, Wakefulness,
REM and Non-REM epochs by taking into account the p ercentage
of mis-classified samples and an estimation of the number of sam-
ples that were rejected by the classifiers on each class.
Consider a binary classifier C, with a reject option that maps
each sample into one of three labels l ∈ {p, n, r}where p, n and r
denote Positive, Negative and Reject.
Smartphone Based Monitoring System for Long-Term Sleep Assessment 399
with Tp, Fn, Fp, Tn, Rp and Rn the True Positives, False Negatives,
False Positives, True Negatives, Rejected Positives and Rejected
Negatives respectively.
The positive (θp,i) and negative (θn,i) correction factors and the
fraction of rejected samples per class (ωp,i and ωn,i) are computed
for each training dataset as
Tpi + Fpi
θ p,i =
Tpi + Fn i
Fn + Tn i
θ n ,i = i
Fpi + Tn i
Rpi
ωp,i =
Rpi + Rn i
Rn i
ωn ,i =
Rpi + Rn i
With i ∈ [1, …, M] and M the number of training sets.
Let N(.) represent a counting operator, the number of pre-
dicted samples in each class can be corrected taking into account
the performance of the classifier as
N ( p)
N ( p˘ ) =
θp
And estimating the number of rejected samples from each class as
N ( rp ) = ωp N ( r )
The expressions used to calculate the Sleep parameters finally
become:
N ( s˘ )
SE =
N ( s ) + N ( w)
NREM p =
( )
+ N (r )
N ns ns
( N ( ns ) + N ( rs ) + N ( r ) ) × SE
400 Alexandre Domingues
REM p =
( )
+ N (r )
N rs ns
( N ( ns ) + N ( rs ) + N ( r ) ) × SE
with SE computed from the output of the SW classifier and NREMp
and REMp from the RN classifier. The three parameters are pre-
sented to the user in percentage.
The described system and sleep parameter estimation method
was validated with a group of 25 healthy volunteers. Each volun-
teer performed a standard PSG, supervised by a trained technician.
The Hypnogram was used as the ground truth, to properly identify
the correct state on each epoch. The sleep parameter estimation
algorithm was tested using a “Leve-One-Patient-Out” policy, i.e.,
the computation of the regularization parameters and the training
of the classifiers was performed considering all the datasets, except
one, which was then used to test the algorithm. This process was
repeated for all the subjects. This means that the classification and
computation of sleep parameters is always performed on data pre-
viously unseen by the algorithm.
The described method achieves an average estimation error of
approximately 4, 10, and 5 % in the estimation of the SE, REM,
and NREM percentages, respectively, when compared with the
values obtained from PSG data.
The estimation error is computed as
true − estimated
error = ×100
true
Where true is the true value of the parameter and estimated the
value computed by the algorithm.
The structure of the sleep monitoring software is shown in
Fig. 6. A new record is started on each night; the system will start
acquiring the ECG and Accelerometer data, and display them in
real-time together with an estimate of the power spectrum of the
RR signal (Heart Rate Variability). After each acquisition, the data
is processed and the sleep parameters computed and stored.
3.3 Sleep The scheme of the Android implementation of the Sleep and
and Dream Diary Dream Diaries are shown in Figs. 7 and 8 The Sleep Diary has a
simple structure, with a main menu from which Events can be cre-
ated, displayed and deleted. The Insert Event menu allows the cre-
ation of a new event, from a list of standard types. Each event and
respective information is stored in a sql database, the implementa-
tion of this database is supported by Android through SQLite, a
software library that implements a self-contained, serverless, zero-
configuration, SQL database engine.
The Dream Diary follows the same structure of the Sleep Diary
although with more complex options. The first option “Record
Smartphone Based Monitoring System for Long-Term Sleep Assessment 401
Fig. 6 Structure of the sleep monitor software. A simple menu allows to create a New Record or retrieve
a previous record. After each record the data is processed and stored in a local database
Fig. 7 Structure of Sleep Electronic Diary (SeD). All the entries are stored in a database, common with the sleep
monitor and DeD
402 Alexandre Domingues
Fig. 8 Structure of Dream Electronic Diary (DeD). All the entries are stored in a database, common with the
sleep monitor and SeD
4 Notes
References
1. Kushida CA et al (2005) Practice parameters 4. Seabra JC et al (2011) Rayleigh mixture model
for the indications for polysomnography and for plaque characterization in intravascular
related procedures: an update for 2005. Sleep ultrasound. IEEE Trans Biomed Eng 58(5):
28(4):499–521 1314–1324
2. Guerreiro J et al (2013) BITalino: a multimodal 5. Lewicke A, Sazonov E, Corwin MJ, Neuman M,
platform for physiological computing. Schuckers S (2008) Sleep versus wake classifica-
Proceedings of the international conference on tion from heart rate variability using computa-
informatics in control, automation and robotics tional intelligence: consideration of rejection in
(ICINCO), Reykjavik, Iceland classification models. IEEE Trans Biomed Eng
3. Clifford GD (2002) Signal processing methods 55(1):108–118
for heart rate variability analysis. Ph.D. disserta-
tion, St Cross College
Chapter 27
Abstract
Mobile technology has recently been introduced for blood pressure measurements or glucose level con-
trols. In surgical disciplines the use of smartphone applications is mostly restricted as training tools or
knowledge resources.
Simple surgical procedures which are performed often in certain disciplines may be performed with
limited accuracy since routine and overwork of medical staff lead to less awareness to possible mistakes. In
these cases simple and effective means are necessary to achieve better patient safety. In this context, a surgi-
cal instrument for ventricular catheter placement in neurosurgical patients was designed which is assisted
by measurements undertaken in a smartphone software application specifically visualizing the use of this
instrument and achieving better accuracy for catheter positioning. On theoretical ground, the angulation
of the catheter trajectory towards the surface of the skull in a coronal reconstructed CT or MR image is
determined as the simplified but the most relevant individual parameter for correct ventricular catheter
placement. Transfer of a CT/MRI image onto the smartphone can be performed via mail as anonymous
file. Using this image, the trajectory measurement can be performed individually in a few steps by calibra-
tion of the image size, definition of the frontal entry point, and virtual placement of the instrument on the
surface of the skull. Then the angulation can be adjusted and measured to place the catheter’s trajectory
towards the ipsilateral ventricle and the catheter length is determined. The parameters are now given by
the app and visualized on the image in order to be applied to the surgical site of the patient.
The tool represents a widely available and cost-effective solution as navigation technique which is
simple to apply in order to achieve better accuracy in ventricular catheter placement for higher safety in a
large cohort of neurosurgical patients.
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_27, © Springer Science+Business Media New York 2015
405
406 Ulrich-W. Thomale
Fig. 1 Schematic drawing of the ventricular anatomy in relation to the brain and skull surface depicted with a
ventricular catheter entered via a frontal (precoronal) paramedian bur hole (a). The possible trajectories are
given in simplified frontal/coronal (b) and longitudinal/sagittal (c) view. The individual measurement of the
trajectory’s angulation in the coronal plane is necessary for correct placement of the ventricular catheter. In
the sagittal plane the insertion angle towards the skull surface is always 90° using a frontal entry point
408 Ulrich-W. Thomale
Fig. 2 The ventricular catheter guide with the guiding tube, and the base which
consists of a semicircular rod with angulation marks and a base. The angulation
can only be adjusted in one plane. The guide must be aligned such that the fixed
angulation of 90° will be applied in sagittal orientation while the individual angu-
lation will be applied in coronal plane
Ventricular Catheter Placed with Smartphone Assistance 409
Fig. 3 Smartphone application software is available in the Apple App Store for iPhone and iPad
2 Materials
Fig. 4 MRI images prepared for transfer to the software app. (a) Coronal image with moderate ventricular
enlargement. (b) Reconstructed image along the catheter trajectory as coronal like section. The trajectory of
the catheter is depicted as solid line towards the ventricle. The bitemporal diameter is measured before trans-
fer in order to warrant image size calibration
Ventricular Catheter Placed with Smartphone Assistance 411
3 Methods
Fig. 5 Workflow for image data transfer to the mobile device, which can be established by mail or with an USB
cable. Image may be given as .png, .jpeg, .bmp, or .tif format
Fig. 6 Measurements as performed in the iPhone app in two representative cases. (a) Angulation of the trajec-
tory (red dotted line) towards the skull surface is 5.8° as deviation from 90° (light blue dotted line) with a tilt
of the guiding tube towards the midline of the patient. Catheter length is 71 mm (green dotted line). (b) In the
trajectory’s inline view reconstruction image the angulation is 6.8° tilt towards midline. The catheter length is
determined with 64 mm (Color figure online)
Fig. 7 Coordinates for the entry point are given as distance from nasion and
distance from midline. While the latter one can be determined in the software
app (usually 2–3 cm), the distance to nasion may be at 11.5 cm
4 Notes
References
1. Carter T, O’Neill S, Johns N et al (2013) 6. Thomale UW, Knitter T, Schaumann A et al
Contemporary vascular smartphone medical (2013) Smartphone-assisted guide for the place-
applications. Ann Vasc Surg 27(6):804–809 ment of ventricular catheters. Childs Nerv Syst
2. Brooke MJ, Thompson BM (2013) Food and 29(1):131–139
Drug Administration regulation of diabetes- 7. Wilson TJ, Stetler WR Jr, Al-Holou WN et al
related mHealth technologies. J Diabetes Sci (2013) Comparison of the accuracy of ven-
Technol 7(2):296–301 tricular catheter placement using freehand
3. Naftel RP, Safiano NA, Falola MI et al (2013) placement, ultrasonic guidance, and stereotac-
Technology preferences among caregivers of tic neuronavigation. J Neurosurg 119(1):
children with hydrocephalus. J Neurosurg 66–70
Pediatr 11(1):26–36 8. O’Neill BR, Velez DA, Braxton EE et al (2008)
4. Jenny JY (2013) Measurement of the knee flex- A survey of ventriculostomy and intracranial
ion angle with a Smartphone-application is pre- pressure monitor placement practices. Surg
cise and accurate. J Arthroplasty 28(5):784–787 Neurol 70(3):268–273
5. Orringer DA, Golby A, Jolesz F (2012) 9. Hayhurst C, Beems T, Jenkinson MD et al (2010)
Neuronavigation in the surgical management of Effect of electromagnetic-navigated shunt place-
brain tumors: current and future trends. Expert ment on failure rates: a prospective multicenter
Rev Med Devices 9(5):491–500 study. J Neurosurg 113(6):1273–1278
Part III
Abstract
Cervical cancer causes 275,000 deaths each year with 85 % of these deaths occurring in the developing
world. One of the primary reasons for the concentration of deaths in developing countries is a lack of effec-
tive screening methods suited for the infrastructure of these countries. In order to address this need, we
have developed a high-resolution microendoscope (HRME). The HRME is a fiber-based fluorescence
microscope with subcellular resolution. Using the vital stain proflavine, we are able to image cell nuclei
in vivo and evaluate metrics such as nuclear-to-cytoplasmic ratio, critical to identifying precancerous epi-
thelial regions. In this chapter, we detail the materials and methods necessary to build this system from
commercially available parts.
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_28, © Springer Science+Business Media New York 2015
421
422 Benjamin D. Grant et al.
2 Materials
2.1 Optical Lenses 1. 10× Plan Achromat Objective: 0.25 NA, 10.6 mm working
and Mirrors distance, infinity corrected (Thorlabs RMS10X).
2. Tube Lens: 1-in. diameter spherical achromatic doublet,
150 mm focal length, 400–700 nm wavelength anti-reflective
coating (Thorlabs AC254-150-A).
3. Condenser Lens: 1-in. diameter aspherical plano-convex con-
denser lens, 17.0 mm focal length, 400–700 nm wavelength
antireflective coating (Newport KPA031-C).
4. Protected Aluminum Mirror: 1-in. diameter, round, protected
aluminum mirror, λ/10 flatness, >90 % reflectivity from 400 to
700 nm (Thorlabs PF10-03-G01).
2.2 Optical Filters 1. Dichroic Mirror: 1-in. diameter, 45° angle of incidence,
485 nm cut-off (Chroma 485dclp).
2. Excitation Filter: 1-in. diameter, thin-film bandpass filter, 452
center wavelength with 45 nm bandwidth (Semrock FF01-
454/45-25).
424 Benjamin D. Grant et al.
2.3 Optomechanics 1. Cage Cube: 30 mm cage cube compatible with Thorlabs’ cage
system (Thorlabs C6W).
2. Blank Cage Cube Cover Plate: cover plate for 30 mm cage
cube (Thorlabs B1C).
3. Fixed Cage Cube Platform: rotating round platform for plac-
ing optics in 30 mm cage cube system (Thorlabs B3C).
4. Cage Cube Optics Mount: optics mount for 1-in. optics com-
patible with the cage cube platform (Thorlabs B5C).
5. End Cap: 1-in. diameter end cap (Thorlabs SM1CP2).
6. Cage Plate: 30 mm square cage plate for 1-in. optics (Thorlabs
CP02).
7. Cage Rods: 6 mm diameter steel rods for Thorlabs 30 mm
cage system, 1.5″ (4× Thorlabs ER1.5), 3.0″ (4× Thorlabs
ER3), 6.0″ (4× Thorlabs ER6).
8. Right-Angle Kinematic Mirror Holder: holds 1″ diameter mir-
ror to redirect light in 30 mm cage systems (KCB1).
9. Lens Tube, 1″: threaded lens tube for 1-in. optics, 1 in. in
length (Thorlabs SM1L10).
10. Lens Tube, 3″: threaded lens tube for 1-in. optics, 3 in. in
length (Thorlabs SM1L30).
11. Focusing Z-Translator: translation mount for focusing in 1 μm
increments (Thorlabs SM1Z).
12. Camera Adapter: SM1 to C-mount adapter to allow c-mount
capable camera to interface with Thorlabs parts (Thorlabs
SM1A9).
13. RM1 Adapter: SM1 to RMS adapter to connect commercial
objective to system (Thorlabs SM1A3).
14. SM1 Coupler: externally threaded coupler for attaching two
internally threaded 30 mm Thorlabs components (Thorlabs
SM1T2 2×).
15. SMA Receptacle: attaches SMA connector to Thorlabs 30 mm
system (Thorlabs SM1SMA).
16. Retaining Rings: holds optical components in lens tubes
(Thorlabs SM1RR).
2.4 Illumination 1. Royal Blue LED: 900 mW royal blue LED with heat sink,
centered at 455 nm (Thorlabs M455L3).
2. High power LED fDriver: 12.0 V constant current driver with
variable current selection from 0 to 1,200 mA (Thorlabs,
LEDD1B).
High-Resolution Microendoscope 425
3 Methods
3.1 Optics Assembly 1. Figure 2 illustrates the assembly of the tube lens, excitation
filter and cage cube. Start by placing the excitation filter [4]
into the 3 in. lens tube [5]. The filter should be placed as far
down into the tube as possible, so that it is coincident with the
beginning of the external threads. The filter should be facing
the male end of the tube [5] where it interfaces with the cage
cube [6] because the light will be entering the filter from this
direction. Next, secure the filter with a 1-in. retaining ring [3].
Now the tube lens [2] should be placed in the lens tube [5]
completely, such that it is flush with the retaining ring [3].
Ensure that the more convex side of the lens faces the male end
[5] of the lens tube, as this is where collimated light will enter
the lens. Secure the lens with a second retaining ring. Finally,
screw the lens tube into the cage cube [6].
2. The assembly of the kinematic mirror mount, the camera and
the lens tube is shown in Fig. 3a. Place the aluminum mirror
[1] in the right-angle kinematic mirror mount [2]. Create four
7.5″ cage rods by screwing each 1.5″ rod [3] into a 6″ rod [4].
Next, screw the 7.5″ cage rod into the four holes on one face
of the kinematic mirror mount [2]. Slide the cage cube [5]
onto the cage rods so that the 3″ lens tube is facing the kine-
matic mirror mount. Next, attach the camera [8] to the kine-
matic mirror mount [2] by using the SM1 coupler [6] and the
C-mount to SM1 camera adapter [7]. At this point, turn the
camera on and the focus the system on an object sufficiently far
away to achieve infinity focus (in order to properly interface
with the infinity-corrected objective). To focus the system,
426 Benjamin D. Grant et al.
Fig. 2 Assembly of primary imaging optics: [1] retaining ring, [2] tube lens, [3]
retaining ring [4] emission filter, [5] lens tube, [6] cage cube
move the cage cube along the cage rods until the image appears
focused on the camera image. Using a 150 mm focal length
lens tube with the Grasshopper camera results in a slight air
gap between the lens tube and the kinematic mirror mount
demonstrated in Fig. 3b.
3. Install the cage cube optics mount [1] on the cage cube plat-
form [2] using the provided bolt. Next, secure the dichroic
mirror [3] into the optics mount [1]. Make sure the dichroic
mirror is facing out of the optics mount as shown in Fig. 4a.
4. Install the cage cube platform [2] onto the cage cube [4].
Rotate the cage cube platform so that the dichroic mirror is
facing at a 45° angle between holes five and six as depicted in
Fig. 4b. Secure the cage cube platform at this angle for the
time being with provided screws. See Note 1 for more
information.
5. Figure 4c demonstrates the installation of the cage cube cover
plate and end caps. Place the blank cage cube cover plate [7]
on the top of the cage cube [4] and cover the rear hole with
the end cap [8]. These help reduce stray light.
6. Figure 5 depicts the installation of the commercial objective
and the SMA adapter to the system. The SMA adapter allows
an SMA-terminated fiber to be integrated directly into the sys-
tem. The fiber screws directly into the SMA adapter and the
SMA adapter screws into the Z-translating stage. Install the
RMS to SM1 adapter [1] on the 10× commercial objective [2].
With the adapter in place, install the objective into the cage
cube [5]. Next, place the SM1 threaded SMA connector [4]
into the Z-translating stage [3]. Attach the Z-stage translator
High-Resolution Microendoscope 427
Fig. 3 Attaching the camera and kinematic mirror mount. (a) Exploded view showing the relative locations of
[1] the aluminum mirror, [2] the kinematic mirror mount, [3] the 1.5″ cage rods, [4] the 6″ cage rods, [5] the
cage cube, [6] the SM1 coupler, [7] the c-mount to SM1 coupler, and [8] the camera. (b) The top view of the
system assembled to this point showing the gap between the kinematic mirror mount and the lens tube
onto the main system by sliding it onto the cage rods. Do not
tighten onto the cage rods until the system is moderately
focused. To focus, install the fiber bundle using the SMA con-
nections. With the camera on, face the distal end of the fiber
directly towards a light. Move the Z-translator manually along
the cage rods until the fiber bundle is relatively focused, then
tighten the Z-translator on the cage rods. Again, while facing
428 Benjamin D. Grant et al.
Fig. 4 Assembly of the dichroic mirror in the cage cube system. (a) Exploded view of [3] the dichroic mirror, [2]
the cage cube platform and [1] the cage cube optic mount. (b) Installation of [2] the cage cube platform onto
[4] the cage cube, illustrating [1] the cage cube optic mount and [3] the dichroic mirror at a 45° angle to open-
ings [5] and [6]. (c) Addition of [7] the blank cover plate and [8] the end cap to block stray light from entering
[4] the cage cube
Fig. 5 An exploded view showing the installation of [2] the objective lens into [5]
the cage cube using an [1] RMS to SM1 adapter and the installation of [3] the
Z-translating stage and [4] the SMA fiber mount
same face of the cage cube assembly [9]. The installation of the
LED assembly to the system is shown in Fig. 6c. Connect the
LED by guiding the LED-cage plate [3] assembly onto the
four 3-in. cage rods [10]. Adjust the position of the LED to
maximize the light intensity exiting the distal end of the fiber
bundle. This can be achieved by aiming the distal end of the
fiber at a power meter while adjusting the LED position.
Secure the LED in this position. Figure 7 shows the entire sys-
tem both with (A) and without (B) major optomechanics.
Fig. 6 Illumination installation. (a) Installation of [1] the LED using an [2] SM1
coupler and [3] the cage plate (b) Exploded view of illumination optics. The [5]
excitation filter is secured into [4] the 1.5″ lens tube using [6] a retaining ring.
Next, [7] a condenser lens is secured using [8] an additional retaining ring
directly behind the filter. [4] The 1.5″ lens tube, now containing the excitation
optics, is screwed into [9] the cage cube system. Finally, [10] the 3-in. cage rods
are installed in each corner of the cage cube (c) The [1–3] LED cage plate
assembly is connected to the system using [10] the cage rods
Fig. 7 Overall Diagram of the HRME. (a) Overall diagram of completed system
with major components labeled: [1] CCD camera, [2] kinematic mirror mount, [3]
3" lens tube, [4] cage cube with cover plate, [5] 10× objective lens, [6]
Z-translating stage, [7] SMA connector, [8] LED. (b) The HRME without major
optomechanics, with major components labeled: [1] CCD Camera, [2] mirror, [3]
emission light path, [4] tube lens, [5] emission filter, [6] dichroic mirror, [7] exci-
tation filter, [8] condenser Lens, [9] excitation light path, [10] LED
the HRME was used to image one control area that appeared nor-
mal by visual inspection. Biopsies were taken from the correspond-
ing areas of the cervix for pathologic diagnosis [12]. Figure 8
provides an example of the HRME’s ability to discriminate between
benign and precancerous acetowhitening. Figure 8a, d are wide-
field images of the cervix after application of acetic acid [12]. The
white arrows in both images indicate suspicious areas based on ace-
towhitening. Figure 8b is an HRME image corresponding to the
suspicious region in Fig. 8a. The HRME image shows normal cel-
lular features: the underlying nuclei are small, evenly spaced and
show low eccentricity. A biopsy of this region was found to be nor-
mal by histopathology and a sample image is provided in
C. Conversely, Fig. 8e, the HRME image from the suspicious
region in D, shows precancerous cellular features. The nuclei are
large, crowded, and irregular, indicative of high grade dysplasia.
Histopathological analysis of the biopsy from this area confirmed
that the underlying disease was high grade cervical interepithelial
neoplasia (CIN 3) [12].
Fig. 8 Comparison of colposcopic images, HRME images, and histologic diagnosis. Both (a) and (d) show col-
poscopic images of cervices with regions undergoing acetowhitening (indicated by white arrows). When the
suspicious lesion in (a) is imaged with the HRME (b), the HRME shows that the nuclei are small, round and
well-spaced. However, when the suspicious lesion (d) is imaged with the HRME (e) the nuclei are ragged,
irregular, and crowded. Histopathologic diagnosis of tissue biopsied from the regions of interest in (a) and (d)
confirm that (a) is a non-neoplastic lesion while that of (d) is high grade dysplasia (CIN3) [12]
High-Resolution Microendoscope 433
Fig. 9 The average N/C ratio versus histopathologic diagnosis for a pilot study
in Botswana [12]
4 Notes
References
1. Jemal A, Bray F, Center MM et al (2011) 8. Pierce MC, Vila PM, Polydorides AD et al
Global cancer statistics. CA Cancer J Clin (2011) Low-cost endomicroscopy in the
61:69–90 esophagus and colon. Am J Gastroenterol
2. Kitchener HC, Castle PE, Cox JT (2006) 106(9):1722–1724
Chapter 7: achievenements and limitations of 9. Pierce MC, Guan Y, Quinn MK et al (2012) A
cervical cytology screening. Vaccine 24S3:63–70 pilot study of low-cost, high-resolution
3. Papanicolaou GN, Traut HF (1941) The microendoscopy as a tool for identifying
diagnostic value of vaginal smears in carci- women with cervical precancer. Cancer Prev
noma of the uterus. Am J Obstet Gynecol Res 5(11):1273–1279
42:193–206 10. Hitchcock CL (2011) The future of telepa-
4. Sankaranarayanan R, Esmy PO, Rajkumar R thology for the developing world. Arch Pathol
et al (2007) Effect of visual screening on cervi- Lab Med 135(2):211–214
cal cancer incidence and mortality in Tamil 11. Suba EJ, Murphy SK, Donelly AD et al (2006)
Nadu, India: a luster-randomised trial. Lancet Systems analysis of real-world obstacles to suc-
370(9585):398–406 cessful cervical cancer prevention in developing
5. Muldoon TJ, Pierce MC, Nida DL et al (2007) countries. Am J Public Health 96(3):480–487
Subcellular-resolution molecular imaging 12. Quinn MK, Bubi TC, Pierce MC et al (2012)
within living tissue by fiber microendoscopy. High-resolution microendoscopy for the
Opt Express 15(25):16413–16423 detection of cervical neoplasia in low-resource
6. Shin D, Pierce MC, Gillenwater AM et al settings. PLoS One 7(9):e44924
(2010) A fiber-optic fluorescence microscope 13. World Health Organization (2012) Prevention
using a consumer-grade digital camera for of cervical cancer through screening using
in vivo cellular imaging. PLoS One 5(6): visual inspection with acetic acid (VIA) and
11218 treatment with cryotherapy. A demonstration
7. Muldoon TJ, Anandasabapathy S, Maru D et al project in six African countries: Malawi,
(2008) High-resolution imaging in Barrett’s Madagascar, Nigeria, Uganda, The United
esophagus: a novel, low-cost endoscopic micro- Republic of Tanzania, and Zambia. WHO
scope. Gastrointest Endosc 68(4):737–744 Document Production Services
Chapter 29
Abstract
This chapter presents the state of the art on mobile teledermoscopy applications, utilizing smartphones
able to store digital images of skin areas depicting regions of interest (lesions) and perform self-assessment
or communicate the captured images with expert physicians. Mobile teledermoscopy systems consist of a
mobile application that can acquire and identify moles in skin images and classify them according their
severity and Cloud infrastructure exploiting computational and storage resources. The chapter presents
some indicative mobile applications for skin lesions assessment and describes a proposed system developed
by our team that can perform skin lesion evaluation both on the phone and on the Cloud, depending on
the network availability.
Key words Image analysis, Skin lesions, Skin cancer, Melanoma, Mobile dermoscopy, Mobile com-
puting, Teledermatology, Cloud infrastructures, Android, iOS, Smartphones
1 Introduction
Skin cancer is among the most frequent types of cancer and one of
the most malignant tumors. Its incidence has increased faster than
that of almost all other cancers and the annual rates have increased on
the order of 3–7 % in fair-skinned populations in recent decades [1].
Currently, between 2 and 3 million non-melanoma skin cancers and
132,000 melanoma skin cancers occur globally each year. Skin cancer
is the most common form of cancer in the USA. More than 3.5
million skin cancers in over two million people are diagnosed annu-
ally One in every three cancers diagnosed is a skin cancer and, accord-
ing to Skin Cancer Foundation Statistics, one in every five Americans
will develop skin cancer in their lifetime [2]. Each year there are
more new cases of skin cancer than the combined incidence of
cancers of the breast, prostate, lung, and colon [3]. Treatment of
non-melanoma skin cancers increased by nearly 77 % between 1992
and 2006 [4]. Over the past three decades, more people have had
skin cancer than all other cancers combined [3].
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_29, © Springer Science+Business Media New York 2015
435
436 Charalampos Doukas et al.
Fig. 2 Illustration of (a) typical melanoma, (b) dysplastic nevus, and (c) non-
dysplastic (common) nevus
L = R2 + G 2 + B2 (3)
B
Angle A = cos −1 (4)
L
R
Angle B = cos −1 (5)
L sin ( Angle A )
Fig. 4 Asymmetry border color features; (a) Asymmetry Test, (b) Border Test, (c) Color variegation (source:
http://www.dermoncology.com)
∑ P (i − µ )
2
σi = i, j i (10)
i , j =0
In ref. [8] we have presented a detailed comparison between
the most common methods and research works for analyzing skin
lesion images. Annotation of skin images is also particularly
important for medical decision support systems since medical
experts have difficulties in understanding the criteria of decision in
many existing systems. A semantic taxonomy and hierarchy of skin
lesion images using ontological approached is also presented in
ref. [21] (Fig. 5).
442 Charalampos Doukas et al.
Fig. 5 Differential structures; (a) Pigmented network, (b) Dots, (c) Brown globules, (d) Branched streaks
(source: http://www.dermoncology.com)
1.1 Doctor Mole This application uses augmented reality technology to scan the
Application skin lesions in real time and extracts instant risk feedback on
Asymmetry, Border, Color, Diameter, and Risk. The photos which
are taken by user are stored in the mobile phone for future refer-
ence and examination of the lesion progress through time.
In addition, it is designed to help in the assessment of moles.
This allows the user to easily take a photo of the desired target
mole accurately with the correct lighting and distance. All the cal-
culations are performed on the phone so as results are provided
instantly. At any point user has the ability to review all photos,
results and make comparisons between mole images captured at
different times.
Below, we present a few screenshots of this application (Fig. 6).
1.2 Mole Detective Mole Detective takes pictures of moles on user’s skin and analyzes
the symptoms of melanoma in order to increase the chance of
detecting skin cancer in early stages. The survival rate of melanoma
is a dismal 15 % at stage four. However, when caught early, the sur-
vival rate is 95 %. Users have the ability to indicate the location of
the mole their body and take pictures of it. Then, users can analyze
Evaluating Skin Lesions Utilizing Smart Phones and Cloud Platforms 443
Fig. 6 (a) Compare saved photos. (b) Results of mole. (c) Preview of the mole. (d) Display risk of mole (source:
https://play.google.com/store/apps/details?id=com.revsoft.doctormole)
444 Charalampos Doukas et al.
Fig. 7 (a) Start the app. (b) Location’s mole. (c) Result of scanned mole. (d) Take a photo of mole (source:
https://play.google.com/store/apps/details?id=com.moledetective)
446 Charalampos Doukas et al.
1.5 SpotMole SpotMole provides a simple way to have a quick check of skin spots
and moles. It takes as input an image of a mole which was loaded
by phone’s gallery or was taken from camera. It may detect signs of
melanoma using image processing and pattern recognition tech-
niques. Moreover, it has embedded an automatic mole analysis
using the device’s camera/gallery and algorithms.
Firstly every user should have to take a close-up and well cen-
tered snapshot of a skin spot or mole and next run an analysis.
SpotMole automatic assessment software uses standard visual anal-
ysis procedures to check spots on the skin. The features extracted
are common in dermatology and widely employed in visual inspec-
tion of the skin spots. These are Asymmetry, Border, Color,
Diameter, and Evolution of the mole (ABCDE). Furthermore, this
application provides extra settings, which each user has the chance
to change the parameters of image processing and list with the
doctors who are closer to your location (Fig. 10).
Fig. 9 (a) Select the doctor. (b) Take a photo. (c) List with any mole. (d) Describe the problem (source: https://
play.google.com/store/apps/details?id=pkg.onlinedermclinic)
448 Charalampos Doukas et al.
Fig. 10 (a) Details of melanoma. (b) Basic menu. (c) In progress of analysis. (d) Result image (source: https://
play.google.com/store/apps/details?id=com.spotmole)
Evaluating Skin Lesions Utilizing Smart Phones and Cloud Platforms 449
2 Material
Table 1
Overview of processing method (local or remote) of related applications
2.1 The Mobile The mobile application provides all the essential functionality for
Application acquiring the image (either from a storage media device like an
SD card or through the mobile’s camera) (see Note 2). It is also
responsible for segmenting the pigmented skin lesion and extract-
ing the essential features. It also allows users to add contextual
information (like age, inheritance factor in melanoma, exposure
to UV radiation) and information about the assessed skin lesion
(like multiplicity factor, age estimation of the lesion, etc.).
The image segmentation and feature extraction is performed
on the mobile device using a method presented in ref. [9]. The
method does not require the usage of specific image processing
libraries and thus can be implemented on every mobile platform.
Initially the skin lesion region is segmented and then texture (like
ASM and GMSM), size (like area, perimeter), asymmetry index,
and color features are generated. More information on the features
utilized can also be found in ref. [4]. The features along with the
contextual information are then encrypted using AES symmetrical
Evaluating Skin Lesions Utilizing Smart Phones and Cloud Platforms 451
2.2 The Cloud Part The Cloud part consists of a Java EE (Java Enterprise Edition for
Web applications) application that provides both the management
graphical interface and the interfaces for the communication with
the sensors. As a Cloud infrastructure the Jelastic platform has
been selected. The Jelastic [12] is a Platform as a Service (PaaS)
type Cloud provider that allows users to deploy Java-based applica-
tions providing all the essential components (application server
instances, databases, load balancers, etc.) and all the appropriate
scalability. Jelastic provides full access to the application server run-
time environment, which enables the deployment of additional
Java extensions like encryption and authentication libraries.
For the specific application the Tomcat application server along
with a MySQL database has been utilized. Data decryption has
been achieved using the java cryptographic extension implement-
ing a symmetric (AES) mechanism using the same encryption key
with the mobile application. Communication with the mobile
application is performed through a REST Web Service API. Once
the data has been received from the mobile device and decrypted,
the contextual data is stored into the database for future usage.
The features are then processed by the data classification module
for characterizing the skin lesion. The module utilizes the WEKA
classification engine [14]. The classification engine uses train
models that have been previously created using the WEKA tool.
Several models can be used to achieve the best accuracy; a train
model can initially validate a new feature set for discriminating the
corresponding image between three classes: melanoma, dysplastic
nevus, and benign nevus. In case a melanoma or a benign nevus is
estimated, a second model can be utilized that characterizes features
to melanoma or nevus providing this way best accuracy to the user.
The train models can be updated with more data or advanced clas-
sification techniques and parameters at any time without affecting
the use of the mobile application and without the need for an
update. The Cloud application has been designed to enable also
452 Charalampos Doukas et al.
3 Methods
3.1 Description For the initial evaluation of the proposed system’s mobile part, an
of the System Android application has been developed and deployed on an
Implementation Android Samsung Galaxy S Plus phone (Android OS v2.3,
1.4 GHz CPU, 512 MB RAM). User initially enters the contextual
information like age, UV exposure, estimated skin lesion age, etc.
(see Fig. 12). Then the skin lesion image is provided, either
through browsing the image file on a local storage media (e.g., an
SD memory card) or by capturing it using the mobile’s camera.
The lesion is segmented, features are extracted and the segmented
image presented to the user (see Fig. 12).
Taking into account the processing capabilities of modern
smartphones a fast algorithm based on a local thresholding tech-
nique was adopted for image segmentation. The window size, the
threshold value and degree of overlap between successive moving
windows were the procedure parameters. These parameters were
tuned so that skin lesions separation was performed satisfactory.
Image thresholding is performed using only the intensity value;
therefore the image is firstly converted into gray scale. Furthermore,
image pixels are smoothed using a standard Gaussian filter, whose
moving window size value is appropriately tuned, for reducing
noise. According to the proposed method, image intensity is
directly compared to the average value of this specific feature com-
puted for all pixels that reside within a wide rectangular area (win-
dow) around this pixel. If the pixel feature value is less that the
average window value minus a characteristic threshold value the
pixel is assumed to be part of the skin lesions region (the pixel is
interpreted as “dark” pixel). If the window is wide enough to con-
tain the entire image for all pixels within it, the technique is called
global thresholding. Typically, to avoid individual image intensity
differences, the feature values are normalized by the average fea-
ture value for all pixels in the image. Thus threshold is provided as
a per-cent fraction of this value. Local (or global) thresholding
partitions the image into objects that can be interpreted as “islands”
of “dark” pixels within the frame. These objects contain image
Evaluating Skin Lesions Utilizing Smart Phones and Cloud Platforms 453
Fig. 12 Screenshots of the Android mobile application. On the left: the initial screen for entering contextual
data. On the middle: A skin lesion image segmented by the mobile application. On the right: history data of
two images
3.2 Initial Evaluation For the initial evaluation of the system a dataset consisting of over
Results 3,000 skin lesion image sets of manually classified images has been
utilized. The dataset contained about 800 images with melanoma,
600 with dysplastic nevus and the rest 1,600 images with benign nevi.
A subset of them, e.g., 80 % of the images is used as learning set and
the other 20 % of the samples are used for testing using the trained
classifier. The images in both learning and test subsets are exchanged
for all possible combinations to avoid bias in the solution.
We have evaluated a number of different classification algo-
rithms provided by the WEKA tool. Accuracy (%) in correctly clas-
sifying instances, Root Mean Square Error (RMS), True Positive
Rates (TPR), False Positive Rates (FPR), and Area under ROC
Curve (AUC) have been also utilized as evaluation metrics. The
latter metrics are considered established evaluation techniques in
machine learning and classification problems [17–19]. The first
experiment involves the evaluation of the algorithms in character-
izing melanoma versus dysplastic and non-dysplastic (i.e., benign)
skin lesions. The corresponding results are presented in Table 2.
The TPR and FPR refer to the detection of melanocytic class.
Evaluating Skin Lesions Utilizing Smart Phones and Cloud Platforms 455
Table 2
Classification results for the characterization of melanocytic, dysplastic,
and Non-dysplastic skin lesions
Table 3
Performance results (in seconds) for the mobile application
4 Notes
References
9. Argenziano G et al (2003) Dermoscopy of melanoma diagnosis by one click? Semin Cutan
pigmented skin lesions: results of a consensus Med Surg 28(3):203–205, ISSN 1085-5629
meeting via the Internet. J Am Acad Dermatol 15. AlShahwan F, Moessner K, Carrez F. (2011)
48(5):680–693 ‘Distributing resource intensive mobile web
10. Argenziano G, Fabbrocini G, Carli P, De services’. 2011 International Conference on
Giorgi V, Sammarco E, Delfino M (1998) Innovations in Information Technology, IIT
Epiluminescence microscopy for the diagnosis 2011, pp. 41–46
of doubtful melanocytic skin lesions. 16. AlShahwan F, Moessner K, Carrez F (2011)
Comparison of the ABCD rule of dermatos- Distributing resource intensive mobile web
copy and a new 7-point checklist based on services. 2011 International conference on
pattern analysis. Arch Dermatol 134(12): innovations in information technology, IIT
1563–1570 2011, 25–27 Apr, Abu Dhabi, pp 41–46
11. Betta G, Di Leo G, Fabbrocini G, Paolillo A, 17. Hall M, Frank E, Holmes G, Pfahringer B,
Scalvenzi M (2005) Automated application of Reutemann R, Witten IH (2009) The WEKA
the “7-point checklist” diagnosis method for data mining software: an update. SIGKDD
skin lesions: estimation of chromatic and shape Explor Newsl 11(1):10–18, doi:10.1145/
parameters. In: Proceedings of the IEEE 1656274.1656278, http://doi.acm.org/
instrumentation and measurement technology 10.1145/1656274.1656278
conference (IMTC ‘05), May 2005, pp
18. Walter SD (2005) The partial area under the sum-
1818–1822
mary ROC curve. Stat Med 24(13):2025–2040
12. Maglogiannis I (2003) Automated segmenta-
19. Gatsonis C, Paliwal P (2006) Meta-analysis of
tion and registration of dermatological images.
diagnostic and screening test accuracy evalua-
J Math Model Algorithm 2:277–294
tions: methodologic primer, research, funda-
13. Maglogiannis I, Pavlopoulos S, Koutsouris D mentals of clinical research for radiologists. Am
(2005) An integrated computer supported J Roentgenol 187:271–281
acquisition, handling and characterization sys-
20. Okeanos Cloud Infrastructure. https://okeanos.
tem for pigmented skin lesions in dermatologi-
grnet.gr/home/
cal images. IEEE Trans Inf Technol Biomed
9(1):86–98 21. Maragoudakis M, Maglogiannis I (2011) A
14. Massone C, Brunasso AMG, Campbell TM, medical ontology for intelligent web-based skin
Peter Soyer H (2009) Mobile teledermoscopy: lesions image retrieval. Health Informatics J
17(2):140–157
Chapter 30
Abstract
Smartphones of the latest generation featuring advanced multicore processors, dedicated microchips for
graphics, high-resolution cameras, and innovative operating systems provide a portable platform for run-
ning sophisticated medical screening software and delivering point-of-care patient diagnostic services at a
very low cost. In this chapter, we present a smartphone digital dermoscopy application that can analyze
high-resolution images of skin lesions and provide the user with feedback about the likelihood of malig-
nancy. The same basic procedure has been adapted to evaluate other skin lesions, such as the flesh-eating
bacterial disease known as Buruli ulcer. When implemented on the iPhone, the accuracy and speed achieved
by this application are comparable to that of a desktop computer, demonstrating that smartphone applica-
tions can combine portability and low cost with high performance. Thus, smartphone-based systems can be
used as assistive devices by primary care physicians during routine office visits, and they can have a significant
impact in underserved areas and in developing countries, where health-care infrastructure is limited.
Key words Skin cancer detection, Melanoma screening, Buruli ulcer, Smartphones in health care,
Handheld devices
1 Introduction
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0_30, © Springer Science+Business Media New York 2015
459
460 George Zouridakis et al.
1.1 Skin Cancer Risk Over two million people are diagnosed with some form of skin
cancer annually, including malignant melanoma, basal cell carci-
noma, and squamous cell carcinoma [7, 8]. Melanoma represents
only 5 % of all new cases; however, it accounts for the vast majority
of skin cancer deaths globally [9]. In the USA, melanoma is esti-
mated to claim 12,650 lives in 2013 [7], whereas melanoma in situ,
a very early stage of melanoma, can add tens of thousands of more
cases, as it is growing at 15 % per year [10]. For patients whose
melanoma is detected early the overall 5-year survival rate is about
98 %, but this rate falls to 62 % when the disease reaches the lymph
nodes, and to only 15 % when the disease metastasizes to distant
organs [7]. Thus, early detection is of paramount importance.
The most effective method for early detection of melanoma is
skin self-examination [11]. In fact, a substantial percentage of mel-
anomas are detected initially by individuals who examine them-
selves visually [12–14] according to the “ABCDE” rules
recommended by the American Academy of Dermatology [15]
and report to the doctor any suspicious skin changes. However,
the differentiation of early melanoma from other pigmented skin
lesions, such as benign neoplasms that mimic melanoma, is not
trivial, and even primary care physicians may underestimate a mela-
noma in its early stage [16]. This has motivated researchers to
develop automated systems for screening early melanoma that can
be deployed to the general population using the now-a-days ubiq-
uitous smartphones for image acquisition and real time analysis.
1.2 Diagnostic The first studies for automated classification of pigmented skin
Criteria lesions appeared in the literature more than two decades ago [17],
but today the majority of the analysis procedures follow a set of
objective dermoscopic criteria that classify a lesion based on the
presence or absence of certain characteristic features of melanoma.
These criteria were established after several methods were evalu-
ated by an international group of experts during the 2000
Consensus Net Meeting on Dermoscopy [1]. Here we present
only the three most popular lesion-scoring schemes, since these are
the ones we have implemented in our iPhone-based system pre-
sented in later sections. All three scoring schemes are summarized
in Table 1.
462 George Zouridakis et al.
Table 1
Common melanoma detections schemes: diagnostic criteria and scoring rules
1.3 Image Analysis Several melanoma detection systems have been developed during
the past two decades, most of which follow a three-stage procedure
to analyze a lesion, namely, image segmentation, feature extrac-
tion, and lesion classification.
Image segmentation is a critical step as it defines the contour
and thus the symmetry of the lesion, which is a basic feature used
Skin Lesion Screening using Smartphones 463
Fig. 1 Handheld cameras used by popular melanoma screening systems: (left) Melafind, (middle) SolarScan,
and (right) SIAscope V
Fig. 2 iPhone attachments for skin cancer screening: (left ) DermLite and (right ) Handyscope attachments
2 Materials
2.1 Hardware
Implementation A block diagram of a self-contained handheld system for automated
detection of melanoma is shown in Fig. 3. The hardware presented is
a prototype for the handheld skin cancer detector and is an alternative
466 George Zouridakis et al.
Fig. 3 Block diagram of a Texas Instruments TMS320DM642-based automated melanoma detection system
digital-to-
analog converter, converts the digital signal back to
analog for real time image display on an LCD. A boot flash (AMD
AM29LV033C) is used for program storage. A universal asynchro-
nous receiver-transmitter (UART transceiver TI TLC16C752B_9)
is added to the system for interfacing with the Bluetooth module
(TAIYO YUDEN EYMF2CAMM). A keypad and LCD display
terminal provide the human machine interface (HMI) functions.
This prototype system is designed with a HMI board, and includes
buttons and LEDs, which enable the user to control and monitor
system function. The power management module (PWR.MGMT.)
includes various switching regulators to meet the voltage require-
ments of the different blocks in the system. An 8-bit latch extends
control I/Os, and power regulators generate 1.4 V core voltage
and 3.3 V I/O voltage for the TMS320DM642 and other chips.
The TMS320DM642 has several power-down modes to minimize
power consumption when idle. The 1.4 V core voltage can be
reduced to 1.2 V with optimized software (reduced MIPS con-
sumption) and CPU clock can be slowed down for further power
savings. A picture of the prototype for the handheld skin cancer
detector is shown in Fig. 4.
2.2 Software Early on a desktop application was developed [30, 52, 55, 59]
Implementation based on Matlab (MathWorks Inc., Natick, MA) that could auto-
matically analyze a skin lesion image and calculate the probability
Video
Out
Video
Encoder
3 Methods
3.1 The System Our skin cancer detection system consists of two main modules,
namely, image acquisition and image analysis. We implement image
acquisition using the DermLite II Multispectral dermoscope (www.
dermlite.com) which can provide white light cross- polarization
(XLM) epiluminescence, blue light for surface coloration, yellow
light for superficial vascularity, and red light for deeper coloration
and vascularity, using 32 bright white LEDs—8 per color. This der-
moscope gives the choice between free-floating “dry” skin imaging
and oil-immersion epiluminescence, at any color. The dermoscope
can be attached to a high-resolution camera, e.g., the Sony
Cybershot DSC-W300, which provides a resolution of 13.5 MP.
Under this configuration, image analysis must be implemented off
line on a laptop or tablet computer. Real time analysis is accom-
plished when the dermoscope is attached directly to an iPhone
4/5/5s that combines image acquisition and image analysis in one
device. Furthermore, if multispectral imaging is not required, a
smaller dermoscope, such as the pocket Dermlite DL1, can also be
used with the iPhone. Figure 5 shows various configuration exam-
ples, namely, the DermLite II Multispectral d ermoscope (left), the
same dermoscope attached to an off-the-shelf Sony camera (mid-
dle), and the Dermlite DL1 attached to an iPhone 5s (right).
3.2 Overall Analysis The entire procedure used in our system is graphically depicted in
Procedure Fig. 6. Initially, one or more images of a lesion acquired with light
of different frequencies or under different illumination modalities,
including transillumination (TLM) and cross-polarization (XLM)
epiluminescence, are preprocessed and then segmented with four
different methods. The segmented images are fed into a scoring
stage that takes into account statistical properties of the lesion and
selects the best segmentation result out of which a refined lesion
boundary is computed and superimposed onto the original images.
Subsequently, geometrical, statistical, texture, and color features are
Skin Lesion Screening using Smartphones 469
Fig. 5 The DermLite II Multispectral dermoscope (left) can be attached to an off-the-shelf Sony Cybershot
camera (middle). Similarly, the Dermlite DL1 can be attached to an iPhone 5s (right)
Fig. 6 Software architecture implemented by our automated system for lesion classification. Multispectral
images acquired under transillumination (TLM) or cross-polarization (XLM) epiluminescence imaging undergo
distinct stages of analysis before a probability of malignancy is computed for the lesion under evaluation
Fig. 7 Software architecture optimized for the iPhone. A suspicious lesion is evaluated for malignancy sepa-
rately using the ABCD rules, Menzies’ method, and the 7-point checklist
3.3.1 Stage 1: The very first processing step involves conversion of color RGB
Preprocessing images to gray scale (Gray) using the standard formula [64] for
averaging the R, G, and B channels, Gray = 0.299 × R + 0.587 × G +
0.114 × B.
Hair artifacts and salt-and-pepper noise are also removed, or
minimized, using median filtering [65, 66], which in general
reduces noise while it preserves image edges. We use a fast two-
dimensional algorithm [67] with two structuring elements of 5 × 5
and 11 × 11 pixels. In this algorithm, a histogram is used to store
the neighboring pixel values. When moving the center pixel over
the image, only a part of the histogram is modified, thus reducing
the computational complexity from O(n2) to O(n). The exact steps
of the algorithm in a generic pseudocode language are shown in
Fig. 8.
Histogram Equalization, needed to increase the contrast of
images with pixel intensity levels grouped in a narrow range, is
performed by spreading out the most frequent intensity values.
Given a gray scale image of size M × N pixels, we start by comput-
ing the intensity histogram H(j) and the cumulative density func-
tion, cdf(i) = ∑ j = 0iH(j), and then we use monotonic nonlinear
3.3.2 Stage 2: Image To identify the border that separates the foreground lesion from
Segmentation the background healthy skin, in our early implementations [70, 71]
we used four classical segmentation methods based on sigmoid
histogram remapping [72], principal component transformation
[32, 73], histogram Gaussian modeling [73], and fuzzy clustering
[74], and later we employed evolutionary strategy-based segmen-
tation. The objective function we adopted favors an ellipse that
divides the image into two homogeneous areas with minimum
variation in both regions, and it is given by F(X, Y, a, b, θ) = ∫ ω|I(x, y)
− c1|2dxdy + ∫ Ω\ω|I(x, y) − c2|2dxdy, where I(x, y) represents the image
intensity value at coordinates (x, y), ω is the area enclosed by the
ellipse defined by (X, Y, a, b, θ), Ω is the area of pixels with non-
zero intensity value, and c1 and c2 represent the average intensity
value of the pixels inside and outside ω, respectively.
More recently, however, we developed a better method based
on active contours [75] and narrow band graph partitioning
that includes geodesic active contours and gradient vector flow
snakes that rely on gradient information. The method follows
active contour region fusion [53] in a hierarchical approach. Briefly,
it first segments the lesion into small regions based on strict con-
straints on homogeneity and edge strength using region-based
active contours, and then merges the regions together based on
centroid criteria and gradient information. To further improve
robustness and computational efficiency, we followed narrow band
graph partitioning. This approach is more robust against weak or
false edges and against lesion asymmetry. The performance of the
Skin Lesion Screening using Smartphones 473
Fig. 9 Segmentation of an asymmetric lesion with weak border: (a) modified level set method and (b) narrow
band energy based approach (right) optimized for the iPhone
Fig. 13 Example of image segmentation using the Fuzzy C Means and Active Contour algorithms
Fig. 14 Example showing the need for postprocessing the mask identified by the segmentation algorithm to
remove the area inside the lesion
Connected Component We apply this algorithm first to the foreground pixels to remove
Labeling regions outside the lesion and then to the background pixels to
remove holes inside the lesion. More specifically, we start the pro-
cedure by scanning all the pixels in a binary image and grouping
Skin Lesion Screening using Smartphones 477
3.3.4 Stage 4: Feature After segmentation, we extract a large number of low-level features
Extraction that relate to a lesion’s shape, texture, and color. Typically, this
requires image sampling as a first step. The sampling operator
Lesion Sampling Strategies
selects N pixels inside a lesion and then it centers a patch of p × p
pixels at each location. The number of patches sampled affects the
memory required and the speed of algorithm execution; therefore,
minimization of both parameters is of paramount importance in
handheld devices (see Note 2).
The full desktop system relies on a combination of heuristic
rules, autoregressive modeling, Laws masks, Gabor features, and
wavelet transforms (WTs) to define features that relate to the high-
level dermoscopic criteria of Subheading 2 [51, 52, 70, 71].
However, through exhaustive parameter exploration, we found
[30, 85] that the handheld implementation can use only a subset
of features that constitute a reasonable trade-off between classifica-
tion accuracy and computation speed.
Skin Lesion Screening using Smartphones 479
Fig. 16 Various steps to obtain a smooth lesion boundary after segmentation: (a) original boundary, (b) basis
function boundary, (c) minor ridges, (d) smooth boundary, and (e) smooth boundary superimposed on the origi-
nal image
Lesion Geometry Analysis Lesion shape, one of the most important for human perception
low-level lesion features, is highly irregular in several lesions.
Therefore, we extract three measures for estimating lesion irregu-
LesionArea
larity [67]: Total Irregularity, given by IRtot = 4π ;
LesionPerimeter 2
1 4
Sector Irregularity, given by IR sec = ∑ min area ( Si ) − area ( S j ) ,
4 i −1 j =1...4, j ≠ i
where Si, i = 1…4, are four possibly overlapping sectors
defined by four points on the lesion boundary that intersect
with the minimum rectangle bounding the lesion; and
n −1
larity, given by
Contour Irregu IRcon = ∑ d j − d j −1 , where
j =0
y j − y j−w y j −1 − y j − w−1
d j = arctan − arctan represents the curvature
x j − x j−w x j −1 − x j − w−1
at a boundary point pj = (xj, yj), when the boundary is represented
by a series of points {p0, p1, …, pn − 1}, with p0 = pn.
Lesion Texture Analysis Texture analysis relies on wavelet [86] and local binary pattern
[87] analysis. Wavelets are used to hierarchically decompose a
given continuous-time signal into different frequency components.
In general, a continuous function can be represented as the sum of
480 George Zouridakis et al.
φ j , m, n = 2 j / 2 φ ( 2 j x − m, 2 j y − n )
ψ ij ,m ,n = 2 j / 2ψ i ( 2 j x − m, 2 j y − n )
where the index i = {H,V,D} identifies horizontal (H), vertical (V),
and diagonal (D) directional wavelets. The DWT of image f(x,y) of
size M × N pixels is given by
M −1 N −1
1
Wφ ( j ,m,n ) =
MN
∑∑ f ( x,y )φ j 0, m , n ( x ,y )
x =0 y =0
1 M −1N −1
Wψi ( j ,m,n ) = ∑∑
MN x = 0 y = 0
f ( x,y )ψ ij 0,m ,n ( x,y )
Typically we set j0 = 0 and N = M = 2j, with j = 0, 1 … J − 1 and m,
n = 0, 1 … J − 1, so that the image is decomposed into sub-bands
that are dyadically subsampled. After one level of decomposition,
four sub-band images are obtained, but the approximation image
can be further decomposed using the same procedure. In general,
a DWT with d decomposition levels will yield a total of 3d + 1 sub-
bands. DWT provides information on the various structural pat-
terns present in a skin lesion used by the dermoscopic criteria of
Subheading 2, such as pigmentation networks, vascular structures,
and dots and globules [88].
Haar Wavelet As wavelet function we use the Haar transform, which is given by
ψij(x) = ψ(2jx − i), where i = 0, …, 2j − 1 and
1 if 0 ≤ t < 1 / 2
ψ ( t ) = −1 if 1 / 2 ≤ t < 1
0 otherwise
Previous work [45] and our own studies [30] have demonstrated
the effectiveness of Haar wavelet coefficients and local binary pat-
terns [89] for melanoma detection. Images are decomposed into
three levels, each resulting in ten sub-bands images. The extracted
features used by the classifier are the mean and standard deviation
of each sub-band image. The Haar wavelet transform algorithm for
two-dimensional images is given in Fig. 17, and the feature extrac-
tion process is summarized in Fig. 18. For local feature extraction,
the image is divided into overlapping patches of K × K pixels cen-
tered on a square grid of size M pixels. When the algorithm runs
Skin Lesion Screening using Smartphones 481
Local Binary Pattern (LBP) LBP is a robust texture operator [89] defined on gray scale images.
It is invariant to both monotonic transformation of intensity and
to rotation. It can be derived using a symmetric neighborhood of
P members on a circle of radius R, where the parameter P repre-
sents the quantization of angular space in the circular neighbor-
hood, and R represents the spatial resolution. A limited number of
transitions or discontinuities (0/1 changes in LBP) are allowed to
reduce the noise and improve discrimination of features; however,
we restrict the maximum number of transitions to P, so that transi-
tions greater than P are considered equal. An occurrence histo-
gram, denoted by LBPPRriu, provides statistical and structural
information on the image and is computed following the steps
shown in Fig. 19.
Lesion Color Analysis Several dermoscopic criteria listed in Subheading 2 analyze color
intensity and patterns of color distribution to characterize a skin
Color Histograms
lesion. Examples include the presence of brown dots, blue-whitish
veil, and regression, which consist of mixtures of certain colors.
482 George Zouridakis et al.
Fig. 18 Feature extraction process based on Haar Wavelets optimized for the
iPhone
2
The RGB color space uses separate channels for red, green, and blue color
intensity, HSV uses separate channels for hue, saturation, and brightness,
whereas LAB uses one channel for luminance L, and two color channels,
A and B, that correspond to perceived intensity of complementary colors.
Skin Lesion Screening using Smartphones 483
Fig. 21 Various steps used during the training and testing phases of classifier
development
3.3.5 Stage 5: All the features extracted from a lesion, such as shape Si, color
Classification histogram Ci, Haar wavelet Hi, and local binary patterns Li are
combined to form a singe feature vector Fi = {Si, Ci, Hi, Li}.
Depending on what particular dermoscopic criteria we want to use
for lesion assessment, we filter the feature vector to select a subset
of Fi keeping only those features with the highest classification
accuracy for the particular dermoscopic rule. Correlation coeffi-
cient values between Fi and each criterion are used to rank the fil-
ters. Each criterion requires both training and testing before it is
deployed. To build a support vector machine (SVM) classifier, typi-
cally a dataset is divided into several folds, each containing both
malignant and benign lesions. The majority of the folds are used
for training of the classifier, whereas the remaining folds are used
for testing. The optimum number of features and parameters of
the linear SVM classifier are obtained during the training phase by
exhaustive search. The two phases of building a classifier are shown
in Fig. 21.
Even though the full version of the system employs several clas-
sification algorithms, the handheld device uses only the so-called
“2C” formulation of the SVM algorithm [91] that can successfully
484 George Zouridakis et al.
3.4 Application This section illustrates skin lesion analysis on the Apple iPhone.
Examples For this particular example, we used images from the Interactive
CD of Dermoscopy,3 a large commercial library of skin cancer
3.4.1 Detection
images annotated by expert dermatologists. The library contains
of Skin Cancer
360 lesions, 270 benign and 90 melanomas; however, 13 images
that did not produce satisfactory segmentation results were
excluded from this analysis.
All analyses were done on an iPhone 4, using our menu-driven
application that implements all the algorithms of the automated
procedure for lesion classification described in the previous sections.
The user can select any dermoscopic image from the database or
take a new one using the DermLite attachment, analyze it using
3
http://www.dermoscopy.org/atlas/cd-review.asp
Skin Lesion Screening using Smartphones 485
Fig. 22 Example of automatic skin lesion analysis on the iPhone: (a) splash screen and (b) classification meth-
ods available for lesion analysis; (c) final lesion classification: the black line represents the border of the seg-
mented lesion; the score according to the 7-point checklist is shown at the bottom of the screen
Table 2
Confusion matrix computed as the average of tenfold cross-validation
across ten test sets and the resulting average sensitivity and specificity
of the automated analysis procedure on the iPhone
iPhone
Table 3
Classification results using individual and combined criteria of the 7-point
checklist on the iPhone
Execution Time We contrast the time needed for classification using the ISODATA
segmentation algorithm as implemented on the Apple iPhone 3G
against a typical desktop computer (2.26 GHz Intel Core 2 Duo
with 2GB RAM). The choice of the iPhone 3G would provide the
worst-case scenario for the execution time for our algorithm, since
all the following iPhone models are much faster.4 Classification
time includes time needed for feature extraction.
4
The iPhone 3G features a single core 0.6 GHz ARM Cortex processor,
0.25 GB of RAM memory, and quad core 150 MHz PowerVR SGX535 GPU
with screen resolution of 164 ppi, whereas the latest model iPhone 5s features
a dual core 1.3 GHz ARM Cyclone (64 bit), 1 GB of RAM memory, and
quad core 300 MHz, PowerVR Series 6 Rogue G6430 GPU with screen reso-
lution of 325 ppi (source: http://goo.gl/88YB3U).
Skin Lesion Screening using Smartphones 487
Table 4
Average computation time in seconds for the iPhone and desktop
platforms
3.4.2 Detection of Buruli The procedures presented in the previous sections can be adapted
Ulcer Disease and applied to other skin diseases that require early detection.
We have recently developed [59, 62] an application for automatic
detection of Buruli ulcer [63, 95, 96], a devastating flesh-eating
bacterial infection that, each year, affects thousands of people,
mostly children under the age of 15 years [97, 98], and results in
life debilitating amputations. However, if detected early, BU can
be easily treated with oral medication and cured [99].
BU image analysis starts with image acquisition, using either a
camera or the iPhone with a DermLite IIm attachment, which can
acquire multispectral images. Once an image is loaded, the system
can automatically perform lesion preprocessing, segmentation, fea-
ture extraction, feature selection, classification, and graphically
provide a probability of the lesion being Buruli, as shown in Fig. 23
(see Note 4).
3.5 New Framework The process of implementing new methods on mobile devices is
to Facilitate rather challenging, as a new algorithm typically undergoes a series
Development of steps before it can be implemented on the handheld device: it is
on Mobile Device first implemented in Matlab (for testing), then it is ported to the
C/C++ language and compiled for use by Matlab as a “mex”
library, and finally it is ported to the host language for the target
device. These steps also require modifying the C code to use mem-
ory management and threading functions from the operating system,
and writing or modifying a substantial amount of Objective-C
code to expose the new algorithm to the user interface and display
the results. If, as is often the case, the original algorithm is later
modified/improved using Matlab, some or all of the intermediate
steps must be repeated.
488 George Zouridakis et al.
Fig. 24 Overview of the new framework that optimizes code development and
maintenance for the Apple iOS 7.1 operating system and iPhone 5s architecture
the native programming language of the host device; and takes full
advantage of multiprocessor platforms, graphical processing units
(GPUs), and other recently developed hardware features.
4 Notes
Fig. 25 Examples showing different lesion sampling strategies based on (a) dense square-grid, (b) random,
(c) key-point, and (d) saliency sampling. Blue circles correspond to the patch centers selected by various
sampling methods
5 Concluding Remarks
Acknowledgment
This research has been supported in part by NSF grant 521527 and
NIH grant 5R21AR057921-02. This chapter was developed while
GZ was a Visiting Professor at the Basque Center on Cognition,
Brain, and Language, in Donostia-San Sebastián, Spain, and a
Fellow of the Ikerbasque Foundation, Basque Country, Spain.
492 George Zouridakis et al.
References
1. Argenziano G, Soyer HP, Chimenti S, 16. Pariser RJ, Pariser DM (1987) Primary care
Talamini R, Corona R, Sera F et al (2003) physicians errors in handling cutaneous disor-
Dermoscopy of pigmented skin lesions: ders. J Am Acad Dermatol 17:239–245
results of a consensus meeting via the Internet. 17. Cascinelli N, Ferrario M, Tonelli T, Leo E
J Am Acad Dermatol 48(5):679–693 (1987) A possible new tool for clinical diag-
2. Argenziano G, Soyer HP, De Giorgi V, nosis of melanoma: the computer. J Am Acad
Piccolo D, Carli P, Delfino M (2002) Dermatol 16(2):361–367
Dermoscopy: a tutorial. EDRA, Medical 18. Stolz W, Riemann A, Cognetta AB, Pillet L,
Publishing & New Media, Milan Abmayr W, Holzel D et al (1994) Abcd rule
3. Bafounta ML, Beauchet A, Aegerter P et al of dermatoscopy: a new practical method for
(2001) Is dermoscopy (epiluminescence early recognition of malignant-melanoma.
microscopy) useful for the diagnosis of mela- Eur J Dermatol 4(7):521–527
noma? Arch Dermatol 137:1343–1350 19. Nachbar F, Stolz W, Merkle T, Cognetta A,
4. Kittler H, Pehamberger H, Wolff K et al Vogt T, Landthaler M, Bilek P, Braun-Falco
(2002) Diagnostic accuracy of dermoscopy. O, Plewig G (1994) The ABCD rule of der-
Lancet Oncol 3:159–165 matoscopy: high prospective value in the
5. Menzies SW (2013) Evidence-based dermos- diagnosis of doubtful melanocytic skin lesions.
copy. Dermatol Clin 31(4):521–524 J Am Acad Dermatol 30(4):551–559
6. Menzies SW, Bischof L, Talbot H, Gutenev A 20. Menzies SW, Ingvar C, Crotty KA, McCarthy
et al (2005) The performance of SolarScan: WH (1996) Frequency and morphologic
an automated dermoscopy image analysis characteristics of invasive melanomas lacking
instrument for the diagnosis of primary mela- specific surface microscopic features. Arch
noma. Arch Dermatol 141(11):1388–1396 Dermatol 132(10):1178
7. ACS: American Cancer Society, Cancer Facts & 21. Argenziano G, Fabbrocini G, Carli P, De
Figures 2013. http://goo.gl/rgD56, Accessed Giorgi V, Sammarco E, Delfino M (1998)
31 Sep 2013 Epiluminescence microscopy for the diagno-
8. Rogers HW, Weinstock MA, Harris AR et al sis of doubtful melanocytic skin lesions: com-
(2010) Incidence estimate of nonmelanoma parison of the ABCD rule of dermatoscopy
skin cancer in the United States, 2006. Arch and a new 7-point checklist based on pattern
Dermatol 146(3):283–287 analysis. Arch Dermatol 134(12):1563
9. Parkin D, Bray F, Ferlay J, Pisani P (2005) 22. Johr RH (2002) Dermoscopy: alternative
Global cancer statistics, 2002. CA Cancer J melanocytic algorithms—the ABCD rule of
Clin 55(2):74–108 dermatoscopy, menzies scoring method, and
10. Thorn M, Penten F, Johansson B et al (1998) 7-point checklist. Clin Dermatol 20(3):
Rapid increase in diagnosis of cutaneous mel- 240–247
anoma in situ in Sweden, 1968–1992. Cancer 23. Silveira M, Nascimento JC, Marques JS,
Detect Prev 22:430 Marçal AR, Mendonça T, Yamauchi S et al
11. Pollitt R, Geller A, Brooks D, Johnson T, (2009) Comparison of segmentation meth-
Park E, Swetter S (2009) Efficacy of skin self- ods for melanoma diagnosis in dermoscopy
examination practices for early melanoma images. IEEE J Sel Top Signal Process 3(1):
detection. Cancer Epidemiol Biomarkers Prev 35–45
18:3018–3023 24. Wang H, Chen X, Moss RH, Stanley RJ,
12. Brady MS, Oliveria SA, Christos PJ et al Stoecker WV, Celebi ME et al (2010)
(2000) Patterns of detection in patients with Watershed segmentation of dermoscopy
cutaneous melanoma. Cancer 89(2):342–347 images using a watershed technique. Skin Res
13. Epstein DS, Lange JR, Gruber SB, Mofid M, Technol 16(3):378–384
Koch SE (1999) Is physician detection associ- 25. Celebi ME, Kingravi HA, Uddin B, Iyatomi
ated with thinner melanomas? JAMA H, Aslandogan YA, Stoecker WV, Moss RH
281(7):640–643 (2007) A methodological approach to the
14. McGuire ST, Secrest AM, Andrulonis R, classification of dermoscopy images. Comput
Ferris LK (2011) Surveillance of patients for Med Imaging Graph 31(6):362–373
early detection of melanoma: patterns in der- 26. Iyatomi H, Oka H, Celebi ME, Hashimoto
matologist vs patient discovery. Arch M, Hagiwara M, Tanaka M, Ogawa K (2008)
Dermatol 147(6):673–678 An improved internet-based melanoma
15. AAD: American Academy of Dermatology. screening system with dermatologist-like
How to perform a self-exam. http://goo. tumor area extraction algorithm. Comput
gl/6D9E1d. Accessed 27 Oct 2013 Med Imaging Graph 32(7):566–579
Skin Lesion Screening using Smartphones 493
27. Nascimento JC, Marques JS (2005) Adaptive 39. Green A, Martin N, Pfitzner J, O’Rourke M,
snakes using the EM algorithm. IEEE Trans Knight N (1994) Computer image analysis in
Image Process 14(11):1678–1686 the diagnosis of melanoma. J Am Acad
28. Seidenari S, Pellacani G, Grana C (2005) Dermatol 31(6):958–964
Pigment distribution in melanocytic lesion 40. Ercal F, Chawla A, Stoecker WV, Lee HC,
images: a digital parameter to be employed Moss RH (1994) Neural network diagnosis
for computer‐aided diagnosis. Skin Res of malignant melanoma from color images.
Technol 11(4):236–241 IEEE Trans Biomed Eng 41(9):837–845
29. Situ N (2011) Melanoma recognition using 41. Rubegni P, Cevenini G, Burroni M, Perotti R,
mid-level features and high-level annotations. Dell'Eva G, Sbano P et al (2002) Automated
Doctoral dissertation, University of Houston diagnosis of pigmented skin lesions. Int J
30. Situ N, Yuan X, Chen J, Zouridakis G (2008) Cancer 101(6):576–580
Malignant melanoma detection by bag-of- 42. Blum A, Luedtke H, Ellwanger U, Schwabe R,
features classification. In 30th annual interna- Rassner G, Garbe C (2004) Digital image anal-
tional conference of the IEEE Engineering in ysis for diagnosis of cutaneous melanoma.
Medicine and Biology Society, 2008 (EMBS Development of a highly effective computer
2008). IEEE, pp 3110–3113 algorithm based on analysis of 837 melanocytic
31. Zhou H, Chen M, Rehg JM (2009) lesions. Br J Dermatol 151(5):1029–1038
Dermoscopic interest point detector and 43. Ganster H, Pinz P, Rohrer R, Wildling E,
descriptor. In IEEE international symposium Binder M, Kittler H (2001) Automated mela-
on biomedical imaging: from nano to macro, noma recognition. IEEE Trans Med Imaging
2009. ISBI'09. IEEE, pp 1318–1321 20(3):233–239
32. Celebi ME, Aslandogan YA, Bergstresser PR 44. Caetano Ruela AM (2012) What is the role of
(2005) Unsupervised border detection of color in dermoscopy analysis? Master of Science
skin lesion images. In International confer- Thesis, Biomedical Engineering, Instituto
ence on information technology: coding and Superior Técnico, Lisbon, Portugal, 2012
computing, 2005 (ITCC 2005), vol 2. IEEE, 45. Elbaum M, Kopf AW, Rabinovitz HS, Langley
pp 123–128 RG, Kamino H, Mihm MC Jr et al (2001)
33. Hoffmann K, Gambichler T, Rick A, Kreutz Automatic differentiation of melanoma from
M, Anschuetz M, Grünendick T et al (2003) melanocytic nevi with multispectral digital
Diagnostic and neural analysis of skin cancer dermoscopy: a feasibility study. J Am Acad
(DANAOS). A multicentre study for collec- Dermatol 44(2):207–218
tion and computer‐aided analysis of data from 46. Monheit G, Cognetta AB, Ferris L, Rabinovitz
pigmented skin lesions using digital dermos- H, Gross K, Martini M et al (2011) The per-
copy. Br J Dermatol 149(4):801–809 formance of MelaFind: a prospective multi-
34. Rubegni P, Burroni M, Cevenini G, Perotti R, center study. Arch Dermatol 147(2):188
Dell'Eva G, Barbini P et al (2002) Digital 47. Moncrieff M, Cotton S, Claridge E, Hall P
dermoscopy analysis and artificial neural net- (2002) Spectrophotometric intracutaneous
work for the differentiation of clinically atypi- analysis: a new technique for imaging pig-
cal pigmented skin lesions: a retrospective mented skin lesions. Br J Dermatol 146(3):
study. J Investig Dermatol 119(2):471–474 448–457
35. Serrano C, Acha B (2009) Pattern analysis of 48. Karargyris A, Karargyris O, Pantelopoulos A
dermoscopic images based on Markov random (2012) DERMA/care: an advanced image-
fields. Pattern Recognit 42(6):1052–1057 processing mobile application for monitoring
36. Tanaka T, Torii S, Kabuta I, Shimizu K, skin cancer. In IEEE 24th international con-
Tanaka M (2008) Pattern classification of ference on tools with artificial intelligence
nevus with texture analysis. IEEE Trans Elect (ICTAI), vol 2, 2012. IEEE, pp 1–7
Electron Eng 3(1):143–150 49. Doukas C, Stagkopoulos P, Kiranoudis CT,
37. Andreassi L, Perotti R, Rubegni P, Burroni Maglogiannis I (2012) Automated skin lesion
M, Cevenini G, Biagioli M et al (1999) assessment using mobile technologies and
Digital dermoscopy analysis for the differen- cloud platforms. In annual international con-
tiation of atypical nevi and early melanoma: a ference of the IEEE Engineering in Medicine
new quantitative semiology. Arch Dermatol and Biology Society (EMBC), 2012. IEEE,
135(12):1459 pp 2444–2447
38. Green A, Martin N, McKenzie G, Pfitzner J, 50. Celi LA, Sarmenta L, Rotberg J, Marcelo A,
Quintarelli F, Thomas BW et al (1991) Clifford G (2009) Mobile care (moca) for
Computer image analysis of pigmented skin remote diagnosis and screening. J Health
lesions. Melanoma Res 1(4):231–236 Inform Dev Ctries 3(1):17
494 George Zouridakis et al.
51. Yuan X, Situ N, Zouridakis G (2008) 63. Johnson PD, Azuolas J, Lavender CJ, Wishart
Automatic segmentation of skin lesion images E, Stinear TP, Hayman JA et al (2007)
using evolution strategies. Biomed Signal Mycobacterium ulcerans in mosquitoes cap-
Process Control 3:220–228 tured during outbreak of Buruli ulcer, south-
52. Situ N, Wadhawan T, Hu R, Lancaster K, eastern Australia. Emerg Infect Dis 13(11):1653
Yuan X, Zouridakis G (2011). Evaluating 64. Gonzalez R, Woods R (2002) Digital image
sampling strategies of dermoscopic interest processing. Prentice Hall, Upper Saddle
points. In IEEE international symposium on River, NJ, pp 349–408
biomedical imaging: from nano to macro, 65. Tukey J (1977) Exploratory data analysis.
2011. IEEE, pp 109–112 Addison-Wesley, Menola Park, CA
53. Yuan X, Situ N, Zouridakis A (2009) A nar- 66. Huang T, Yang G, Tang G (1979) A fast two-
row band graph partitioning method for skin dimensional median filtering algorithm. IEEE
lesion segmentation. Pattern Recognit Trans Acoust Speech Signal Process 27(1):
42(6):1017–1028 13–18
54. Hu R, Queen CM, Zouridakis G (2012) 67. Huang PW, Lai YH (2010) Effective segmen-
Lesion border detection in Buruli ulcer tation and classification for HCC biopsy
images. In annual international conference of images. Pattern Recognit 43(4):1550–1563
the IEEE Engineering in Medicine and 68. Acharya T, Ray AK (2005) Image processing:
Biology Society (EMBC), 2012. IEEE, principles and applications. Wiley-
pp 5380–5383 Interscience, New York
55. Wadhawan T, Situ N, Rui H, Lancaster K, 69. Fries R, Modestino J (1979) Image enhance-
Yuan X, Zouridakis G (2011) Implementation ment by stochastic homomorphic filtering.
of the 7-point checklist for melanoma detec- IEEE Trans Acoust Speech Signal Process
tion on smart handheld devices. In annual 27(6):625–637
international conference of the IEEE 70. Zouridakis G, Doshi M, Mullani N (2004)
Engineering in Medicine and Biology Society, Early diagnosis of skin cancer based on seg-
EMBC, 2011. IEEE, pp 3180–3183 mentation and measurement of vasculariza-
56. Zouridakis G, Yuan X, Chen J, Stotzer E, Wu tion and pigmentation in nevoscope images.
Y (2012) Device and software for screening In 26th annual international conference of
the skin. University of Houston, July 2012. the IEEE Engineering in Medicine and
US 08213695 Biology Society, 2004. IEMBS’04, vol 1.
57. Zouridakis G, Yuan X, Chen J, Stotzer E, Wu Y IEEE, pp 1593–1596
(2008) Device and software for screening the 71. Zouridakis G, Doshi M, Duvic M, Mullani NA
skin, September 2008. US 20080226151-A1 (2005) Transillumination imaging for early
58. Wu Y, Chen J, Yuan X, Zouridakis G (2009) A skin cancer detection. Biomedical Imaging
programmable DSP development platform for Lab, Department of Computer Science,
automated detection of melanoma. Int J Intel University of Houston, Houston, TX. http://
Comput Med Sci Image Process 3(1):1–8 goo.gl/NPkLBp
59. Hu, R (2013) Automated recognition of 72. Song B, Chan T (2002) A fast algorithm for
Buruli ulcer images on smart handheld devices. level set based optimization. UCLA Cam
Doctoral dissertation, University of Houston Report, 02-68
60. Lancaster K, Zouridakis G (2014) A software 73. Sapiro G (2000) Segmenting skin lesions with
framework to facilitate development of image partial-differential-equations-based image
analysis applications on mobile and embed- processing algorithms. IEEE Trans Med
ded devices. PECCS 2014: International con- Imaging 19(7):763–767
ference on pervasive and embedded 74. Erkol B, Moss RH, Joe Stanley R, Stoecker
computing and communication systems, 7–9 WV, Hvatum E (2005) Automatic lesion
Jan 2014, Lisbon, Portugal boundary detection in dermoscopy images
61. Wadhawan T, Situ N, Lancaster K, Yuan X, using gradient vector flow snakes. Skin Res
Zouridakis G (2011) SkinScan©: a portable Technol 11(1):17–26
library for melanoma detection on handheld 75. Kass M, Witkin A, Terzopoulos D (1988)
devices. In IEEE International Symposium Snakes: active contour models. Int J Comput
on biomedical imaging: from nano to macro, Vision 1(4):321–331
2011. IEEE, pp 133–136 76. Ridler TW, Calvard S (1978) Picture thresh-
62. Hu R, Queen CM, Zouridakis G (2013) olding using an iterative selection method.
Detection of Buruli ulcer disease: preliminary IEEE Trans Syst Man Cybern 8(8):630–632
results with dermoscopic images on smart 77. Qureshi S (2005) Embedded image process-
handheld devices. In Point-of-Care Healthcare ing on the TMS320C6000TM DSP: exam-
Technologies (PHT), 2013 IEEE. IEEE, ples in code composer studioTM and
pp 168–171 MATLAB. Springer, New York
Skin Lesion Screening using Smartphones 495
78. Bezdek JC (1981) Pattern recognition with international joint conference on artificial
fuzzy objective function algorithms. Kluwer intelligence, vol 1999. pp 55–60
Academic Publishers, Norwell 92. Wasikowski M, Chen XW (2010) Combating
79. Dunn JC (1973) A fuzzy relative of the the small sample class imbalance problem
ISODATA process and its use in detecting using feature selection. IEEE Trans Knowl
compact well-separated clusters. J Cybern Data Eng 22(10):1388–1400
3(3):32–57 93. Forman G, Cohen I (2004) Learning from
80. Chan TF, Vese LA (2001) Active contours little: comparison of classifiers given little train-
without edges. IEEE Trans Image Process ing. In: Knowledge discovery in databases:
10(2):266–277 PKDD 2004. Springer, Berlin, pp 161–172
81. Chan TF, Sandberg BY, Vese LA (2000) 94. Cui YJ, Davis S, Cheng CK, Bai X (2004) A
Active contours without edges for vector- study of sample size with neural network. In
valued images. J Vis Commun Image Proceedings of 2004 international conference
Represent 11(2):130–141 on machine learning and cybernetics, 2004,
82. Ramlakhan K, Shang Y (2011) A mobile vol 6. IEEE, pp 3444–3448
automated skin lesion classification system. In 95. Rezze GG, Sá BCSD, Neves RI (2006)
23rd IEEE international conference on tools Dermoscopy: the pattern analysis. An Bras
with artificial intelligence (ICTAI), 2011. Dermatol 81(3):261–268
IEEE, pp 138–141 96. WHO, World Health Organization: Buruli ulcer
83. Shapiro LG, Stockman GC (2001) Computer disease, Fact sheet N 199, Revised Mar 2007
vision. Prentice Hall, Upper Saddle River, NJ 97. Boleira M, Lupi O, Lehman L, Asiedu KB,
84. Pehamberger H, Steiner A, Wolff K (1987) In Kiszewski AE (2010) Buruli ulcer. An Bras
vivo epiluminescence microscopy of pig- Dermatol 85(3):281–301
mented skin lesions. I. Pattern analysis of pig- 98. Debacker M, Aguiar J, Steunou C, Zinsou C,
mented skin lesions. J Am Acad Dermatol Meyers WM, Guédénon A et al (2004)
17(4):571–583 Mycobacterium ulcerans disease (Buruli
85. Situ N, Yuan X, Zouridakis G, Mullani N ulcer) in rural hospital, Southern Benin,
(2007) Automatic segmentation of skin 1997–2001. Emerg Infect Dis 10(8):1391
lesion images using evolutionary strategy. In 99. Drummond C, Butler JR (2004) Myco
IEEE international conference on image bacterium ulcerans treatment costs, Australia.
processing, 2007 (ICIP 2007), vol 6. IEEE, Emerg Infect Dis 10(6):1038
pp VI–277 100. Kubat M, Holte R, Matwin S (1997)
86. Stollnitz EJ, DeRose AD, Salesin DH (1995) Learning when negative examples abound.
Wavelets for computer graphics: a primer. 1. In: Machine learning: ECML-97. Springer,
IEEE Comput Graph Appl 15(3):76–84 Berlin, pp 146–153
87. Marghoob AA, Swindle LD, Moricz CZ, 101. Leung T, Malik J (2001) Representing and
Sanchez Negron FA, Slue B, Halpern AC, recognizing the visual appearance of materials
Kopf AW (2003) Instruments and new using three-dimensional textons. Int J
technologies for the in vivo diagnosis of Comput Vision 43(1):29–44
melanoma. J Am Acad Dermatol 49(5): 102. Nowak E, Jurie F, Triggs B (2006) Sampling
777–797 strategies for bag-of-features image classifica-
88. Binder M, Schwarz M, Winkler A, Steiner A, tion. In: Computer vision–ECCV 2006.
Kaider A, Wolff K, Pehamberger H (1995) Springer, Berlin, pp 490–503
Epiluminescence microscopy: a useful tool for 103. Lowe DG (2004) Distinctive image features
the diagnosis of pigmented skin lesions for from scale-invariant keypoints. Int J Comput
formally trained dermatologists. Arch Vision 60(2):91–110
Dermatol 131(3):286 104. Parikh D, Zitnick CL, Chen T (2008)
89. Ojala T, Pietikainen M, Maenpaa T (2002) Determining patch saliency using low-level
Multiresolution gray-scale and rotation context. In: Computer vision–ECCV 2008.
invariant texture classification with local Springer, Berlin, pp 446–459
binary patterns. IEEE Trans Pattern Anal 105. Chen XW, Gerlach B, Casasent D (2005)
Mach Intell 24(7):971–987 Pruning support vectors for imbalanced
90. Van De Sande KE, Gevers T, Snoek CG data classification. In Proceedings. 2005
(2010) Evaluating color descriptors for object IEEE international joint conference on neural
and scene recognition. IEEE Trans Pattern networks, 2005 (IJCNN'05), vol 3. IEEE,
Anal Mach Intell 32(9):1582–1596 pp 1883–1888
91. Veropoulos K, Campbell C, Cristianini N 106. Kubat M, Matwin S (1997) Addressing the
(1999) Controlling the sensitivity of sup- curse of imbalanced training sets: one-sided
port vector machines. In Proceedings of the selection. In: Proceedings of the fourteenth
496 George Zouridakis et al.
international conference on machine learn- 112. Kira K, Rendell LA (1992) The feature selec-
ing, vol 97. Morgan Kaufmann, San Mateo, tion problem: traditional methods and a new
CA, pp 179–186 algorithm. In: Proceedings of the tenth
107.
Chawla NV, Bowyer KW, Hall LO, national conference on artificial intelligence.
Kegelmeyer WP (2011) SMOTE: synthetic AAAI Press, Menlo Park, CA, pp 129–134
minority over-sampling technique. arXiv pre-
113. Wolf JA, Moreau JF, Akilov O, Patton T,
print arXiv:1106.1813 English JC, Ho J, Ferris LK (2013) Diagnostic
108. Estabrooks A, Jo T, Japkowicz N (2004) A inaccuracy of Smartphone applications for mel-
multiple resampling method for learning anoma detection. JAMA Dermatol 149(4):
from imbalanced data sets. Comput Intell 422–426
20(1):18–36 114. Chao JT, Loescher LJ, Soyer HP, Curiel-
109. Schölkopf B, Platt JC, Shawe-Taylor J, Smola Lewandrowski C (2013) Barriers to mobile
AJ, Williamson RC (2001) Estimating the teledermoscopy in primary care. J Am Acad
support of a high-dimensional distribution. Dermatol 69(5):821–824
Neural Comput 13(7):1443–1471 115.
Kroemer S, Frühauf J, Campbell TM,
110. Raskutti B, Kowalczyk A (2004) Extreme re- Massone C, Schwantzer G, Soyer HP,
balancing for SVMs: a case study. ACM Hofmann‐Wellenhof R (2011) Mobile tele-
SIGKDD Explor Newslett 6(1):60–69 dermatology for skin tumour screening: diag-
111. Manevitz LM, Yousef M (2002) One-class
nostic accuracy of clinical and dermoscopic
SVMs for document classification. J Mach image tele-evaluation using cellular phones.
Learn Res 2:139–154 Br J Dermatol 164(5):973–979
INDEX
Avraham Rasooly and Keith E. Herold (eds.), Mobile Health Technologies: Methods and Protocols, Methods in Molecular Biology,
vol. 1256, DOI 10.1007/978-1-4939-2172-0, © Springer Science+Business Media New York 2015
497
MOBILE HEALTH TECHNOLOGIES
498 Index
Benzene, toluene, ethylbenzene and xylenes (BTEX) (cont.) Charge-coupled device (CCD)
and VOCs...................................................................203 detector ............................................... 248, 250, 255, 258
wearable detection system...........................................208 device .................................................................. 140, 144
Biotinylated anti-PfHRP2 IgG synthesis...........................78 Chemical sensors ..............................................................202
Blood analysis Circularity index (CIRC) .................................................439
cell-phone based imaging ...................................187–188 Circulating tumor cells (CTCs)
cost-effective translation .............................................172 auxiliary electronics.....................................................131
smart application ................................................181–182 auxiliary fluidics ..........................................................131
static microscopic imaging cancer..........................................................................124
hemoglobin concentration ............................186–188 challenges ...................................................................123
imaging process methods ..............................182–184 clinical applications.....................................................151
optical design ................................................177–180 detection .............................................................133–134
red blood cell density ....................................185–186 drug response ......................................................134–136
smart application ..........................................181–182 electronics ...........................................................128–129
white blood cell density ................................184–185 engineers .....................................................................123
Botulinum neurotoxin A (BoNT-A) ................................252 finite element simulations-fluidic .......................132–133
Buruli ulcer finite element simulations-magnetic...................131–132
automatic detection ............................................487, 488 giant magnetoresistance ..............................................126
skin cancer ..................................................................468 immunomagnetic detection ........................................124
magnetic labeling ........................................................131
C
magnetic nanomaterials ......................................129–130
Camera. See Mobile camera magnetic nanomaterials and micro-Hall detectors ...........126
Cancer detection....................................... 436, 465, 467, 468 μHall chip (see μHall chip)
Capillary μHall sensors .......................125, 126, 128, 129, 132, 133
3D detection systems ..................................................232 microfluidics ...............................................................125
fluorophore .................................................................237 mobile mobile flow cytometer ....................................140
imaging detector .........................................................237 superparamagnetic MNPs ..........................................126
LED ...........................................................................237 Clinical assays, artificial sera samples ...........................96–97
optical amplification ...................................................232 Cloud infrastructures ........................................ 436, 451, 457
Capillary-driven microfluidics ............................................86 CMOS camera. See Complementary metal oxide
Cardiovascular diseases (CVDs) .......................................293 semiconductor (CMOS) camera
CCD. See Charge-coupled device (CCD) Colorimetric method
Cell-phone based diagnostics paper-based mobile platform ..................................42–43
application ..................................................................174 tuberculosis diagnostics.................................................42
blood analysis (see Blood analysis) Colorimetric non-cross-linking assay
chemical reagents........................................................174 gold nanoprobe characterization .............................49–50
cost-effective translation .............................................172 MTBC DNA detection ....................................46, 50–52
flow-cytometry (see Cell-phone based Commercial off-the-shelf instrumentation.
flow-cytometry) See Spectrophotometry, CMOS cameras
imaging cytometer ......................................................189 Complementary metal oxide semiconductor (CMOS)
manual counting .........................................................172 camera.....................................................140, 144
micro-fluidic chip .......................................................172 “iOS Default” .............................................................310
optical components .....................................................174 iPhysioMeter ..............................................................316
opto-fluidic design ......................................................188 LED flash ...................................................................320
opto-fluidic pumping/excitation scheme ....................173 lnNPV values ..............................................................313
PDMS based micro-fluidic chip .................................188 PPG measurement..............................................306, 309
ratio of f/f 2.................................................................188 Computational image enhancement .........................239, 240
telemedicine devices ...................................................172 Computer aided drawing (CAD) .....................................250
Cell-phone based flow-cytometry Computer control .............................................................233
nHD mode .................................................................175 Connected component algorithm .....................................478
optical design ..............................................................175 CTCs. See Circulating tumor cells (CTCs)
and optical microscopy ...............................................172 Cyan magenta yellow key (CMYK) .................................261
SYTO16 solution .......................................................174 Cytometry. See also Mobile flow cytometer
WBC density ......................................................176–177 imaging flow ...............................................................145
Cell phone camera ..............................................................94 POC clinical flow .......................................................151
MOBILE HEALTH TECHNOLOGIES
Index
499
D EEG recording
eSenses, attention and meditation ......................383–384
Deoxyribonucleic acid (DNA) modes .................................................................382–383
amplification components.......................................45, 49 noise frequencies .........................................................378
and control samples ................................................45, 46 physiological and behavioral .......................................381
fluorescent dyes.............................................................35 poor signal ..................................................................382
isolation Electric field
components.......................................................44–45 and flow circulation ......................................................58
extraction with QIAamp DNA mini kit ...........48–49 with and without vibration ...........................................66
sample decontamination .........................................48 Electrocardiograph (ECG)
LAMP ..........................................................................18 ACT ................................................................... 396, 400
Loopamp™ DNA amplification kit .............................21 HRV/PRV ..................................................................322
on-chip isolation ...........................................................25 and iPhysioMeter .......................................................323
Depolymerization .............................................................215 laboratory NIR PPG ..................................................322
Developing world and PPG instruments .................................................321
HRME system ...........................................................422 and QRS complex detection .......................................394
oral and cervical cancer (see Oral and cervical cancer) Electrochemiluminescence (ECL). See Paper microfluidics
Diagnostics. See also Improve lateral-flow immunoassay Electroencephalogram (EEG)
(LFIA) diagnostics ADHD ...............................................................378, 387
health care services .........................................................3 adverse symptoms .......................................................378
plastic microfluidic cassette ............................................6 Ag/AgCl electrodes ....................................................377
point-of-care........................................... 71, 72, 119, 121 brain electrical activity ................................................375
polycarbonate (PC) tubing .............................................6 computerized test .......................................................387
Diaphragm-based Fabry–Perot interferometric disruption, visual processing .......................................378
(DFPI) ....................................................160–162 frontal bands and accommodative lag .........................387
Diblock copolymer synthesis ..............................................76 frontal lobe .................................................................377
Dielectrophoresis (DEP) ..............................................58, 59 Mindset/ThinkGear (see MindSet)
Diffuse reflectance spectroscopy (DRS) recording.............................................................382–384
calibration process ......................................................158 SPSS syntax (see SPSS syntax)
self-calibration channels .....................................160, 163 types of waves .....................................................375–376
unreliable and defective ..............................................168 Electrohydrodynamic (EHD) flow ...............................59, 60
visible wavelength range (VIS-DRS) .................156, 158 Environmental monitoring .......................................201, 207
Digital auscultation Enzymatic
amplification ...............................................................298 amplification .................................................................17
audio playback ....................................................300, 301 cellulose-based FTA membrane ...................................30
AVC ...........................................................................298 detect fluorescent signals ........................................15–16
high pass filter ............................................................298 molecular assays ............................................................16
internal processing ..............................................296, 297 Enzyme-linked immunosorbent assay (ELISA).................58
listening modes ...................................................298–299 capture ..........................................................................93
low pass filter ......................................................296, 298 indirect..........................................................................93
optimal positions, body ...............................................299 μTADs ....................................................................86–88
DMP-pNIPAAm synthesis..........................................77–78 Essential tremor................................................ 359, 362, 371
Dorsolateral prefrontal cortex (DLPFC) ..................377, 378
3D printing technologies, stethoscope F
audio processing and analysis..............................331–332
Fabrication
iPhone (see iPhone)
μTADs ....................................................................89–90
shapeways ...................................................................330
printed microplates .................................................88–89
test and record ............................................................331
Fabrication methods .........................................................234
Dream diary ..................................................... 392, 400–402
FCM. See Fuzzy C Means (FCM)
DRS. See Diffuse reflectance spectroscopy (DRS)
FDM. See Fused deposition modeling (FDM)
Dysplastic nevi .................................................................437
Fiber-optic sensor
E mobile .................................................................160–162
pressure test ................................................................164
ECG. See Electrocardiograph (ECG) smartphone based spectrometers ................................168
EEG. See Electroencephalogram (EEG) Field of view (FOV) .........................................................146
MOBILE HEALTH TECHNOLOGIES
500 Index