Application of Flow Cyto DiagCyto

Embed Size (px)

Citation preview

  • 8/2/2019 Application of Flow Cyto DiagCyto

    1/9

    Application of Flow Cytometry forBiomarker-Based Cervical CancerCells DetectionJian Ling, Ph.D.,1* Urs Wiederkehr, B.S.,2 Spring Cabiness, B.S.,3

    Kenneth R. Shroyer, Ph.D., M.D.,4 and J. Paul Robinson, Ph.D.5

    The Pap test used for cervical cancer screening is subjective,labor-intensive, and has relatively low sensitivity and specificityfor the detection of underlying clinically significant lesions. Theobjective of this study is to develop a biomarker/flow cytometry-

    based approach for cervical cancer screening. Immunofluores-cence technology to quantify cervical cell expression of two bio-markers p16INK4A and Mcm5 was developed and evaluated byboth microcopy and flow cytometry. The capability of using bio-marker/flow cytometry approach to detect rare-event dysplasticcells in a large background of benign epithelial and inflammatorycells was evaluated. The results indicate that flow cytometrycould detect 0.01% dysplastic cells in a background of normalcervical epithelial cells with the combination of the two bio-markers. Thirty-two clinical specimens were used to test the bio-marker/flow cytometry-based approach and the results were com- pared with the liquid-based cervical cytology. The experimentyielded 100% sensitivity and 93% specificity with reference to theliquid-based cervical cytology. This study indicates the promiseof using multi-color fluorescence flow cytometry for biomarker-

    based cervical cancer screening. This molecular-based, poten-tially high-throughput and automated method is expected to pro-vide an alternative/auxiliary means of cervical cancer screening.Diagn. Cytopathol. 2008;36:7684. ' 2008 Wiley-Liss, Inc.

    Key Words: cervical cancer, cancer screening; flow cytometry;cancer markers; rare-event detection

    Cervical cancer is the second most common cancer in

    women, with 500,000 new cases reported each year and

    250,000 deaths worldwide. Eighty percent of the deaths

    occur in developing countries1

    due to the lack of wide-spread screening programs. In developed countries, the

    death rate from cervical cancer has been reduced signifi-

    cantly through the adoption of population-wide screening

    programs. According to the American Cancer Society,2

    the cervical cancer death rate in the U.S. declined 48%

    between 1973 and 1993.

    Current Screening Methods

    The recognized leading tools used in cervical cancer

    screening programs are the Pap smear, pioneered by Dr.

    George Papanicolaou in the 1930s, and liquid-based cervi-

    cal cytology, introduced in the mid 1990s. In both meth-ods, cell specimens are collected by gently scraping the

    surface of the cervix with a sampling device, such as a

    plastic spatula or cytobrush. In the Pap smear, cells from

    the spatula or cytobrush are smeared directly on a slide

    and then fixed and stained using the Papanicolaou stain.

    In liquid-based cervical cytology, the sample is first

    rinsed into a liquid fixation solution to preserve the cells

    and, thin layer or monolayer preparations are prepared

    using density gradient centrifugation or filter membrane

    technology using automated systems. Both Pap smears

    and liquid-based cytology slides are subsequently stained

    using the Papanicolaou stain.

    Cervical cytology slides are initially screened by micro-

    scopic examination by either a cytotechnologist or pathol-

    ogist. Federal regulations require that all potentially

    abnormal specimens be reviewed and diagnosed by a

    qualified pathologist. Slides that are screened as normal,

    however, may be reported without requiring pathologist

    review. Cytologic abnormalities that may reflect underly-

    ing cervical dysplasia or squamous cell carcinoma are

    categorized under the Bethesda 2001 system as atypical

    squamous cells of undetermined significance (ASC-US),

    1Medical System Department, Automation and Data System Division,Southwest Research Institute, San Antonio, Texas

    2Cytolution, Inc., San Jose, California3Applied Physics Division, Southwest Research Institute, San Antonio,

    Texas4

    Department of Pathology, University of Colorado Health ScienceCenter, Denver, Colorado

    5Cytometry Laboratories, Bindley Bioscience Center, Purdue

    University, West Lafayette, IndianaContract grant sponsor: Cytolution Inc.; Contract grant sponsor: NIH

    (National Cancer Institute); Contract grant number: 1R21CA125370-01.*Correspondence to: Jian Ling, Ph.D., Southwest Research Institute,

    6220 Culebra Rd., San Antonio, TX 78238. E-mail: [email protected] 18 June 2007; Accepted 6 October 2007DOI 10.1002/dc.20763Published online in Wiley InterScience (www.interscience.wiley.com).

    76 Diagnostic Cytopathology, Vol 36, No 2 ' 2008 WILEY-LISS, INC.

  • 8/2/2019 Application of Flow Cyto DiagCyto

    2/9

    atypical squamous cells suspicious but not diagnostic for

    a high grade squamous intraepithelial lesion (ASC-H),

    low-grade squamous intraepithelial lesion (LSIL), high-

    grade squamous intraepithelial lesion (HSIL), and squa-

    mous cell carcinoma (SCC).3 Figure 1 illustrates the

    morphological changes that are characteristic of the devel-

    opment of precursor lesions. A general feature of the

    high-grade dysplastic cells is that they typically have high

    nuclear-to-cytoplasmic volume ratios and this ratio

    increases as the severity of the lesion increases (Fig. 1).

    Current guidelines provided by the American Cancer

    Society recommend screening for women 21 year of age

    and older. The preferred screening frequency is annual

    unless there are three consecutive normal, technically sat-

    isfactory Pap tests but is often increased to every 36 mo

    if the Pap test indicates an abnormality.4 About 50 million

    Pap tests are performed each year in the United States

    and about 110 million worldwide.5,6

    Human papillomavirus (HPV) is the main cause of cer-

    vical dysplasia and carcinoma. Although HPV vaccines

    are likely to be highly effective in preventing infection by

    HPV vaccine types, cervical cancer screening programs

    will still play crucially important roles for the detection ofcytologic abnormalities in currently infected patients and in

    the detection of disease associated with nonvaccine types.

    Limitations of Current Screening Methods

    The major challenge for cervical cytology is the need to

    detect rare-events. A liquid-based cervical cytology speci-

    men contains a minimum of 5,000 normal squamous

    cells; most samples contain 50,000 or more normal cervi-

    cal squamous epithelial cells, as well as benign endocervi-

    cal cells, and inflammatory components. High-grade squa-

    mous intraepithelial lesions (HSILs), on the other hand,

    may often be based on the detection of only a very small

    number of abnormal cells, frequently in the range of 10

    100 dysplastic cells/slide.

    Federal guidelines permit cytotechnologists to screen

    up to 100 slides in a normal 8-hr workday.7 Assuming a

    minimum number of 5,000 cells per slide, cytotechnolo-

    gist would review at least 500,000 cells/day and be

    required to detect as few as 1050 dysplastic cells in a

    positive specimen. Since *90% of all cases in most diag-

    nostic practices are negative for cytologic abnormalities,

    most of the screeners time and energy is expended look-

    ing at healthy cells.8 Fatigue and monotony can reduce

    the acuity of the screener and increase the chance that

    rare positive cells could be overlooked.7

    Current methods of cervical cancer screening are not

    only labor-intensive but are also highly subjective and

    have relatively low sensitivity and specificity for the

    detection of some high-grade clinically significant lesions.

    With the liquid-based Pap test, the sensitivity of cervical

    screening has increased to about 80% from the 65% in

    conventional Pap smear,9 resulting in an improvement of

    the overall clinical, economic, and patient outcomes.

    However, the specificity of liquid-based Pap test dropped

    from 95% with conventional Pap smear to about 75%.9

    Recently, the FDA approved the use of high risk HPV

    testing in combination with the liquid-based cervical cy-

    tology for primary screening of women over age 30.

    Biomarkers for Cancer Screening

    With the significant advances in genomics and proteomicsover the last decade, hundreds of articles have been pub-

    lished on the subject of understanding the molecular

    pathogenesis of cervical cancer.10 Molecular changes

    have been recognized to be the earliest indication of cell

    abnormalities. The objective of the current study was to

    develop a method that can achieve a true molecular mea-

    surement using immunofluorescence technology and flow

    cytometry technologies. Such a molecular-based cervical

    cancer screening method is expected to have higher sensi-

    tivity and specificity compared to the current cervical cy-

    tology methods.

    A large number of biomarkers have been identified that

    are overexpressed in cervical cancer cells.11 Some of themarkers that appear to have potential for cervical cancer

    screening include p16INK4A (a cyclin-dependent kinase in-

    hibitor protein), Mcm (minichromosome maintenance) pro-

    teins, Cdc (cell division cycle) proteins, topoisomerase 2

    alpha, PCNA, Ki-67, Cyclin E, p-53, and Rb (retinoblas-

    toma) proteins.10,1215 This report is focused on the analy-

    sis of two of these markers, p16INK4A 1628 and Mcm5.29,30

    p16INK4A

    protein. The p16INK4A protein has been used as

    an immunohistochemical and immunocytochemical marker

    in several studies to detect cervical cancer. In cervical car-

    cinomas, viral DNA integration into the host genome may

    result in disruption of the E2 open reading frame, resulting

    in unregulated overexpression of HPV oncogenes E6 and

    E7, E7-mediated catabolism of pRb, and the reciprocal

    overexpression of p16INK4A.26 Almost 100% of high-grade

    cervical dysplasias and invasive cancers have been shown

    to express very high levels of p16INK4A, whereas normal

    cervical squamous cells do not test positive for

    p16INK4A.31,32 Several studies16,17,2628 have demonstrated

    the successful combination of p16INK4A immunocytochem-

    ical assay with the liquid-based Pap test. In studies per-

    formed by Bibbo et al.,16 very high levels of p16INK4A

    Fig. 1. Morphological changes (the increase of nucleus-to-cytoplasm ra-tio) of precursor lesions of cervical carcinoma.

    BIOMARKER-BASED CERVICAL CANCER CELLS DETECTION

    Diagnostic Cytopathology, Vol 36, No 2 77

    Diagnostic Cytopathology DOI 10.1002/dc

  • 8/2/2019 Application of Flow Cyto DiagCyto

    3/9

    were detected in almost 100% of high-grade cervical dys-

    plasias and invasive cancers, whereas no p16INK4A-positive

    stain was found in normal cervical epithelia using the same

    antibodies. A study performed by Murphy et al.21 also

    reported that p16INK4A identified dysplastic squamous and

    glandular cells of the cervix with a sensitivity of 99.9%

    and a specificity of 100%. A practical limitation to the use

    of p16INK4A as a cytologic diagnostic adjunct, however, is

    that sporadic expression of this marker is also sometimes

    present in scattered benign endocervical glandular cells

    and in tuboendometrial metaplasia of the cervical mucosa,

    which could lead to false positive classification of test

    results.31

    Mcm5 protein. The MCM proteins form a hexameric

    helicase that denatures DNA at the initiation of DNA rep-

    lication.10 Mcm5 has been extensively studied as a marker

    for cellular proliferation expressed during the normal cell

    cycle and recent studies indicate that Mcm5 may be a

    marker for the presence of cervical intraepithelial neopla-

    sia and carcinoma but is also expressed in low grade dys-

    plastic lesions and in some normal proliferating squamous

    cells.21,29,30

    Although Mcm5 may be expressed in a lower proportion

    of high grade dysplastic cells than is typically observed for

    p16INK4A the expression of Mcm5 has not been reported in

    benign endocerrvial glandular cells. Thus, dual (multiplex)

    staining of both Mcm5 and p16INK4A could theoretically

    increase overall test performance because these two bio-

    markers are complementary in nature.

    Flow cytometry for cervical cancer detection. Flow

    cytometry is an ideal format for the analysis of single-cell

    suspensions, quantifying cell structural and molecular fea-tures, and for the detection of rare events. The potential for

    the use of flow cytometry for cervical cancer screening

    began in the 1970s and was widely reported during the

    1980s and early 1990s.3340

    Most of these studies focused

    on methods that use fluorescent dyes to stain nucleic acids

    and use flow cytometry to measure DNA content (aneu-

    ploidy) as a prognostic indicator of solid tumors. However,

    the use of DNA content as an independent prognostic indi-

    cator is uncertain and remains controversial.

    This study took a different approach in the use of flow

    cytometry by including the evaluation of p16INK4A and

    Mcm5 as sensitive and specific markers for the detection

    of cervical dysplasia and carcinoma.

    Materials and Methods

    Sample Preparation

    Control samples. The cervical cancer-derived HeLa cell

    line, which has been shown to overexpress both p16INK4A

    and Mcm5 proteins, was used as the positive control in

    this study. HeLa cells were fixed and preserved with a

    methanol-based fixative (PreservCyt1 solution, Cytyc

    Corp., Marlborough, MA). A previous study41 has shown

    that the PreservCyt1 solution will preserve both cell

    morphology and cellular molecular markers for at least

    30 days. PreservCyt1 solution is also known to permeab-

    ilize cells so that fluorochromes-labeled antibodies can

    penetrate cells.

    Clinical cervical specimens. Residual cervical cytology

    specimens from PreservCyt1 vials were obtained from

    the cytopathology laboratories at the University of Texas

    Health Science Center at San Antonio and the University

    of Colorado Health Sciences Center at Denver, following

    IRB application approval of the study protocol. These

    specimens have been reviewed by experienced cytopatho-

    logists and classified by Bethesda 2001 terminology as

    negative, ASC-US, LSIL, or HSIL. The clinical

    specimens were filtered with 70-lm nylon mesh filter to

    remove cell clusters before flow cytometry measurement.

    Fluorescence Labeling

    Antibodies and conjugation with fluorochrome. Mousemonoclonal antibodies to p16INK4A (Clone ZJ11) and

    Mcm5 (Clone CRCT5.1) from Labvison Inc. (Fremont,

    CA) were selected in the study. These two antibodies

    were directly conjugated with PE and APC fluorochromes

    using commercially available labeling kits (ProZyme Inc.,

    San Leandro, CA). The conjugates were denoted as

    p16INK4A-PE and Mcm5-APC antibodies. Corresponding

    mouse IgG1 and IgG2b isotypes were also obtained and

    conjugated to PE and APC, respectively, as the isotype

    control.

    Immunofluorescence staining. Before staining, a sample

    was washed twice with phosphate buffered saline (PBS)

    to remove the fixation solution. The second wash used astaining buffer (PBS plus 1% bovine serum albumin

    (BSA) and 0.01% sodium azide) to block the intracellu-

    lar nonspecific binding sites. The sample was concen-

    trated to 100 lL and then simultaneously stained with a

    cocktail of p16INK4A-PE and Mcm5-APC antibodies. In

    immunofluorescence imaging, 1 lg/mL concentration of

    antibody was used to stain the samples. In flow cytome-

    try, the optimal antibody concentration was about 0.1

    0.25 lg/mL. Flow cytometry is more sensitive for detec-

    tion of the fluorescence signal owing to the use of a

    laser as the excitation source and a photomultiplier tube

    (PMT) as the detector.

    The staining tube was kept on ice or in a 48C dark re-frigerator for 30 min. Then the stained cells were washed

    twice with the staining buffer to remove the unbound con-

    jugates. The same procedure and same concentration were

    followed for isotype staining.

    Quantitative Microscopy

    Before performing the flow cytometry experiment, micro-

    scopic imaging was performed to (1) verify the effective-

    ness of the fluorescence stain, and (2) verify whether the

    LING ET AL.

    78 Diagnostic Cytopathology, Vol 36, No 2

    Diagnostic Cytopathology DOI 10.1002/dc

  • 8/2/2019 Application of Flow Cyto DiagCyto

    4/9

    overexpression of biomarkers is correlated to the abnor-

    mal morphology of dysplastic cervical cells. A Nikon

    Eclipse TE2000E inverted microscope and computer sys-

    tem was used for the fluorescence imaging. Three fluores-

    cence filters in the FITC, PE, and APC bands (i.e., the

    530-nm, 575-nm, and 660-nm emission bands) were used.

    During imaging, the microscope was first set in differen-

    tial-interference-contrast (DIC) video mode and visually

    focused on an imaging area which contained multiple

    nonoverlapping cells. Then, four images (three fluores-

    cence images in the FITC, PE, APC bands, and a DIC

    image) were obtained for each imaging area. The DIC

    image illustrates the morphology of the cells. The PE and

    APC images show the expression of p16INK4A and Mcm5

    markers in the cells. The FITC image measures cell auto-

    fluorescence in the FITC band, which is used to correct

    the autofluorescence in the PE and APC bands on a cell-

    by-cell basis (see Data Analysis below).

    Flow Cytometry

    A FACS Aria flow cytometer (Becton-Dickinson, San

    Jose, CA) and an FC 500 flow cytometer (Beckman-

    Coulter, Miami, FL) were used in the flow experiments.

    Five parameters were measured: forward-scatter (FS), side

    scatter (SS), and FITC, PE, and APC fluorescence bands.

    The FS and SS measurements were used to gate out cell

    debris. The cell autofluorescence measured in the FITC

    band was used to correct the autofluorescence in the PE

    and APC bands on a cell-by-cell basis using a postcompen-

    sation method (see Discussion). The remaining fluores-

    cence measured in the PE and APC bands reflects the

    expression levels of biomarkers p16INK4A

    and Mcm5,respectively, in each cell. Before each flow experiment,

    the flow cytometer was calibrated using fluorescence beads

    to minimize the day-to-day variation of optics. The calibra-

    tion procedure ensured the measurement of different

    samples under similar conditions.

    Data Analysis

    For imaging data, a MATLAB program was developed

    to quantitatively compare cell-to-cell average stain inten-

    sities in fluorescent images. The software automatically

    segments the fluorescent images to locate individual

    cells. The average fluorescence intensities or the fluores-

    cence density in the FITC, PE, and APC bands were

    determined for each individual cell. The fluorescence

    density, calculated by normalizing the total staining in-

    tensity by cell area, provided a fair comparison of the

    biomarker expression among different types of cervical

    cells, which usually have large variation in size (from 25

    to 65 lm in diameter).

    For flow cytometry data, FCS Express (De Novo Soft-

    ware, Thornhill, Canada) was used to perform gating

    and autofluorescence correction. Fluorescence pulse peak

    instead of pulse integral was used to represent the fluores-

    cence density or biomarker expression of each cell

    because pulse peak is not significantly affected by cell

    size, as is the pulse integral (see Discussion).

    Results

    Microscopy Imaging Experiment

    Comparison of antibody stain and isotype stain. Fixed

    HeLa cells were stained with a cocktail of p16INK4A-PE

    and Mcm5-APC antibodies. Matched aliquots of fixed

    HeLa cells were stained with a cocktail of PE and APClabeled isotypes. The dot plot in Figure 2 shows that the

    antibody-stained cells (denoted by light quadrangular

    symbol) have significantly higher stain intensities in both

    the PE and APC bands than that of the isotype-stained

    cells.

    Comparison of normal and dysplastic cervical cells.

    Eleven cervical cytology specimens, including five nega-

    tive and six positive specimens (1 ASC-US, 2 LSIL, and

    3 HSIL), were used in a pilot imaging study. Each speci-

    men was divided into two parts. One part was unstained

    and used to establish autofluorescence compensation coef-

    ficients, and the other part was stained with the cocktail

    of p16INK4A-PE and Mcm5-APC antibodies. About

    70 cells, including cells with differing morphology, were

    imaged for each specimen. The average fluorescence

    intensities were computed for each cell in imaging areas.

    Figure C-1 gives an example of the DIC and fluorescence

    images of a normal and an abnormal HSIL cell from one

    of the HSIL cervical specimens. The autofluorescence in-

    tensity of the cells in the PE and APC band has been

    eliminated based on their autofluorescence intensity in the

    FITC band. Figure C-1 indicates that the average PE and

    Fig. 2. PE vs. APC fluorescence intensities of the HeLa cells stained withcocktail of isotypes (dark triangular symbol) and stained with cocktail ofp16

    INK4A-PE and Mcm5-APC antibodies (light quadrangular symbol).

    BIOMARKER-BASED CERVICAL CANCER CELLS DETECTION

    Diagnostic Cytopathology, Vol 36, No 2 79

    Diagnostic Cytopathology DOI 10.1002/dc

  • 8/2/2019 Application of Flow Cyto DiagCyto

    5/9

    APC intensities of the HSIL cell are significantly higher

    than those of the normal cells.

    The experiment also shows that most cells in the eleven

    specimens had low stain intensities in both the PE and

    APC bands. Only a small number of cells in the ASC-US,

    LSIL, and HSIL specimens had high stain intensities.

    This suggests that the biomarker overexpressed cells are

    rare-events, which is similar to the morphology-based

    Fig. C-1C-3. Fig. C-1. DIC (upper left), FITC (upper right), PE (lower left) and APC (lower right) images of a normal cell (a), and a HSIL dysplas-tic cell (b), from a HSIL cervical specimen. The numbers under the cells are the average fluorescence intensities of the cells. Fig. C-2. The dot plot(left) illustrates HeLa cells (red dots) identified and separated from normal cervical cells (blue dots) after staining with p16

    INK4A-PE and Mcm5-APC

    antibodies. The scatter plot (right) indicates the linear relationship between the number of spiked and the number of p16INK4A and Mcm5 positiveHeLa cells. Fig. C-3. Dot plots of PE (P16INK4A) vs. APC (Mcm5) immunofluorescence intensities of the cells in a negative (left) and a positive HSILcervical specimen (right). Each plot contains about 75,000 cervical cells.

    LING ET AL.

    80 Diagnostic Cytopathology, Vol 36, No 2

    Diagnostic Cytopathology DOI 10.1002/dc

  • 8/2/2019 Application of Flow Cyto DiagCyto

    6/9

    detection. The experiment also suggested that the overex-

    pression of both the p16INK4A and Mcm5 biomarkers is

    closely related to the abnormality of cell morphology.

    Flow Cytometry Experiment

    Spiking experiment. This experiment was conducted toshow the feasibility of biomarker-based flow cytometry in

    the detection of rare cervical cancer cells among a large

    number of normal cells in a cervical specimen. Aliquots

    of normal cells were taken from six Pap test negative

    specimens and combined to create a normal cervical cell

    pool. The normal cells were counted and divided among

    seven tubes, with each tube containing about 77,000 be-

    nign squamous cells. One tube was left unstained for ref-

    erence. The other six tubes were spiked with 10, 20, 50,

    100, 500, and 1,000 HeLa cells using the serial dilution

    method. The spiked samples were stained with the cock-

    tail of p16INK4A-PE and Mcm5-APC antibodies.

    The dot plot in the left panel of Figure C-2 illustratesan example of the PE versus APC intensities in the 500-

    HeLa-spiked sample. The HeLa cells highly stained with

    p16INK4A and Mcm5 (red dots) are identified and sepa-

    rated from the normal cervical cells (blue dots). The scat-

    ter plot in the right panel of Figure C-2 illustrates the lin-

    ear relationship between the number of spiked and the

    number of detected HeLa cells. The discrepancy between

    the detected cells and the number of spiked cells could be

    due to the following reasons: (1) the number of HeLa

    cells that were actually added into each sample varied;

    (2) cells were lost during the poststaining washing steps;

    (3) a small portion of the samples could not be measured

    due to the dead space between pipette and test tube; and(4) some HeLa cells did not have a high staining of

    p16INK4A and Mcm5.

    This study indicates that it is possible to use flow

    cytometry to detect as low as 0.01% cancer cells among a

    large number of normal cervical cells. The outcome

    exceeded the expectation of detecting less than 0.1%

    abnormal cells among normal cells, which is considered

    by pathologists as being an acceptable limit for a cervical

    cancer screening method.

    Clinical specimens experiment. Thirty-two residual cer-

    vical specimens from routine cervical cancer screening

    were involved in this study. They were categorized as 15

    negative and 17 positive (2 ASC-US, 1 LSIL, and 14

    HSIL) cases. Each specimen was split into two aliquots.

    One aliquot was unstained and used to measure the cell

    autofluorescence in the three fluorescence bands: FITC,

    PE, and APC. The other aliquot, containing around 75,000

    cells, was stained with a cocktail of p16INK4A-PE and

    Mcm5-APC antibodies and then run on the flow cytometer.

    Figure C-3 shows two dot plots of a negative and a

    positive (HSIL) specimen generated from flow measure-

    ment. These two plots clearly show that the HSIL speci-

    men has significantly more cells with high intensities in

    both PE and APC bands than the negative specimen. The

    high intensity in the PE and APC bands indicates that

    both biomarkers p16INK4A and Mcm5 are overexpressed.

    The detection threshold was set arbitrarily in this

    experiment to maximize the separation between negative

    and positive (ASC-US) specimens. In Table I, the clas-

    sification of the thirty-two specimens determined by multi-

    parameter flow cytometry is compared with the classifica-

    tion by liquid-based Pap test. Using the Pap test as the

    reference, the sensitivity and specificity of the flow

    cytometry method to classify cervical specimens into

    negative and positive (ASC-US) was 100 and93%, respectively.

    Discussion

    Autofluorescence Compensation

    One of the major challenges in the use of flow cytometry

    to measure biomarker expression of cervical cancer cells

    is the autofluorescence problem. Fixed cervical cells have

    very strong autofluorescence. Figure 3a shows a flow

    cytometry plot of FITC versus PE of an unstained sample

    that was mixed with HeLa and normal cervical cells. The

    autofluorescence is present not only in the green band but

    also in the yellow and even in red frequency bands. Inaddition, the cell-to-cell autofluorescence intensities can

    vary over a 1,000-fold range in a specimen. Without cor-

    rection for such a large variance of autofluorescence, it is

    impossible to detect biomarker signals in PE and APC

    bands. This is illustrated in Figures 3b and c, the dot plot

    of the same sample but stained with p16INK4A-PE and

    Mcm5-APC antibodies. These two figures indicate that

    the PE and APC staining intensities of HeLa cells are

    much lower than some of the normal cells that have high

    autofluorescence in the same bands. Although cell-to-cell

    autofluorescence has large variation, the dot plots suggest

    that the intensities between the autofluorescence in the

    yellow or red band and the autofluorescence in the green

    band are linearly correlated among different cells. Using

    this feature, the cell autofluorescence measured in the

    FITC band (from the samples not stained with FITC

    dyes) can be used to correct the autofluorescence in the

    PE and APC bands on a cell-by-cell basis using a post-

    compensation method. The ratios between the autofluores-

    cence in PE or APC bands versus that in FITC band can

    be determined beforehand from the unstained specimen.

    Figure 3d illustrates the data in Figure 3b and c after

    Table I. Comparison of the Classification of 32 Specimens BetweenFlow Cytometry and Pap Test

    Positive in Pap test Negative in Pap test

    Positive in flow cytometry TP 17 FP 1Negative in flow cytometry FN 0 TN 14

    BIOMARKER-BASED CERVICAL CANCER CELLS DETECTION

    Diagnostic Cytopathology, Vol 36, No 2 81

    Diagnostic Cytopathology DOI 10.1002/dc

  • 8/2/2019 Application of Flow Cyto DiagCyto

    7/9

    autofluorescence compensation. HeLa cells are clearly

    separated from normal cervical cells based on PE and

    APC staining intensities.

    The Measurement of Cervical Cell FluorescenceIntensity in Flow Cytometry

    Another major challenge in the study, which is usually

    not involved in the hematological application of flow

    cytometry, is how to correctly measure the biomarker

    expression levels in flow cytometry when cells of large

    different sizes are mixed together. Cervical epithelial cells

    in a sample can vary from 25 to 65 lm in size. The fluo-

    rescence pulse integral (or area), often used in flow

    cytometry to measure fluorescence intensity, is not appro-

    priate in this case to compare the biomarker expression

    (indicated by fluorescence dyes per unit volume or fluo-

    rescence density) among cells of different sizes. For

    example, the pulse integral of a small-size cell with high

    biomarker expression may be smaller than a large-size

    cell but with low biomarker expression. In this study,

    pulse peak was used to estimate the fluorescence density

    instead of pulse integral. As the size (2565 lm) of cervi-

    cal cells is larger than the height (9 lm) of the excitation

    laser beam in the flow cytometer, the pulse integral is

    more significantly affected by cell size than the pulse

    peak. The flow cytometry applications to large-size epi-

    thelial cells are different from the applications to blood

    cells, which are usually smaller than or comparable to the

    excitation laser beam. A better way of estimating fluores-

    cence density is to normalize pulse integral by pulse

    width (or time-of-flight).42 However, pulse width mea-

    surement from most commercially available flow cytome-

    ters has a large variance. A slit-scanning system with

    small focal spot size43 might provide a reliable measure-

    ment of pulse width. A system with Coulter volume mea-

    Fig. 3. (a) Dot plots of FITC vs. PE for an unstained cervical sample that was mixed with HeLa and normal cervical cells. ( b, c) FITC vs. PE andFITC vs. APC of the same sample in (a) but stained with p16

    INK4A-PE and Mcm5-APC antibodies. HeLa cells were separated from normal cells

    because of the additional stain intensity onto the autofluorescence intensity. (d) PE vs. APC of the stained sample after the autofluorescence in PE andAPC bands were compensated. HeLa cells, with high stain intensity in both PE and APC bands, were clearly separated from normal cells.

    LING ET AL.

    82 Diagnostic Cytopathology, Vol 36, No 2

    Diagnostic Cytopathology DOI 10.1002/dc

  • 8/2/2019 Application of Flow Cyto DiagCyto

    8/9

    surement may also be used to obtain a better estimation

    of fluorescence density. How to modify flow cytometer

    for epithelial cells analysis is still a topic needs to be

    investigated. As pointed out by Dong et al., it is a crucial

    step to obtain an optimal flow cytometry setting suitable

    for analysis of epithelial cells.44

    Summary and Future Research

    This study demonstrated the feasibility of (1) using multi-

    plex detection of p16INK4A and Mcm5 to detect dysplastic

    cervical cells by immunofluorescence, (2) using multipara-

    meter flow cytometry to detect rare-event dysplastic cells

    from large background of normal cells, and (3) using mul-

    tiparameter flow cytometry to identify positive cervical

    specimens. Although the results were based on a limited

    number of clinical specimens, this experiment demon-

    strated the promise of using multiparameter flow cytometry

    for biomarker-based cervical cancer screening. This molec-

    ular-based, potentially high-throughput and automated

    method is expected to provide an alternative/auxiliary

    means of cervical cancer screening. The method developed

    for cervical cancer screening in this study can be extended

    to the diagnosis of other nonhematological cancer.

    Future studies will make this technology more robust.

    First, the threshold or gating to detect dysplastic cells was

    arbitrarily set in this preliminary study. A cell sorting and

    validation experiment is needed to optimize the threshold

    setting and to reduce the false negatives and false positives

    in the detection of abnormal cells. Second, the fluorescence

    contrast between the biomarker positive and negative cells

    needs to be enhanced, especially for the p16INK4A

    bio-

    marker. Third, cervical cells tend to cluster together. Toprovide enough cells in single suspension for flow cytome-

    try measurement, sample preparation must include a proce-

    dure of cluster desegregation. The potential solutions to

    these problems will be investigated in future studies.

    Acknowledgment

    We thank Elizabeth Branch for her assistance in the prep-

    aration of this manuscript.

    References1. Cancer Mondial 20002003, World Health OrganizationInterna-

    tional Agency for Research on Cancer. Available at http://www.dep.iarc.fr.

    2. Cancer Facts and Figures 19972003, American Cancer Society.

    Available at http://www.cancer.org/docroot/STT/STT_0.asp.

    3. DeMay RM. Practical principles of cytopathology. Chicago: Ameri-

    can Society Clinical Pathology Press; 1999.

    4. Cervical Cancer Home Page, National Cancer Institute. Available at

    http://www.cancer.gov/cancer_information/cancer_type/cervical/.

    5. What tests are used to screen and diagnose cervical cancer. Univer-

    sity of Maryland Medicine, 2004. Available at http://www.umm.edu/patiented/articles/what_tests_used_screen_diagnose_cervical_cancer_000046_8.htm.

    6. Cytyc Corporation 2002, Annual Report 2002. Available at http://

    ir.cytyc.com/downloads/cytc02.pdf.

    7. Ducatman BS, Wang HH, The pap smear: Controversies in practice.

    London: Arnold Publisher; 2002.

    8. Simon H, Cannistra SA. Cervical Cancer, A.D.A.M. Inc., 2003.

    Available at http://www.healthandage.com/html/well_connected/pdf/doc46.pdf.

    9. Preventing and controlling cervical cancer in the new millennium.In: EUROGIN, Paris; 2003.

    10. Baldwin P, Laskey R, Coleman N. Translational approaches toimproving cervical screening. Nat Rev Cancer 2003;3:217226.

    11. Santin AD, Zhan FH, Bignotti E, et al. Gene expression profiles ofprimary HPV16- and HPV18-infected early stage cervical cancersand normal cervical epithelium: Identification of novel candidatemolecular markers for cervical cancer diagnosis and therapy. Virol-ogy 2005;331:269291.

    12. Davidson B, Goldberg I, Lerner-Geva L, et al. Expression of topo-isomerase II and Ki-67 in cervical carcinomaClinicopathologicalstudy using immunohistochemistry. APMIS 2000;108:209.

    13. Tae KY, Kyoung CE, Hoon CN, et al. Expression of cyclin E andp27(KIP1) in cervical carcinoma. Cancer Lett 2000;153:4150.

    14. Shroyer KR, Homer P, Heinz D, et al. Validation of a novel immu-nocytochemical assay for topoisomerase II-alpha and minichromo-some maintenance protein 2 expression in cervical cytology. Cancer2006;108:324330.

    15. von Knebel DM. Biomarkers in screening of cervical cancer. In:Monsonego J, editor. Emerging issues on HPV infections: From sci-ence to practice. Basel: Karger; 2006. p 119.

    16. Bibbo M, Klump WJ, DeCecco J, et al. Procedure for immunocyto-chemical detection of P16INK4A antigen in thin-layer, liquid-basedspecimens. Acta Cytol 2002;46:2529.

    17. Bibbo M, DeCecco J, Kovatich AJ. P16INK4A as an adjunct test inliquid-based cytology. Anal Quant Cytol Histol 2003;25:811.

    18. Harris CP, Lu XY, Narayan G, et al. Comprehensive molecularcytogenetic characterization of cervical cancer cell lines. GenesChromosomes Cancer 2003;36:233241.

    19. Klaes R, Friedrich T, Spitkovsky D, et al. Overexpression ofp16(INK4A) as a specific marker for dysplastic and neoplastic epi-thelial cells of the cervix uteri. Int J Cancer 2001;92:276284.

    20. Klaes R, Benner A, Friedrich T, et al. p16INK4a immunohisto-chemistry improves interobserver agreement in the diagnosis of cer-vical intraepithelial neoplasia. Am J Surg Pathol 2002;26:13891399.

    21. Murphy N, Ring M, Killalea AG, et al. p16INK4A as a marker forcervical dyskaryosis: CIN and cGIN in cervical biopsies and Thin-Prep smears. J Clin Pathol 2003;56:5663.

    22. Nuovo GJ, Plaia TW, Belinsky SA, et al. In situ detection of thehypermethylation-induced inactivation of the p16 gene as an earlyevent in oncogenesis. Proc Natl Acad Sci USA 1999;96:1275412759.

    23. Park JS, Dong SM, Kim HS, et al. Detection of p16 gene alterationin cervical cancer using tissue microdissection and LOH study. Can-cer Lett 1999;136:101108.

    24. Pientong C, Ekalaksananan T, Swadpanich U, et al. Immunocyto-

    chemical detection of p16INK4a protein in scraped cervical cells.Acta Cytol 2003;47:616623.

    25. Riethdorf L, Riethdorf S, Lee KR, et al. Human papillomaviruses,expression of p16, and early endocervical glandular neoplasia. HumPathol 2002;33:899904.

    26. Saqi A, Pasha TL, McGrath CM, et al. Overexpression ofp16INK4A in liquid-based specimens (SurePath) as marker of cervi-cal dysplasia and neoplasia. Diagn Cytopathol 2002;27:365370.

    27. von Knebel Doeberitz M. New molecular tools for efficient screen-ing of cervical cancer. Dis Markers 2001;17:123128.

    28. von Knebel Doeberitz M. New markers for cervical dysplasia tovisualise the genomic chaos created by aberrant oncogenic papillo-mavirus infections. Eur J Cancer 2002;38:22292242.

    BIOMARKER-BASED CERVICAL CANCER CELLS DETECTION

    Diagnostic Cytopathology, Vol 36, No 2 83

    Diagnostic Cytopathology DOI 10.1002/dc

  • 8/2/2019 Application of Flow Cyto DiagCyto

    9/9

    29. Laskey R. Initiation of DNA replication in normal and neoplasticcells. Presented at Fifth congress of the European haematologyassociation, Birmingham, UK, 2000.

    30. Williams GH, Romanowski P, Morris L, et al. Improved cervicalsmear assessment using antibodies against proteins that regulateDNA replication. Proc Natl Acad Sci USA 1998;95:1493214937.

    31. Tringler B, Gup CJ, Shroyer KR, Correlation between p16 and pRb

    expression in cervical squamous and glandular neoplasia. HumanPathol 2004;35:689696.

    32. Willman JH, Heinz D, Golitz LE, et al. Correlation of p16 and pRbexpression with HPV detection in Bowens disease. J CutaneousPathol 2006;33:629633.

    33. Nishiya I, Kikuchi T, Moriya S, et al. Cytophotometric study ofpremalignant and malignant cells of the cervix in an approachtowards automated cytology. Acta Cytol 1977;21:271275.

    34. Watts KC, Husain OA, Campion MJ, et al. Quantitative DNA anal-ysis of low grade cervical intraepithelial neoplasia and human papil-lomavirus infection by static and flow cytometry. Br Med J (ClinRes Ed). 1987;295:10901092.

    35. Wheeless LL, Patten SF, Berkan TK, et al. Multidimensional slit-scan prescreening system: Preliminary results of a single blind clini-cal study. Cytometry 1984;5:18.

    36. Titley I, Tee DE, Driver M, et al., Can flow cytometry reduce the

    workload for cervical screening? The results of a series of 622specimens. Cytopathology 1991;2:193203.

    37. Magtibay PM, Perrone JF, Stanhope CR, et al. Flow-cytometricDNA analysis of early stage adenocarcinoma of the cervix. GynecolOncol 1999;75:2024.

    38. Bocking A, Motherby H, Assessment of cervical dysplasia withDNA image cytometry. Pathologe 1999;20:2533.

    39. van Leeuwen AM, Ploem-Zaaijer JJ, Pieters WJ, et al. The suitabil-ity of DNA cytometry for the prediction of the histological diagno-

    sis in women with abnormal cervical smears. Br J Obstet Gynaecol1996;103:359365.

    40. Kimmig R, Wimberger P, Kapsner T, et al. Flow cytometric DNAanalysis using cytokeratin labeling for identification of tumor cellsin carcinomas of the breast and the female genital tract. Anal CellPathol 2001;22:165178.

    41. Lin WM, Ashfaq R, Michalopulos EA, et al. Molecular Papanico-laou tests in the twenty-first century: Molecular analyses with fluid-based Papanicolaou technology. Am J Obstet Gynecol 2000;183:3945.

    42. Shapiro H. Beam geometry and pulse characteristics. In: Practicalflow cytometry, 4th ed. New Jersey: Wiley; 2003. p 183184.

    43. Shapiro H. Intrinsic cellular parameters. In: Practical flow cytome-try, 4th ed. New Jersey: Wiley; 2003. p 285293.

    44. Dong HP, Holth A, Berner A, Davidson B, Risberg B. Flow cyto-metric immunphenotyping of epithelial cancer cells in effusions

    Technical considerations and pitfalls. Cytometry Part B 2007:72B:332343.

    LING ET AL.

    84 Diagnostic Cytopathology, Vol 36, No 2

    Diagnostic Cytopathology DOI 10.1002/dc