WO2001004828A1 - Automated detection of objects in a biological sample - Google Patents

Automated detection of objects in a biological sample Download PDF

Info

Publication number
WO2001004828A1
WO2001004828A1 PCT/US2000/019046 US0019046W WO0104828A1 WO 2001004828 A1 WO2001004828 A1 WO 2001004828A1 US 0019046 W US0019046 W US 0019046W WO 0104828 A1 WO0104828 A1 WO 0104828A1
Authority
WO
WIPO (PCT)
Prior art keywords
interest
slide
image
candidate object
stage
Prior art date
Application number
PCT/US2000/019046
Other languages
French (fr)
Inventor
Gina Mclaren
Bob Ellis
Original Assignee
Chromavision Medical Systems, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chromavision Medical Systems, Inc. filed Critical Chromavision Medical Systems, Inc.
Priority to JP2001510159A priority Critical patent/JP2003504627A/en
Priority to AU60934/00A priority patent/AU6093400A/en
Priority to EP00947299A priority patent/EP1203343A4/en
Publication of WO2001004828A1 publication Critical patent/WO2001004828A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N1/00Sampling; Preparing specimens for investigation
    • G01N1/28Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
    • G01N1/30Staining; Impregnating ; Fixation; Dehydration; Multistep processes for preparing samples of tissue, cell or nucleic acid material and the like for analysis
    • G01N1/31Apparatus therefor
    • G01N1/312Apparatus therefor for samples mounted on planar substrates
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06VIMAGE OR VIDEO RECOGNITION OR UNDERSTANDING
    • G06V20/00Scenes; Scene-specific elements
    • G06V20/60Type of objects
    • G06V20/69Microscopic objects, e.g. biological cells or cellular parts

Definitions

  • the invention relates generally to light
  • microscopy and, more particularly, to automated light
  • biological specimen such as bone marrow, lymph nodes,
  • peripheral blood cerebrospinal fluid, urine,
  • method of cell specimen preparation is to react a
  • nucleic acid which is reactive with a component of
  • the cells of interest such as tumor cells.
  • reaction may be detected using an enzymatic reaction
  • alkaline phosphatase such as alkaline phosphatase or glucose oxidase or
  • HER2 (neu) is a growth factor receptor and
  • hybridization methods are typically used for HER2/neu
  • rare events e.g., finding one tumor cell in a
  • the present invention provides a method
  • a slide carrier which preferably holds
  • the slide carriers are loaded into an
  • the system automatically locates a scan area for each slide during slide processing.
  • magnification image is acquired and processed to
  • a high magnification image is
  • control slides including positive
  • candidate objects of interest for a slide may be
  • cytotechnologist may view the mosaic or may also
  • the mosaic may be stored on magnetic media for future
  • invention preferably includes the steps of
  • thresholded image to determine the presence of one or
  • the scan area is automatically determined by scanning
  • the derived focal plane enables subsequent
  • the focal plane is determined by
  • a focal plane across the array Preferably, a focal plane
  • acquired image is calculated to form a set of
  • the region of interest is a region of interest
  • n preferably n columns wide, where n is a power of 2.
  • the pixels of this region are then processed using a
  • a slide is mounted onto a
  • the slide carrier is positioned in an input
  • the invention is a HER2/neu expression in tissue.
  • tissue especially breast tissue.
  • FIG. 1 is a perspective view of an apparatus for
  • Fig. 2 is a block diagram of the apparatus shown
  • Fig. 3 is a block diagram of the microscope
  • Fig. 4 is a plan view of the apparatus of Fig. 1
  • Fig. 5 is a side view of a microscope subsystem
  • Fig. 6a is a top view of a slide carrier for use
  • Fig. 6b is a bottom view of the slide carrier of
  • Fig. 7a is a top view of an automated slide
  • Fig. 7b is a partial cross-sectional view of the
  • Fig. 8 is an end view of the input module of the
  • Fig. 9a-9d illustrate the output operation of
  • Fig. 10 is a flow diagram of the procedure for
  • Fig. 11 shows the scan path on a prepared slide
  • Fig. 12 illustrates an image of a field acquired
  • Fig. 13A is a flow diagram of a preferred embodiment
  • Fig. 13B is a flow diagram of a preferred embodiment
  • Fig. 14 is a flow diagram of a procedure for
  • Fig. 15 shows an array of slide positions for
  • Fig. 16 is a flow diagram of a procedure for
  • Fig. 17A is a flow diagram of an overview of the
  • Fig. 17B is a flow diagram of a procedure for
  • Fig. 18 is a flow diagram of a procedure for
  • Fig. 19 is a flow diagram of a procedure for
  • Fig. 20 is a flow diagram of a procedure for
  • Fig. 21 is a flow diagram of a procedure for
  • Fig. 22 illustrates a mosaic of cell images
  • Fig. 23 is a flow diagram of a procedure for
  • Fig. 24 illustrates the apparatus functions
  • the invention provides an automated tissue image
  • proliferative disorder for example those associated with
  • MIB-I Microvessel density analysis, the oncogene
  • nucleoli HIV p24
  • HPV human papiloma virus
  • mitotic index generally any combination
  • ACISTM Cellular Imaging System
  • a worklist is a group of
  • a specimen group consists of
  • H/E staining protocol is compatible with
  • the specimen may
  • third antibody i.e., patient negative
  • This calibration consists of radiometric
  • the output of the worklist is a stained
  • a cellular specimen (a "sample") is split to
  • sample provides two or more subsamples.
  • sample provides two or more subsamples.
  • cellular material derived from a subject includes cellular material derived from a subject.
  • Such samples include but are not limited to hair,
  • tissue refers to a mass of connected cells (e.g.,
  • biological fluid refers to liquid material derived from a
  • sample also includes media containing isolated
  • reaction may be determined by one skilled in the art
  • the HER2/neu method uses an anti-HER2/neu
  • kit like that provided by DAKO (Carpinteria, CA) .
  • slide includes a cover-slip medium to protect the
  • coverslip covers the entire prepared specimen.
  • control slides are run with every worklist. The set
  • H/E hematoxylin/eosin
  • the staining procedure is as follows: (1) Bring
  • the invention also provides automated methods
  • the estrogen and progesterone receptors The estrogen and progesterone receptors,
  • immunohistochemistry system such as that provided by
  • a micrometastases is
  • metastatic recurring disease is similar to
  • micrometastasis but is detected after cancer therapy
  • MIB-1 is an antibody that can be used in
  • the clinical stage at first presentation is related
  • metastasis death from neoplasia, low disease-free
  • Angiogenesis is a complex regional vascular endothelial fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblast fibroblasts, hematoma, hematoma, hematoma, hematoma, hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hematoma hem
  • Intratumor microvessel density can be assessed by anti-CD34
  • malignancies including neoplasms of breast, colon,
  • p53 is performed using an anti-p53 antibody, for
  • a nucleoli is an
  • Cervical dysplasia Cervical dysplasia
  • dysplastic cells are of the same kind but of a lesser amount
  • HIV HIV p24 antigen levels
  • the HIV p24 protein test is used for the
  • the test can be used to detect the virus, measure the amount of virus, and
  • HPV Human Papiloma Virus
  • HPV transmitted virus which causes warts .
  • the patient data entry interface is
  • HER2/neu is composed of tissue samples that are
  • One slide is prepared with the HER2/neu
  • worklist is the: (1) worklist name or identifier of protocol (e.g., HER2/neu) ; barcode of the positive
  • Case is the: (4) patient ID; (5) accession number;
  • the input hopper is composed of all the
  • the system reads industry standard barcodes .
  • the barcode relates the type of protocol the
  • the HER2-positive application (1) finds the
  • the HER2-negative application (1) finds the
  • the HER2 application (1) finds the specimen, (2)
  • the Slide Display Interface displays the list of
  • the set of patient slides is a
  • H/E image is
  • pathologist determines whether or not the cancer is
  • This image is displayed. This image can be reduced to
  • the pathologist can have the features available
  • the pathologist has the following capabilities: (1)
  • pathologist has the following capabilities: (1) to
  • a User Interface lists all the patient cases that have already been reviewed and analyzed by
  • the pathologist can choose one or
  • the interface that displays the patient reports
  • Network connectivity is site specific.
  • Site specificity is desired for sending reports via
  • the instrument can have at least two user modes .
  • One user mode is a locked protocol consistent with
  • a second user mode is modifiable by
  • the apparatus 10 comprises a
  • microscope subsystem 32 housed in a housing 12.
  • housing 12 includes a slide carrier input hopper 16
  • the housing 12 secures the microscope subsystem from
  • the computer subsystem further includes a computer
  • peripherals including storage device 21, pointing
  • external power supply 24 is also shown for powering
  • the apparatus 10 further includes a CCD camera 42 for acquiring images through the microscope
  • control of system processor 23 controls a number of
  • controller 31 provides displacement of the microscope
  • microscope subsystem 32 further includes a motorized
  • objective turret 44 for selection of objectives.
  • the purpose of the apparatus 10 is for the
  • tumor cells e.g., tumor cells.
  • the preferred embodiment may be utilized for rare event detection in which
  • biological specimen may be prepared with a reagent to
  • apparatus of the present invention is used to detect
  • a slide carrier 60 is illustrated in
  • Each slide carrier holds up to 4 slides. Up to 25 slide

Abstract

A method, system (Fig. 2), and apparatus are provided for automated light microscopic (fig. 2, 31) for detection of proteins associated with cell proliferative disorders.

Description

AUTOMATED DETECTION OF OBJECTS IN A BIOLOGICAL SAMPLE
CLAIM OF PRIORITY
This application claims the benefit of priority,
under 35 USC §119 (e) (1), of U.S. provisional
application 60/143,823, filed July 13, 1999, and this
application is a continuation of U.S. Patent
Application Serial No. 08/758,436, filed November 27,
1996, which claims the benefit of U.S. Provisional
Application No. 60/026,805, filed on November 30,
1995.
TECHNICAL FIELD
The invention relates generally to light
microscopy and, more particularly, to automated light
microscopic methods and an apparatus for detection of
proteins associated with cell proliferative
disorders . BACKGROUND OF THE INVENTION
In the field of medical diagnostics including
oncology, the detection, identification, quantitation
and characterization of cells of interest, such as
cancer cells, through testing of biological specimens
is an important aspect of diagnosis. Typically, a
biological specimen such as bone marrow, lymph nodes,
peripheral blood, cerebrospinal fluid, urine,
effusions, fine needle aspirates, peripheral blood
scrapings or other materials are prepared by staining
the specimen to identify cells of interest. One
method of cell specimen preparation is to react a
specimen with a specific probe which can be a
monoclonal antibody, a polyclonal antiserum, or a
nucleic acid which is reactive with a component of
the cells of interest, such as tumor cells. The
reaction may be detected using an enzymatic reaction,
such as alkaline phosphatase or glucose oxidase or
peroxidase to convert a soluble colorless substrate
to a colored insoluble precipitate, or by directly
conjugating a dye to the probe. Examination of biological specimens in the past has been performed
manually by either a lab technician or a pathologist.
In the manual method, a slide prepared with a
biological specimen is viewed at a low magnification
under a microscope to visually locate candidate cells
of interest. Those areas of the slide where cells of
interest are located are then viewed at a higher
magnification to confirm those objects as cells of
interest, such as tumor or cancer cells. The manual
method is time consuming and prone to error including
missing areas of the slide. Automated cell analysis
systems have been developed to improve the speed and
accuracy of the testing process. One known
interactive system includes a single high power
microscope objective for scanning a rack of slides,
portions of which have been previously identified for
assay by an operator. In that system, the operator
first scans each slide at a low magnification similar
to the manual method and notes the points of interest
on the slide for later analysis. The operator then
stores the address of the noted location and the associated function in a data file. Once the points
of interest have been located and stored by the
operator, the slide is then positioned in an
automated analysis apparatus which acquires images of
the slide at the marked points and performs an image
analysis.
A number of cellular proteins are related to
cell proliferation and cell signaling. Many of these
proteins are critical for normal cell growth. For
example, HER2 (neu) is a growth factor receptor and
when found within tumor cells amounts to an
aggressively growing tumor. Studies have determined
that a significantly decreased disease-free survival
and overall survival of a patient with over-
expression of HER2. Before an oncologist prescribes
an anti-HER2/neu therapeutic, an immunohistochemistry
(IHC) assessment for HER2/neu is desirable.
Therapeutic availability increases the need for a
standard methodology for assessing the expression of
HER2/neu. Three general methods are currently available
for the detection of HER2/neu: genetic detection,
protein expression, and protein activity. In situ
hybridization methods are typically used for HER2/neu
genetic detection. Immunohistochemistry methods are
used for the assessment of HER2/neu protein
expression.
SUMMARY OF THE INVENTION
A problem with the foregoing automated system is
the continued need for operator input to initially
locate cell objects for analysis. Such continued
dependence on manual input can lead to errors
including cells of interest being missed. Such
errors can be critical especially in assays for so-
called rare events, e.g., finding one tumor cell in a
cell population of one million normal cells.
Additionally, manual methods can be extremely time
consuming and can require a high decree of training
to identify and/or quantify cells . This is not only
true for tumor cell detection, but also for other applications ranging from neutrophil alkaline
phosphatase assays, reticulocyte counting and
maturation assessment, and others. The associated
manual labor leads to a high cost for these
procedures in addition to the potential errors that
can arise from long, tedious manual examinations. A
need exists, therefore, for an improved automated
cell analysis system which can quickly and accurately
scan large amounts of biological material on a slide.
Accordingly, the present invention provides a method
and apparatus for automated cell analysis which
eliminates the need for operator input to locate cell
objects for analysis.
In accordance with the present invention, a
slide prepared with a biological specimen and reagent
is placed in a slide carrier which preferably holds
four slides. The slide carriers are loaded into an
input hopper of the automated system. The operator
may then enter data identifying the size, shape, and
location of a scan area on each slide, or,
preferably, the system automatically locates a scan area for each slide during slide processing. The
operator then activates the system for slide
processing. At system activation, a slide carrier is
positioned on an X-Y stage of an optical system. Any
bar codes used to identify slides are then read and
stored for each slide in a carrier. The entire slide
is rapidly scanned at a low magnification, typically
lOx. At each location of the scan, a low
magnification image is acquired and processed to
detect candidate objects of interest. Preferably,
color, size, and shape are used to identify objects
of interest. The location of each candidate object
of interest is stored.
At the completion of the low level scan for each
slide in the carrier on the stage, the optical system
is adjusted to a high magnification such as 40x or
60x, and the X-Y stage is positioned to the stored
locations for the candidate objects of interest on
each slide in the carrier. A high magnification
image is acquired for each candidate object of
interest and a series of image processing steps are performed to confirm the analysis which was performed
at low magnification. A high magnification image is
stored for each confirmed object of interest.
Additionally, control slides including positive
and negative controls, may be used to determine
background staining. For example, positive control
slides for a particular staining technique can be run
followed by a negative control in order to determine
a delta for the controls . Subsequent scanning for
objects of interest may then differentiate such
objects based upon their color or intensity above the
delta.
These images are then available for retrieval by
a pathologist or cytotechnologist to review for final
diagnostic evaluation. Having stored the location of
each object of interest, a mosaic comprised of the
candidate objects of interest for a slide may be
generated and stored. The pathologist or
cytotechnologist may view the mosaic or may also
directly view the slide at the location of an object
of interest in the mosaic for further evaluation. The mosaic may be stored on magnetic media for future
reference or may be transmitted to a remote site for
review and/or storage. The entire process involved
in examining a single slide takes on the order of 2-
15 minutes depending on scan area size and the number
of detected candidate objects of interest.
The present invention has utility in the field
of oncology for the early detection of minimal
residual disease ( λ 'micrometastases" ) . Other useful
applications include prenatal diagnosis of fetal
cells in maternal blood and in the field of
infectious diseases to identify pathogens and viral
loads, alkaline phosphatase assessments, reticulocyte
counting, and others. The processing of images
acquired in the automated scanning of the present
invention preferably includes the steps of
transforming the image to a different color space;
filtering the transformed image with a low pass
filter; dynamically thresholding the pixels of the
filtered image to suppress background material;
performing a morphological function to remove artifacts from the thresholded image; analyzing the
thresholded image to determine the presence of one or
more regions of connected pixels having the same
color; and categorizing every region having a size
greater than a minimum size as a candidate object of
interest .
According to another aspect of the invention,
the scan area is automatically determined by scanning
the slide; acquiring an image at each slide position;
analyzing texture information of each image to detect
the edges of the specimen; and storing the locations
corresponding to the detected edges to define the
scan area . According to yet another aspect of the
invention, automated focusing of the optical system
is achieved by initially determining a focal plane
from an array of points or locations in the scan
area . The derived focal plane enables subsequent
rapid automatic focusing in the low power scanning
operation. The focal plane is determined by
determining proper focal positions across an array of
locations and performing an analysis such as a least squares fit of the array of focal positions to yield
a focal plane across the array. Preferably, a focal
position at each location is determined by
incrementing the position of a Z stage for a fixed
number of coarse and fine iterations . At each
iteration, an image is acquired and a pixel variance
or other optical parameter about a pixel mean for the
acquired image is calculated to form a set of
variance data. A least squares fit is performed on
the variance data according to a known function. The
peak value of the least squares fit curve is selected
as an estimate of the best focal position.
In another aspect of the present invention,
another focal position method for high magnification
locates a region of interest centered about a
candidate object of interest within a slide which
were located during an analysis of the low
magnification images. The region of interest is
preferably n columns wide, where n is a power of 2.
The pixels of this region are then processed using a
Fast Fourier Transform to generate a spectra of component frequencies and corresponding complex
magnitude for each frequency component . Magnitudes
of the frequency components which range from 25% to
75% of the maximum frequency component are squared
and summed to obtain the total power for the region
of interest. This process is repeated for other Z
positions and the Z position corresponding to the
maximum total power for the region of interest is
selected as the best focal position.
According to still another aspect of the
invention, a method and apparatus for automated slide
handling is provided. A slide is mounted onto a
slide carrier with a number of other slides side-by-
side. The slide carrier is positioned in an input
feeder with other slide carriers to facilitate
automatic analysis of a batch of slides. The slide
carrier is loaded onto the X-Y stage of the optical
system for the analysis of the slides thereon.
Subsequently, the first slide carrier is unloaded
into an output feeder after automatic image analysis
and the next carrier is automatically loaded.
In a specific embodiment the invention provides
an automated system for the quantitation of proteins
associated with cell proliferative disorders, such as
HER2/neu expression in tissue. The invention is
useful to determine the over-expression of HER2 in
tissue, especially breast tissue.
DESCRIPTION OF THE DRAWINGS
The above and other features of the invention
including various novel details of construction and
combinations of parts will now be more particularly
described with reference to the accompanying drawings
and pointed out in the claims . It will be understood
that the particular apparatus embodying the invention
is shown by way of illustration only and not as a
limitation of the invention. The principles and
features of this invention may be employed in varied
and numerous embodiments without departing from the
scope of the invention. Fig. 1 is a perspective view of an apparatus for
automated cell analysis embodying the present
invention.
Fig. 2 is a block diagram of the apparatus shown
in Fig. 1.
Fig. 3 is a block diagram of the microscope
controller of Fig. 2.
Fig. 4 is a plan view of the apparatus of Fig. 1
having the housing removed.
Fig. 5 is a side view of a microscope subsystem
of the apparatus of Fig . 1.
Fig. 6a is a top view of a slide carrier for use
with the apparatus of Fig. 1.
Fig. 6b is a bottom view of the slide carrier of
Fig. 6a.
Fig. 7a is a top view of an automated slide
handling subsystem of the apparatus of Fig. 1.
Fig. 7b is a partial cross-sectional view of the
automated slide handling subsystem of Fig. 7a taken
on line A-A. Fig. 8 is an end view of the input module of the
automated slide handling subsystem.8a-8d illustrate
the input operation of the automatic slide handling
subsystem.
Fig. 9a-9d illustrate the output operation of
the automated slide handling subsystem.
Fig. 10 is a flow diagram of the procedure for
automatically determining a scan area.
Fig. 11 shows the scan path on a prepared slide
in the procedure of Fig. 10.
Fig. 12 illustrates an image of a field acquired
in the procedure of Fig. 10.
Fig. 13A is a flow diagram of a preferred
procedure of redetermining focal position.
Fig. 13B is a flow diagram of a preferred
procedure for determining a focal position for
neutrophils stained with Fast Red and counterstained
with hemotoxylin.
Fig. 14 is a flow diagram of a procedure for
automatically determining initial focus. Fig. 15 shows an array of slide positions for
use in the procedure of Fig. 14.
Fig. 16 is a flow diagram of a procedure for
automatic focusing at a high magnification.
Fig. 17A is a flow diagram of an overview of the
preferred process to locate and identify objects of
interest in a stained biological specimen on a slide.
Fig. 17B is a flow diagram of a procedure for
color space conversion.
Fig. 18 is a flow diagram of a procedure for
background suppression via dynamic thresholding.
Fig. 19 is a flow diagram of a procedure for
morphological processing.
Fig. 20 is a flow diagram of a procedure for
blob analysis.
Fig. 21 is a flow diagram of a procedure for
image processing at a high magnification.
Fig. 22 illustrates a mosaic of cell images
produced by the apparatus . Fig. 23 is a flow diagram of a procedure for
estimating the number of nucleated cells in an
objective field.
Fig. 24 illustrates the apparatus functions
available in a user interface of the apparatus .
DETAILED DESCRIPTION
Overview
The invention provides an automated tissue image
analysis method for the detection of a cell
proliferative disorder, for example those associated
with HER2/neu. Other automated tissue image analysis
methods, such as immunohistochemical analyses of
estrogen receptor and progesterone receptor are also
provided. Other methods are within the scope of the
invention, such as analyses for MM/MRD for tissue,
MIB-I, Microvessel density analysis, the oncogene
p53, immunophenotyping, hematoxylin/eosin (H/E)
morphological analysis, undifferentiated tumor
classification, antibody titering, prominence of
nucleoli, HIV p24, human papiloma virus (HPV; for cervical biopsy) , and mitotic index; generally any
diagnostic method utilizing staining techniques is
within the scope of the present invention. The
invention has utility in the field of oncology for
the early detection of minimal residual disease
( λ,micrometastases" ) .
For the HER2/neu application, the Automated
Cellular Imaging System (ACIS™) analyzes at least two
subsamples : the H/E prepared slide and the
immunohistochemistry prepared slide. A histological
reconstruction of subsamples is displayed and used
for the appropriate reviewing. An instrument
automatically selects an area of interest, using
color space transformations (and morphological
filtering, if necessary) to identify and quantify the
expression of the HER2/neu in the area of tissue
selected from the immunohistochemistry prepared
slide. The quantitative result, as well as any
selected areas of interest from either slide, is
incorporated into the patient report . The following acronyms and terminology are used
throughout this document: A worklist is a group of
specimens that is prepared with the same staining
techniques and typically analyzed with the same
automated application. A specimen group consists of
one or more patient cases . A histological
reconstruction is an image of the whole specimen that
has been mounted on a slide. This image is created
by piecing together more than one fields of view at
any objective. Unless otherwise defined, all
technical and scientific terms used herein have the
same meaning as commonly understood by one of
ordinary skill in the art to which this invention
belongs .
Although methods and materials similar or
equivalent to those described herein can be used in
the practice or testing of the present invention,
suitable methods and materials are described below.
All publications, patent applications, patents,
and other references mentioned herein are
incorporated by reference in their entirety. In case of conflict, the present application, including
definitions, will control. In addition, the
materials and methods described herein are
illustrative only and not intended to be limiting.
Other features and advantages of the invention will
be apparent from the following detailed description,
the drawings, and from the claims.
Preferred Workflow
A preferred workflow is provided in the
following outline:
1. Specimens arrive in the laboratory. The
specimens are labeled in accordance with
standard hospital procedures to insure the
chain of custody.
2. Sample Preparation.
2.1. Two or three tissue sections per
patient case are preferable for this
test .
2.2. Mount one piece of the tissue to a
slide, to be stained with H/E. The H/E staining protocol is compatible with
HER2/neu immunohistochemistry staining.
2.3. Mount the second piece of the tissue to
a slide, to be stained with the anti-
HER2 immunohistochemistry staining
system.
2.4. If deemed necessary, mount a third
piece of the tissue to a third slide,
to be stained with a third antibody
(i.e., patient negative control) and
the anti-HER2 staining system.
2.5. To avoid ACIS™ mechanical limitations,
the specimen placement specifications
is followed. Considering the frosted
end of the slide as the top, specimen
must be below 30 mm and above 65 mm
from the top edge . The specimen may
reach to the far side edges of the
slide.
2.6. Other patient samples? If yes, repeat
section 2. 2.7. Include two stain control slides.
3. Sample Staining.
3.1. Stain all H/E prepared slides with the
H/E staining protocol .
3.2. Stain all immunohistochemistry slides
with anti-HER2 staining system.
3.3. If deemed necessary, stain all negative
immunohistochemistry slides with a
third antibody (i.e., patient negative
control) and the anti-HER2 staining
system.
3.4. Stain the positive and negative control
slides with the anti-HER2 staining
system and the appropriate antibodies .
4. Barcode all prepared slides and place
within open slots in the slide carriers.
5. Manually study the positive and negative
control slides and verify the staining is
in control. Manually study the patient's
positive and negative immunohistochemistry slides and verify patient doesn't have an
abnormality in background staining.
6. Work-list entry
6.1. Enter the information that is
consistent per work-list.
6.1.1. Specify the work-list
identification.
6.1.2. Specify the
protocol/application (HER2/neu)
6.1.3. Specify the barcodes of the
positive and negative control
slides
6.2. Enter the information that is related
to each patient case.
6.2.1. Accession number and Patient
Identification (ID)
6.2.2. Specify the two/three barcodes
of the samples for this case (H/E,
immunohistochemistry, patient
negative control) 6.2.3. Patient specific information:
Age (integer), Sex (M/F),Race
(character - predefined
selections) , Diagnosis (character
- 2S6) , Date of diagnosis (Date
Format) , Date of last treatment
(Date Format) , and Description of
last treatment (character - 512) .
7. If daily calibration of the instrument
hasn't been completed, please do so before
starting the work-list (5 min per
calibration, necessary once per a day) .
This calibration consists of radiometric,
geometric, and color standard calibration.
8. Load the carriers containing the control
slides and patient cases into the input
hopper .
9. Start the automated analysis of the work-
list.
9.1. Should the stain controls not have a
large enough delta Δ, the instrument skips the rest of the slides within the
worklist .
10. After completion of the analysis, review
the results of the work-list and construct
the appropriate reports online. For each
case, the low power H/E image appears
automatically with the selection of the
patient ID, accession number, or sample
identification .
10.1. While viewing the low power H/E
Histological Reconstruction (HR) the
user has the following choices:
10.1.1. Select an area of the H/E HR
to zoom on .
10.1.2. Choose to go to the low power
immunohistochemistry HR.
10.1.3. Select an area to save within
the pre-constructed report format.
10.2. While viewing the high power H/E
HR the user has the following choices : 10.2.1. Choose to go back to the low
power H/E HR.
10.3. While viewing the low power
immunohistochemistry HR the user has
the following choices :
10.3.1. Select an area of the
immunohistochemistry HR to zoom
on.
10.3.2. Select an area of the low
power immunohistochemistry HR and
query for the following
quantitative results . The
quantitative results are relative
to the scoring of the positive
stain control and the patient's
negative immunohistochemistry
slide.
10.3.2.1. Over-expression within
selected area reported as
0, 1+, 2+, or 3+. 10.3.2.2. Percentage of positive
cells within the selected
area.
10.4. While viewing the high power
immunohistochemistry HR the user has
the following choices :
10.4.1. Select an area of the high
power immunohistochemistry HR and
query for the following
quantitative results . The
quantitative results are relative
to the scoring of the positive
stain control and the patient's
negative immunohistochemistry
slide.
10.4.1.1. Over-expression within
selected area reported as
0, 1+, 2+, or 3+.
10.4.1.2. Percentage of positive
cells within the selected
area. 10.4.2. Choose to go back to the low
power immunohistochemistry HR.
10.5. Other samples to review?
10.5.1. If yes, repeat section 8.
10.6. Should the analysis result in the
tissue not being scanned, reload slide,
open application, and adjust scan area
to location of the tissue, if
necessary. Should the scan area be
adequate, but the focus points not fall
on an area of tissue, resulting in
failed focus, in the application change
the focus inset to a number closer to
one and confirm that the scan area is
centered on an area of tissue. Execute
the application. Repeat steps 10.1-
10.5.
10.7. Manually record results of
analysis .
11. Quality Assurance checks within the
instrument . 11.1. At the completion of analyzing
both the positive and negative control
slides, the Δ between the scores are
calculated. If this Δ isn't large
enough, the analysis of all slides in
this work-list is flagged accordingly
and the instrument warns the user with
an audible alarm.
11.2. If the Δ between the patient's
positive and negative
immunohistochemistry slide isn't large
enough, the patient is flagged
accordingly.
11.3. A montage of tissue sections from
both the positive and negative
controls. Five tissue section images
from each are adequate .
12. Preview the reports online and modify as
necessary.
13. Send the reports to the printer or by the
Internet to the requesting clinicians. The output of the worklist is a stained
positive control slide, a stained negative
control slide, two stained slides per
patient case (one H/E and another IHC) , and
a comprehensive electronic report. The
report includes the selected image
sections, quantitative results, reference
material, and other pertinent data.
Sample Staining
A cellular specimen (a "sample") is split to
provide two or more subsamples. The term "sample"
includes cellular material derived from a subject.
Such samples include but are not limited to hair,
skin samples, tissue sample, cultured cells, cultured
cell media, and biological fluids. The term
"tissue" refers to a mass of connected cells (e.g.,
CNS tissue, neural tissue, or eye tissue) derived
from a human or other animal and includes the
connecting material and the liquid material in
association with the cells. The term "biological fluid" refers to liquid material derived from a
human or other animal . Such biological fluids
include, but are not limited to, blood, plasma,
serum, serum derivatives, bile, phlegm, saliva,
sweat, amniotic fluid, and cerebrospinal fluid (CSF) ,
such as lumbar or ventricular CSF. The term
"sample" also includes media containing isolated
cells . The quantity of sample required to obtain a
reaction may be determined by one skilled in the art
by standard laboratory techniques. The optimal
quantity of sample may be determined by serial
dilution.
The HER2/neu method uses an anti-HER2/neu
staining system, such as a commercially available
kit, like that provided by DAKO (Carpinteria, CA) .
The steps for the immunohistochemistry protocol are
as follows: (1) Prepare wash buffer solution.
(2) Deparaffinize and rehydrate specimens. (3)
Perform epitope retrieval. Incubate 40 min in a 95°C
water bath. Cool slides for 20 min at room
temperature. (4) Apply peroxidase blocking reagent. Incubate 5 min. (5) Apply primary antibody or
negative control reagent. Incubate 30 min +/- 1 min
at room temperature. Rinse in wash solution. Place
in wash solution bath. (6) Apply peroxidase labeled
polymer. Incubate 30 min +/- 1 min at room
temperature. Rinse in wash solution. Place in wash
solution bath. (7) Prepare DAB substrate chromagen
solution. (8) Apply substrate chromogen solution
(DAB) . Incubate 5-10 min. Rinse with distilled
water; (9) Counterstain. (10) Mount coverslips. The
slide includes a cover-slip medium to protect the
sample and to introduce optical correction consistent
with microscope objective requirements. The
coverslip covers the entire prepared specimen.
Mounting the coverslip does not introduce air bubbles
obscuring the stained specimen. This coverslip could
potentially be a mounted 1-1/2 thickness coverslip
with DAKO Ultramount medium. (11) A set of staining
control slides are run with every worklist. The set
includes a positive and negative control. The
positive control is stained with the anti-HER2 antibody and the negative is stained with another
antibody. Both slides are identified with a unique
barcode. Upon reading the barcode, the instrument
recognizes the slide as part of a control set, and
runs the appropriate application. There may be one
or two applications for the stain controls. (12) A
set of instrument calibration slides includes the
slides used for focus and color balance calibration.
(13) A dedicated carrier is used for one-touch
calibration. Upon successful completion of this
calibration procedure, the instrument reports itself
to be calibrated. Upon successful completion of
running the standard slides, the user is able to
determine whether the instrument is within standards
and whether the inter-instrument and intra-instrument
repeatability of test results.
The hematoxylin/eosin (H/E) slides are prepared
with a standard H/E protocol . Standard solutions
include the following: (1) Gills hematoxylin
(hematoxylin 6.0 g; aluminium sulphate 4.2 g; citric
acid 1.4 g; sodium iodate 0.6 g; ethylene glycol 269 ml; distilled water 680 ml) ; (2) eosin (eosin
yellowish 1.0 g; distilled water 100 ml); (3) lithium
carbonate 1% (lithium carbonate 1 g; distilled water
100 g) ; (4) acid alcohol 1% 70% (alcohol 99 ml cone;
hydrochloric acid 1 ml); and (5) Scott's tap water.
In a beaker containing 1 L distilled water, add 20 g
sodium bicarbonate and 3.5 g magnesium sulphate. Add
a magnetic stirrer and mix thoroughly to dissolve the
salts. Using a filter funnel, pour the solution into
a labeled bottle.
The staining procedure is as follows: (1) Bring
the sections to water; (2) place sections in
hematoxylin for 5 min; (3) wash in tap water; (4)
'blue' the sections in lithium carbonate or Scott's
tap water; (5) wash in tap water; (6) place sections
in 1% acid alcohol for a few seconds; (7) wash in tap
water; (8) place sections in eosin for 5 min; (9) wash
in tap water; and (10) dehydrate, clear. Mount
sections .
The results of the H/E staining provide cells
with nuclei stained blue-black, cytoplasm stained varying shades of pink; muscle fibers stained deep
pinky red; fibrin stained deep pink; and red blood
cells stained orange-red.
The invention also provides automated methods
for analysis of estrogen receptor and progesterone
receptor. The estrogen and progesterone receptors,
like other steroid hormone receptors, plays a role in
developmental processes and maintenance of hormone
responsiveness in target cells. From the molecular
viewpoint, estrogen and progesterone receptor
interaction with target genes is of paramount
importance in maintenance of normal cell function and
is also involved in regulation of mammary tumor cell
function. The expression of progesterone receptor
and estrogen receptor in breast tumors is a useful
indicator for subsequent hormone therapy. An anti-
estrogen receptor antibody labels epithelial cells of
breast carcinomas which express estrogen receptor.
An immunohistochemical assay of the estrogen receptor
is performed using an anti-estrogen receptor
antibody, for example the well-characterized 1D5 clone, and the methods of Pertchuk, et al . (Cancer
77: 2514-2519, 1996) or a commercially available
immunohistochemistry system such as that provided by
DAKO (Carpenteria CA; DAKO LSAB2 Immunostaining
System) .
In breast carcinoma cells, immunohistochemistry
immunostaining of progesterone receptor has been
demonstrated in the nuclei of cells from various
histologic subtypes. An anti-progesterone receptor
antibody labels epithelial cells of breast carcinomas
which express progesterone receptor. An
immunohistochemical assay of the progesterone
receptor is performed using an anti-estrogen receptor
antibody, for example the well-characterized 1A6
clone, and the methods of Pertchuk, et al . (Cancer
77: 2514-2519, 1996) .
Still other automated analyses are within the
scope of the invention, including the following:
Micrometastases/Metastatic Recurring Disease
(MM/MRD) . Metastasis is the biological process
whereby a cancer spreads to the distant part of the body from its original site. A micrometastases is
the presence of a small number of tumor cells,
particularly in the lymph nodes and bone marrow. A
metastatic recurring disease is similar to
micrometastasis, but is detected after cancer therapy
rather than before therapy. An immunohistochemical
assay for MM/MRD is performed using a monoclonal
antibody which reacts with an antigen (a metastatic-
specific mucin) found in bladder, prostate and breast
cancers .
MIB-1. MIB-1 is an antibody that can be used in
immunohistochemical assays for the antigen Ki-67.
The clinical stage at first presentation is related
to the proliferative index measured with Ki-67. High
index values of Ki-67 are positively correlated with
metastasis, death from neoplasia, low disease-free
survival rates, and low overall survival rates.
Microvessel Density Analysis. Microvessel
density analysis is a measure of new blood vessel
formation (angiogenesis) . Angiogenesis is
characteristic of growing tumors . Intratumor microvessel density can be assessed by anti-CD34
immunostaining .
The Oncogene p53. Overexpression of the p53
oncogene has been implicated as the most common
genetic alteration in the development of human
malignancies . Investigations of a variety of
malignancies, including neoplasms of breast, colon,
ovary, lung, liver, mesenchyme, bladder and myeloid,
have suggested a contributing role of p53 mutation in
the development of malignancy. The highest frequency
of expression has been demonstrated in tumors of the
breast, colon and ovary. A wide variety of normal
cells do express a wildtype form of p53 but generally
in restricted amounts . Overexpression and mutation
of p53 have not been recognized in benign tumors or
in normal tissue. An immunohistochemical assay of
p53 is performed using an anti-p53 antibody, for
example the well-characterized DO-7 clone.
Immunophenotyping .
Undifferentiated tumor classification.
Antibody titering.
Prominence of Nucleoli. A nucleoli is an
organelle in a cell nucleus. Cervical dysplasia
refers to the replacement of the normal or
metaplastic epithelium with atypical epithelial cells
that have cytologic features that are pre-malignant
(nuclear hyperchromatism, nuclear enlargement and
irregular outlines, increased nuclear-to-cytoplasmic
ratio, increased prominence of nucleoli) and
chromosomal abnormalities . The changes seen in
dysplastic cells are of the same kind but of a lesser
degree than those of frankly malignant cells. In
addition, there are degrees of dysplasia (mild,
moderate, severe) .
HIV p24 Protein. Human immunodeficiency virus
(HIV) p24 antigen levels are measured in an
immunohistochemistry assay using anti-HIV p24
antigen. The HIV p24 protein test is used for the
detection of HIV virus. The test can be used to detect the virus, measure the amount of virus, and
examine the virus' genetic composition.
Human Papiloma Virus (HPV) . HPV is a sexually
transmitted virus which causes warts . HPV is an
important health concern because it may cause cancer
of the cervix. An immunohistochemical assay of HPV
is performed using an antibody associated with
cervical cancer. Several serological responses are
strongly associated with cervical cancer, notably the
IgG response against the HPV 16 epitopes LI: 13,
E2:9, and E7 : 5 , and the IgA response against an HPV
18 E2-derived antigen.
Mitotic Index. When a viral genome is
incorporated into the nuclear genetic material,
uncontrolled malignant growth of the host cell may be
promoted.
Operating the Instrument
While operating the instrument, the user
interacts with a few screens including, but not
limited to, Patient Entry, Review Data, Construct Report, Manual Control, and User Preferences. Rather
than the information entry revolving around the
patient cases, the patient data entry interface is
organized as worklist information composed of
multiple patient cases. For example, a worklist for
HER2/neu is composed of tissue samples that are
prepared in one of two methods, the HER2/neu
immunohistochemistry staining technique or the H/E
technique. The slides within a HER2/neu worklist
consists of a positive control slide (prepared with
the immunohistochemistry technique and the HER2
antibody) , a negative control slide (prepared with
the immunohistochemistry technique and another
antibody) , and two slides for the one or more patient
cases. Per patient case there are two slides
prepared. One slide is prepared with the HER2/neu
immunohistochemistry technique and a second is
prepared with the H/E technique for general tissue
analysis .
The data that is relevant for a HER2/neu
worklist is the: (1) worklist name or identifier of protocol (e.g., HER2/neu) ; barcode of the positive
control slide; and barcode of the negative control
slide. The data that is relevant on a per Patient
Case is the: (4) patient ID; (5) accession number;
(6) social security number; (7) referring hospital,
(8) referring physician; (9) barcode of the
immunohistochemistry prepared slide; and (10) barcode
of the H/E prepared slide.
The user starts analysis with the batch button
and the slide carriers loaded into the input hopper.
The input hopper is composed of all the
immunohistochemistry slides from the patient cases,
all the H/E slides from the patient cases, the
positive control slide, and the negative control
slide. During analysis the instrument reads the
barcode on the slide and determines one of three
situations and starts one of the three appropriate
computer applications: (1) Slide is the positive
control slide and run the HER2-positive application.
(2) Slide is the negative control slide and run the
HER2-negative application. (3) Slide is either the immunohistochemistry or H/E patient slide and run the
HER2 application.
The system reads industry standard barcodes .
The barcode relates the type of protocol the
instrument uses to analyze the slide. The type of
data stored for each slide is determined by the stain
preparation and the protocol used to analyze it .
During the transport of the slide through the system,
therefore, the chain of custody is preserved. When a
barcode is unreadable or isn't found in one of the
defined worklists, the action taken by the instrument
is to ignore the slide altogether and send it to the
output hopper .
The HER2-positive application (1) finds the
specimen, (2) goes to five locations within the
specimen, (3) stores the image from each location,
(4) completes color space transformations to obtain
the intensity of stain at all five locations, and (5)
stores the average value of intensity in the database
(DB) . This value is used as the normalized
expression value for the HER2 quantitative analysis. This value is also used to determine the Δ between
the positive and negative control slides and verify
that the Δ is large enough.
The HER2-negative application (1) finds the
specimen, (2) goes to five locations within the
specimen, (3) stores the image from each location,
(4) completes color space transformations to obtain
the intensity of stain at all five locations, and (5)
stores the average value of intensity in the
database. This value is used to determine the Δ
between the positive and negative control slides and
verify that the Δ is large enough.
The HER2 application (1) finds the specimen, (2)
scans the slide at lOx and constructs a histological
reconstruction object, and (3) stores the object in
the database. Before a slide is analyzed, the
instrument checks to see if the hard drive space has
met a certain capacity for archiving data. If so,
the user is prompted to archive before continuing
instrument analysis. Compression may need to take place prior to the object being stored within the
database.
After completion of running all the slides
within the worklist, the histological reconstruction
objects are analyzed by the pathologist or physician.
The Slide Display Interface displays the list of
slides that were analyzed, or the list of patient
cases. Within tissue analysis, there are always at
least two slides per patient case that are analyzed
by the instrument. The set of patient slides is a
logical unit. Therefore, a pathologist or physician
may choose the patient case by the patient ID or the
accession number.
Within the User Interface that displays the list
of patient cases to be reviewed/analyzed, once the
pathologist chooses the patient case, H/E image is
always displayed first. Using the H/E image, the
pathologist determines whether or not the cancer is
invasive and whether or not analysis of the IHC slide
is necessary. Upon patient case selection, the low power H/E
image is displayed. This image can be reduced to
enable the whole tissue cross-section to be displayed
within the system's monitor (resolution at least 1024
x 760) .
The pathologist can have the features available
to choose more than one patient case to review and
then traverse from one case to the next with the
following features. For each patient case, the
reduced low power H/E image always appears first .
While viewing the reduced low power H/E image
the pathologist has the following capabilities: (1)
to zoom into an area of the low power H/E image for
higher magnification; (2) to choose to go to the
reduced low power immunohistochemistry image for the
current patient case; (3) to select an area to
incorporate within the patient report (Marking an
image section) ; (4) to choose to go to the next
patient case (if selected in previous user
interface) ; and (5) to choose to go to the previous
patient case. While viewing the zoomed H/E image the
pathologist has the following capabilities: (1) to
go back to the reduced low power H/E image; (2) to
choose to go to the reduced low power
immunohistochemistry image for the current patient
case; (3) to select an area to incorporate within the
patient report (Marking an image section) ; (4) to
choose to go to the next patient case (if selected in
previous user interface) ; and (5) to choose to go to
the previous patient case.
While viewing the reduced low power
immunohistochemistry image the pathologist has the
following capabilities: (1) to zoom into an area of
the low power immunohistochemistry image for higher
magnification; (2) to choose to go back to the
reduced H/E image for the current patient case; (3)
to select an area of the reduced immunohistochemistry
image and query for quantitative results; (4) to
select an area to incorporate within the patient
report (Marking an image section) ; (5) to select a
quantitative result to incorporate within the patient report (Save the quantitative result) ; (6) to choose
to go to the next patient case (if selected in
previous user interface) ; and (7) to choose to go to
the previous patient case (if applicable) .
While viewing the zoomed immunohistochemistry
image the pathologist has the following capabilities :
(1) to go back to the reduced low power IHC image;
(2) to choose to go back to the reduced H/E image for
the current patient case; (3) to select an area of
the zoomed immunohistochemistry image and query for
quantitative results; (4) to select an area to
incorporate within the patient report (Marking an
image section) ; (5) to select a quantitative result
to incorporate within the patient report (Save the
quantitative result) ; (6) to choose to go to the next
patient case (if selected in previous user
interface) ; and (7) to choose to go to the previous
patient case (if applicable) .
After completion of the pathologist analysis and
review of all the patient cases, the reports can be
generated. A User Interface lists all the patient cases that have already been reviewed and analyzed by
the pathologist. The pathologist can choose one or
more of the listed patient cases to view the reports
electronically. The HER2/neu report for a patient
case can contain the following information: (1)
Patient Information and Demographics as entered
within the Worklist Information Entry; (2) Referring
Physician and Hospital as entered within the Worklist
Information Entry; (3) Pathologist and Laboratory
Information; (4) Sections of the H/E image marked
during the review/analysis for report inclusion; (5)
sections of the immunohistochemistry image marked
during the review/analysis for report inclusion;
(5) quantitative analysis saved during the
review/analysis for report inclusion; and (6)
HER2/neu application and scoring analysis reference
materials .
The interface that displays the patient reports
online contains the following functionality: (1)
Print; (2) Send To, allows for (linked to e-mail,
when available, and prompting the user for the address of where to send the report) ; (3) Next
Patient Case Report (if more than one case was
selected from the previous interface) ; (4) Previous
Patient Case Report (if applicable) ; and
(5) Save/Apply (if the additional comments area is
modified). Network connectivity is site specific.
Site specificity is desired for sending reports via
the Internet without having to transport the report
over to another host to send it. Direct sending
capabilities are preferred.
The instrument can have at least two user modes .
One user mode is a locked protocol consistent with
FDA regulations . A second user mode is modifiable by
the user. For the second user mode, access to the
following parameters of the application include: (1)
definition of the scan area, center, width, and
height; (2) toggle switch for whether or not the Find
phase is used; (3) Focus Type; (4) Focus Threshold;
and (5) Focus Inset. Automated System
Referring now to the figures, an apparatus for
automated cell analysis of biological specimens is
generally indicated by reference numeral 10 as shown
in perspective view in Fig. 1 and in block diagram
form in Fig. 2. The apparatus 10 comprises a
microscope subsystem 32 housed in a housing 12. The
housing 12 includes a slide carrier input hopper 16
and a slide carrier output hopper 18. A door 14 in
the housing 12 secures the microscope subsystem from
the external environment . A computer subsystem
comprises a computer 22 having a system processor 23,
an image processor 25 and a communications modem 29.
The computer subsystem further includes a computer
monitor 26 and an image monitor 27 and other external
peripherals including storage device 21, pointing
device 30, keyboard 28 and color printer 35. An
external power supply 24 is also shown for powering
the system. Viewing oculars 20 of the microscope
subsystem project from the housing 12 for operator
viewing. The apparatus 10 further includes a CCD camera 42 for acquiring images through the microscope
subsystem 32. A microscope controller 31 under the
control of system processor 23 controls a number of
microscope-subsystem functions described further in
detail . An automatic slide feed mechanism in
conjunction with X-Y stage 38 provide automatic slide
handling in the apparatus 10. An illumination light
source 48 projects light onto the X-Y stage 38 which
is subsequently imaged through the microscope
subsystem 32 and acquired through CCD camera 42 for
processing in the image processor 25. A Z stage or
focus stage 46 under control of the microscope
controller 31 provides displacement of the microscope
subsystem in the Z plane for focusing. The
microscope subsystem 32 further includes a motorized
objective turret 44 for selection of objectives.
The purpose of the apparatus 10 is for the
unattended automatic scanning of prepared microscope
slides for the detection and counting of candidate
objects of interest such as normal and abnormal
cells, e.g., tumor cells. The preferred embodiment may be utilized for rare event detection in which
there may be only one candidate object of interest
per several hundred thousand normal cells, e.g., one
to five candidate objects of interest per 2 square
centimeter area of the slide. The apparatus 10
automatically locates and counts candidate objects of
interest and estimates normal cells present in a
biological specimen on the basis of color, size and
shape characteristics . A number of stains are used
to preferentially stain candidate objects of interest
and normal cells different colors so that such cells
can be distinguished from each other.
As noted in the background of the invention, a
biological specimen may be prepared with a reagent to
obtain a colored insoluble precipitate. The
apparatus of the present invention is used to detect
this precipitate as a candidate object of interest.
During operation of the apparatus 10, a pathologist
or laboratory technician mounts prepared slides onto
slide carriers. A slide carrier 60 is illustrated in
Fig. 8 and will be described further below. Each slide carrier holds up to 4 slides. Up to 25 slide
carriers are then loaded into input hopper 16. The
operator can specify the size, shape and location of
the area to be scanned or alternatively, the system
can automatically locate this area. The operator
then commands the system to begin automated scanning
of the slides through a graphical user 25 interf ce.
Unattended scanning begins with the automatic loading
of the first carrier and slide onto the precision
motorized X-Y stage 38. A bar code label affixed to
the slide is read by a bar code reader 33 during this
loading operation. Each slide is then scanned at a
user selected low microscope magnification, for
example, lOx, to identify candidate objects of
interest based on their color, size, and shape
characteristics. The X-Y locations of candidate
cells are stored until scanning is completed.
After the low magnification scanning is
completed, the apparatus automatically returns to
each candidate object of interest, reimages and
refocuses at a higher magnification such as 40x and performs further analysis to confirm the object
candidate . The apparatus stores an image of the
object for later review by a pathologist. All
results and images can be stored to a storage device
21 such as a removable hard drive or DAT tape or
transmitted to a remote site for review or storage.
The stored images for each slide can be viewed in a
mosaic of images for further review. In addition,
the pathologist or operator can also directly view a
detected object through the microscope using the
included oculars 20 or on image monitor 27.
Having described the overall operation of the
apparatus 10 from a high level, the further details
of the apparatus will now be described. Referring to
Fig. 3, the microscope controller 31 is shown in more
detail. The microscope controller 31 includes a
number of subsystems connected through a system bus .
A system processor 102 controls these subsystems and
is controlled by the apparatus system processor 23
through an RS 232 controller 110. The system
processor 102 controls a set of motor - control subsystems 114 through 124 which control the input
and output feeder, the motorized turret 44, the X-Y
stage 38, and the Z stage 46 (Fig. 2) . A histogram
processor 108 receives input from CCD camera 42 for
computing variance data during the focusing operation
described further herein. The system processor 102
further controls an illumination controller 106 for
control of substage illumination 48. The light
output from the halogen light bulb which supplies
illumination for the system can vary over time due to
bulb aging, changes in optical alignment, and other
factors. In addition, slides which have been "over
stained" can reduce the camera exposure to an
unacceptable level . In order to compensate for these
effects, the illumination controller 106 is included.
This controller is used in conjunction with light
control software to compensate for the variations in
light level . The light control software samples the
output from the camera at intervals (such as between
loading of slide carriers) , and commands the
controller to adjust the light level to the desired levels . In this way, light control is automatic and
transparent to the user and adds no substantial
additional time to system operation. The system
processor 23 may be any processor capable of
performing the operations of the system, for example
dual parallel Intel Pentium 90 MHZ devices, a T.I.
C80 processor for computation and processors capable
of 8 to 32 bit operation on an NT system. The image
processor 25 is preferably a Matrox Imaging Series
640 model. The microscope controller system
processor 102 is an Advanced Micro Devices AMD29K
device .
Figs . 4 and 5 show further detail of the
apparatus 10 is shown. Fig. 4 shows a plan view of
the apparatus 10 with the housing 12 removed. A
portion of the automatic slide feed mechanism 37 is
shown to the left of the microscope subsystem 32 and
includes slide carrier unloading assembly 34 and
unloading platform 36 which in conjunction with slide
carrier output hopper 18 function to receive slide
carriers which have been analyzed. Vibration isolation mounts 40, shown in further detail in Fig.
5, are provided to isolate the microscope subsystem
32 from mechanical shock and vibration that can occur
in a typical laboratory environment. In addition to
external sources of vibration, the high-speed
operation of the X-Y stage 38 can induce vibration
into the microscope subsystem 32. Such sources of
vibration can be isolated from the electro-optical
subsystems to avoid any undesirable effects on image
quality. The isolation mounts 40 comprise a spring
40a and piston 40b submerged in a high viscosity
silicon gel which is enclosed in an elastomer
membrane bonded to a casing to achieve damping
factors on the order of 17 to 20%.
The automatic slide-handling feature of the
present invention will now be described. The
automated slide handling subsystem operates on a
single slide carrier at a time. A slide carrier 60
is shown in Figs . 6a and 6b which provide a top view
and a bottom view respectively. The slide carrier 60
includes up to four slides 70 mounted with adhesive tape 62. The carrier 60 includes ears 64 for hanging
the carrier in the output hopper 18. An undercut 66
and pitch rack 68 are formed at the top edge of the
slide carrier 60 for mechanical handling of the slide
carrier. A keyway cutout 65 is formed in one side of
the carrier 60 to facilitate carrier alignment. A
prepared slide 72 mounted on the slide carrier 60
includes a sample area 72a and a bar code label area
72b. Fig. 7a provides a top view of the slide
handling subsystem which comprises a slide input
module 15, a slide output module 17 and X-Y stage
drive belt 50. Fig. 7b provides a partial cross-
sectional view taken along line A-A of Fig. 7a. The
slide input module 15 comprises a slide carrier input
hopper 16, loading platform 52 and slide carrier
loading subassembly 54. The input hopper 16 receives
a series of slide carriers 60 (Figs. 6a and 6b) in a
stack on loading platform 52. A guide key 57
protrudes from a side of the input hopper 16 to which
the keyway cutout 65 (Fig. 6a) of the carrier is fit
to achieve proper 15 alignment. The input module 15 further includes a revolving indexing cam 56 and a
switch 90 mounted in the loading platform 52, the
operation of which is described further below. The
carrier loading subassembly 54 comprises an infeed
drive belt 59 driven by a motor 86. The infeed drive
belt 59 includes a pusher tab 58 for pushing the
slide carrier horizontally toward the 20 X-Y stage 38
when the belt is driven. A homing switch 95 senses
the pusher tab 58 during a revolution of the belt 59.
Referring specifically to Fig. 7a, the X-Y stage
38 is shown with x position and y position motors 96
and 97 respectively which are controlled by the
microscope controller 31 (Fig. 3) and are not
considered part of the slide handling subsystem. The
X-Y stage 38 further includes an aperture 55 for
allowing illumination to reach the slide carrier. A
switch 91 is mounted adjacent the aperture 55 for
sensing contact with the carrier and thereupon
activating a motor 87 to drive stage drive belt 50
(Fig. 7b) . The drive belt 50 is a double-sided timing belt having teeth for engaging pitch rack 68
of the carrier 60 (Fig. 6b) .
The slide output module 17 includes slide
carrier output hopper 18, unloading platform 36 and
slide carrier unloading subassembly 34. The
unloading subassembly 34 comprises a motor 89 for
rotating the unloading platform 36 about shaft 98
during an unloading operation described further
below. An outfeed gear 93 driven by motor 88
rotatably engages the pitch rack 68 of the carrier 60
(Fig. 6b) to transport the carrier to a rest position
against switch 92. A springloaded hold-down
mechanism holds the carrier in place on the unloading
platform 36.
The slide handling operation will now be
described. Referring to Fig. 8, a series of slide
carriers 60 are shown stacked in input hopper 16 with
the top edges 60a aligned. As the slide handling
operation begins, the indexing cam 56 driven by motor
85 advances one revolution to allow only one slide carrier to drop to the bottom of the hopper 16 and
onto the loading platform 52.
Figs. 8a-8d show the cam action in more detail.
The cam 56 includes a hub 56a to which are mounted
upper and lower leaves 56b and 56c respectively. The
leaves 56b, 56c are semicircular projections
oppositely positioned and spaced apart vertically.
In a first position shown in Fig. 8a, the upper leaf
56b supports the bottom carrier at the undercut
portion 66. At a 20 position of the cam 56 rotated
180°, shown in Fig. 8b, the upper leaf 56b no longer
supports the carrier and instead the carrier has
dropped slightly and is supported by the lower leaf
56c. Fig. 8c shows the position of the cam 56
rotated 270° wherein the upper leaf 56b has rotated
sufficiently to begin to engage the undercut 66 of
the next slide carrier while the opposite facing
lower leaf 56c still supports the bottom carrier.
After a full rotation of 360° as shown in Fig. 8d,the
lower leaf 56c has rotated opposite the carrier stack
and no longer supports the bottom carrier which now rests on the loading platform 52. At the same
position, the upper leaf 56b supports the next
carrier for repeating the cycle.
Referring again to Figs. 7a and 7b, when the
carrier drops to the loading platform 52, the contact
closes switch 90 which activates motors 86 and 87.
Motor 86 drives the infeed drive belt 59 until the
pusher tab 58 makes contact with the carrier and
pushes the carrier onto the X-Y stage drive belt 50.
The stage drive belt 50 advances the carrier until
contact is made with switch 91, the closing of which
begins the slide scanning process described further
herein. Upon completion of the scanning process, the
X-Y stage 38 moves to an unload position and motors
87 and 88 are activated to transport the carrier to
the unloading platform 36 using stage drive belt 50.
As noted, motor 88 drives outfeed gear 93 to engage
the carrier pitch rack 68 of the carrier 60 (Fig. 6b)
until switch 92 is contacted. Closing switch 92
activates motor 89 to rotate the unloading platform
36. The unloading operation is shown in more detail
in end views of the output module 17 (Figs. 9a-9d) .
In Fig. 9a, the unloading platform 36 is shown in a
horizontal position supporting a slide carrier 60.
The hold-down mechanism 94 secures the carrier 60 at
one end. Fig. 9b shows the output module 17 after
motor 89 has rotated the unloading platform 36 to a
vertical position, at which point the spring loaded
hold-down mechanism 94 releases the slide carrier 60
into the output hopper 18. The carrier 60 is
supported in the output hopper 18 by means of ears 64
(Figs. 6a and 6b) . Fig. 9c shows the unloading
platform 36 being rotated back towards the 20
horizontal position. As the platform 36 rotates
upward, it contacts the deposited carrier 60 and the
upward movement pushes the carrier toward the front
of the output hopper 18. Fig. 9d shows the unloading
platform 36 at its original horizontal position after
having output a series of slide carriers 60 to the
output hopper 18. Having described the overall system and the
automated slide handling feature, the aspects of the
apparatus 10 relating to scanning, focusing and image
processing will now be described in further detail.
In some cases, an operator will know ahead of
time where the scan area of interest is on the slide.
Conventional preparation of slides for examination
provides repeatable and known placement of the sample
on the slide. The operator can therefore instruct
the system to always scan the same area at the same
location of every slide which is prepared in this
fashion. But there are other times in which the area
of interest is not known, for example, where slides
are prepared manually with a known smear technique.
One feature of the invention automatically determines
the scan area using a texture analysis process. Fig.
10 is a flow diagram that describes the processing
associated with the automatic location of a scan
area. As shown in this figure, the basic method is
to pre-scan the entire slide area to determine
texture features that indicate the presence of a smear and to discriminate these areas from dirt and
other artifacts.
At each location of this raster scan, an image
such as in Fig. 12 is acquired and analyzed for
texture information at steps 204 and 206. Since it
is desired to locate the edges of the smear sample
within a given image, texture analyses are conducted
over areas called windows 78, which are smaller than
the entire image as shown in Fig. 12. The process
iterates the scan across the slide at steps
208,210,212 and 214.
In the interest of speed, the texture analysis
process is performed at a lower magnification,
preferably at a 4x objective. One reason to operate
at low magnification is to image the largest slide
area at any one time. Since cells do not yet need to
be resolved at this stage of the overall image
analysis, the 4x magnification is preferred. On a
typical slide, as shown in Fig. 11, a portion 72b of
the end of the slide 72 is reserved for labeling with
identification information. Excepting this label area, the entire slide is scanned in a raster scan
fashion 76 to yield a number of adjacent images.
Texture values for each window include the pixel
variance over a window, the difference between the
largest and smallest pixel value within a window, and
other indicators . The presence of a smear raises the
texture values compared with a blank area.
One problem with a smear from the standpoint of
determining its location is its non-uniform thickness
and texture. For example, the smear is likely to be
relatively thin at the edges and thicker towards the
middle due to the nature of the smearing process . To
accommodate for the non-uniformity, texture analysis
provides a texture value for each analyzed area . The
texture value tends to gradually rise as the scan
proceeds across a smear from a thin area to a thick
area, reaches a peak, and then falls off again to a
lower value as a thin area at the edge is reached.
The problem is then to decide from the series of
texture values the beginning and ending, or the
edges, of the smear. The texture values are fit to a square wave waveform since the texture data does not
have sharp beginnings and endings .
After conducting this scanning and texture
evaluation operation, one must determine which areas
of elevated texture values represent the desired
smear 74, and which represent undesired artifacts.
This is accomplished by fitting a step function, on a
line-by-line basis to the texture values in step 216.
This function, which resembles a single square wave
across the smear with a beginning at one edge, and
end at the other edge, and an amplitude provides the
means for discrimination. The amplitude of the best-
fit step function is utilized to determine whether
smear or dirt is present since relatively high values
indicate smear. If it is decided that smear is
present, the beginning and ending coordinates of this
pattern are noted until all lines have been
processed, and the smear sample area defined at 218.
After an initial focusing operation described
further herein, the scan area of interest is scanned
to acquire images for image analysis . The preferred method of operation is to initially perform a
complete scan of the slide at low magnification to
identify and locate candidate objects of interest,
followed by further image analysis of the candidate
objects of interest at high magnification in order to
confirm the objects as cells. An alternate method of
operation is to perform high magnification image
analysis of each candidate object of interest
immediately after the object has been identified at
low magnification. The low magnification scanning
then resumes, searching for additional candidate
objects of interest. Since it takes on the order of
a few seconds to change objectives, this alternate
method of operation would take longer to complete .
The operator can pre-select a magnification
level to be used for the scanning operation. A low
magnification using a lOx objective is preferred for
the scanning operation since a larger area can be
initially analyzed for each acquired scan image. The
overall detection process for a cell includes a
combination of decisions made at both low (lOx) and high magnification (40x) levels. Decision making at
the lOx magnification level is broader in scope,
i.e., objects that loosely fit the relevant color,
size and shape characteristics are identified at the
lOx level.
Analysis at the 40x magnification level then
proceeds to refine the decision-making and confirm
objects as likely cells or candidate objects of
interest. For example, at the 40x level it is not
uncommon to find that some objects that were
identified at lOx are artifacts which the analysis
process will then reject. In addition, closely
packed objects of interest appearing at lOx are
separated at the 40x level.
In a situation where a cell straddles or
overlaps adjacent image fields, image analysis of the
individual adjacent image fields could result in the
cell being rejected or undetected. To avoid missing
such cells, the scanning operation compensates by
overlapping adjacent image fields in both the x and y
directions . An overlap amount greater than half the diameter of an average cell is preferred. In the
preferred embodiment, the overlap is specified as a
percentage of the image field in the x and y
directions .
The time to complete an image analysis can vary
depending upon the size of the scan area and the
number of candidate cells, or objects of interest
identified. For one example, in the preferred
embodiment, a complete image analysis of a scan area
of two square centimeters in which 50 objects of
interest are confirmed can be performed in about 12
to 15 minutes . This example includes not only
focusing, scanning, and image analysis, but also the
saving of 4Ox images as a mosaic on hard drive 21
(Fig. 2) . Consider the utility of the present
invention in a "rare event" application where there
may be one, two or a very small number of cells of
interest located somewhere on the slide. To
illustrate the nature of the problem by analogy, if
one were to scale a slide to the size of a football
field, a tumor cell, for example, would be about the size of a bottle cap. The problem is then to rapidly
search the football field and find the very small
number of bottle caps and have a high certainty that
none have been missed.
However the scan area is defined, an initial
focusing operation must be performed on each slide
prior to scanning. This is required since slides
differ, in general, in their placement in a carrier.
These differences include slight (but significant)
variations of tilt of the slide in its carrier.
Since each slide must remain in focus during
scanning, the degree of tilt of each slide must be
determined. This is accomplished with an initial
focusing operation that determines the exact degree
of tilt, so that focus can be maintained
automatically during scanning.
The initial focusing operation and other
focusing operations to be described later utilize a
focusing method based on processing of images
acquired by the system. This method was chosen for
its simplicity over other methods including use of IR beams reflected from the slide surface and use of
mechanical gauges . These other methods also would
not function properly when the specimen is protected
with a coverglass . The preferred method results in
lower system cost and improved reliability since no
additional parts need be included to perform
focusing. Fig. 13A provides a flow diagram
describing the "focus point" procedure. The basic
method relies on the fact that the pixel value
variance (or standard deviation) taken about the
pixel value mean is maximum at best focus . A
"brute-force" method could simply step through
focus, using the computer controlled Z or focus
stage, calculate the pixel variance at each step, and
return to the focus position providing the maximum
variance. Such a method would be too time consuming.
Therefore, additional features were added as shown in
Fig. 13A.
These features include the determination of
pixel variance at a relatively coarse number of focal
positions, and then the fitting of a curve to the data to provide a faster means of determining optimal
focus. This basic process is applied in two steps,
coarse and fine. During the coarse step at 220-230,
the Z stage is stepped over a user-specified range of
focus positions, with step sizes that are also user-
specified. It has been found that for coarse
focusing, these data are a close fit to a Gaussian
function. Therefore, this initial set of variance
versus focus position data are least-squares fit to a
Gaussian function at 228. The location of the peak
of this Gaussian curve determines the initial or
coarse estimate of focus position for input to step
232.
Following this, a second stepping operation 232-
242 is performed utilizing smaller steps over a
smaller focus range centered on the coarse focus
position. Experience indicates that data taken over
this smaller range are generally best fit by a second
order polynomial. Once this least squares fit is
performed at 240, the peak of the second order curve
provides the fine focus position at 244. Fig. 14 illustrates a procedure for how this
focusing method is utilized to determine the
orientation of a slide in its carrier. As shown,
focus positions are determined, as described above,
for a 3 x 3 grid of points centered on the scan area
at 264. Should one or more of these points lie
outside the scan area, the method senses at 266 this
by virtue of low values of pixel variance. In this
case, additional points are selected closer to the
center of the scan area. Fig. 15 shows the initial
array of points 80 and new point 82 selected closer
to the center. Once this array of focus positions is
determined at 268, a least squares plane is fit to
this data at 270. Focus points lying too far above
or below this best-fit plane are discarded at 272
(such as can occur from a dirty cover glass over the
scan area) , and the data is then refit . This plane
at 274 then provides the desired Z position
information for maintaining focus during scanning.
After determination of the best-fit focus plane,
the scan area is scanned in an X raster scan over the scan area as described earlier. During scanning, the
X stage is positioned to the starting point of the
scan area, the focus (Z) stage is positioned to the
best fit focus plane, an image is acquired and
processed as described herein, and this process is
repeated for all points over the scan area. In this
way, focus is maintained automatically without the
need for time-consuming refocusing at points during
scanning. Prior to confirmation of cell objects at a
40x or 60x level, a refocusing operation is conducted
since the use of this higher magnification requires
more precise focus than the best-fit plane provides.
Fig. 16 provides the flow diagram for this process.
As may be seen, this process is similar to the fine
focus method described earlier in that the object is
to maximize the image pixel variance. This is
accomplished by stepping through a range of focus
positions with the Z stage at 276, 278, calculating
the image variance at each position at 278, fitting a
second order polynomial to these data at 282, and
calculating the peak of this curve to yield an estimate of the best focus position at 284, 286.
This final focusing step differs from previous ones
in that the focus range and focus step sizes are
smaller since this magnification requires focus
settings to within 0.5 micron or better. It should
be noted that for some combinations of cell staining
characteristics, improved focus can be obtained by
numerically selecting the focus position that
provides the largest variance, as opposed to
selecting the peak of the polynomial. In such cases,
the polynomial is used to provide an estimate of best
focus, and a final step selects the actual Z position
giving highest pixel variance . It should also be
noted that if at any time during the focusing process
at 40x or 60x the parameters indicate that the focus
position is inadequate, the system automatically
reverts to a coarse focusing process as described
above with reference to Fig. 13A. This ensures that
variations in specimen thickness can be accommodated
in an expeditious manner. For some biological
specimens and stains, the focusing methods discussed above do not provide optimal focused results. For
example, certain white blood cells known as
neutrophils may be stained with Fast Red, a commonly
known stain, to identify alkaline phosphatase in the
cytoplasm of the cells . To further identify these
cells and the material within them, the specimen may
be counterstained with hemotoxylin to identify the
nucleus of the cells. In cells so treated, the
cytoplasm bearing alkaline phosphatase becomes a
shade of red proportionate to the amount of alkaline
phosphatase in the cytoplasm and the nucleus becomes
blue. However, where the cytoplasm and nucleus
overlap, the cell appears purple. These color
combinations appear to preclude the finding of a
focused Z position using the focus processes
discussed above.
In an effort to find a best focal position at
high magnification, a focus method, such as the one
shown in Fig. 13B, may be used. That method begins
by selecting a pixel near the center of a candidate
object of interest (Block 248) and defining a region of interest centered about the selected pixel (Block
250) . Preferably, the width of the region of
interest is a number of columns which is a power of
2. This width preference arises from subsequent
processing of the region of interest preferably using
a one dimensional Fast Fourier Transform (FFT)
technique. As is well known within, the art,
processing columns of pixel values using the FFT
technique is facilitated by making the number of
columns to be processed a power of two. While the
height of the region of interest is also a power of
two in the preferred embodiment, it need not be
unless a two dimensional FFT technique is used to
process the region of interest.
After the region of interest is selected, the
columns of pixel values are processed using the
preferred one-dimensional FFT to determine a spectra
of frequency components for the region of interest
(Block 252) . The frequency spectra ranges from DC to
some highest frequency component . For each frequency
component, a complex magnitude is computed. Preferably, the complex magnitudes for the frequency
components which range from approximately 25% of the
highest component to approximately 75% of the highest
component are squared and summed to determine the
total power for the region of interest (Block 254) .
Alternatively, the region of interest may be
processed with a smoothing window, such as a Hanning
window, to reduce the spurious high frequency
components generated by the FFT processing of the
pixel values in the region of interest . Such
preprocessing of the region of interest permits all
complex magnitude over the complete frequency range
to be squared and summed. After the power for a
region has been computed and stored (Block 256) , a
new focal position is selected, focus adjusted
(Blocks 258, 260) , and the process repeated. After
each focal position has been evaluated, the one
having the greatest power factor is selected as the
one best in focus (Block 262) .
The following describes the image processing
methods which are utilized to decide whether a candidate object of interest such as a stained tumor
cell is present in a given image, or field, during
the scanning process. Candidate objects of interest
which are detected during scanning are reimaged at
higher (40x or 60x) magnification, the decision
confirmed, and a region of interest for this cell
saved for later review by the pathologist . The image
processing includes color space conversion, low pass
filtering, background suppression, artifact
suppression, morphological processing, and blob
analysis. One or more of these steps can optionally
be eliminated. The operator is provided with an
option to configure the system to perform any or all
of these steps and whether to perform certain steps
more than once or several times in a row. It should
also be noted that the sequence of steps may be
varied and thereby optimized for specific reagents or
reagent combinations; however, the sequence described
herein is preferred. It should be noted that the
image processing steps of low pass filtering,
thresholding, morphological processing, and blob analysis are generally known image processing
building blocks.
An overview of the preferred process is shown in
Fig. 17A. The preferred process for identifying and
locating candidate objects of interest in a stained
biological specimen on a slide begins with an
acquisition of images obtained by scanning the slide
at low magnification (Block 288) . Each image is then
converted from a first color space to a second color
space (Block 290) and the color converted image is
low pass filtered (Block 292) . The pixels of the low
pass filtered image are then compared to a threshold
(Block 294) and, preferably, those pixels having a
value equal to or greater than the threshold are
identified as candidate object of interest pixels and
those less than the threshold are determined to be
artifact or background pixels. The candidate object
of interest pixels are then morphologically processed
to identify groups of candidate object of interest
pixels as candidate objects of interest (Block 296) .
These candidate objects of interest are then compared to blob analysis parameters (Block 298) to further
differentiate candidate objects of interest from
objects which do not conform to the blob analysis
parameters and, thus, do not warrant further
processing. The location of the candidate objects of
interest may be stored prior to confirmation at high
magnification. The process continues by determining
whether the candidate objects of interest have been
confirmed (Block 300) . If they have not been
confirmed, the optical system is set to high
magnification (Block 302) and images of the slide at
the locations corresponding to the candidate objects
of interest identified in the low magnification
images are acquired (Block 288) . These images are
then color converted (Block 290) , low pass filtered
(Block 292) , compared to a threshold (Block 294) ,
morphologically processed (Block 296) , and compared
to blob analysis parameters (Block 298) to confirm
which candidate objects of interest located from the
low magnification images are objects of interest. The coordinates of the objects of interest are then
stored for future reference (Block 303) .
In general, the candidate objects of interest,
such as tumor cells, are detected based on a
combination of characteristics, including size,
shape, and color. The chain of decision-making based
on these characteristics preferably begins with a
color space conversion process. The CCD camera
coupled to the microscope subsystem outputs a color
image comprising a matrix of 640 x 480 pixels. Each
pixel comprises red, green, and blue (ROB) signal
values .
It is desirable to transform the matrix of RGB
values to a different color space because the
difference between candidate objects of interest and
their background, such as tumor and normal cells, may
be determined from their respective colors.
Specimens are generally stained with one or more
industry standard stains (e.g., DAB, New Fuchsin,
AEC) which are "reddish" in color. Candidate
objects of interest retain more of the stain and thus appear red while normal cells remain unstained. The
specimens may also be counterstained with hematoxalin
so the nuclei of normal cells or cells not containing
an object of interest appear blue. In addition to
these objects, dirt and debris can appear as black,
gray, or can also be lightly stained red or blue
depending on the staining procedures utilized. The
residual plasma or other fluids also present on a
smear may also possess some color.
In the color conversion operation, a ratio of
two of the RGB signal values is formed to provide a
means for discriminating color information. With
three signal values for each pixel, nine different
ratios can be formed: R1R, R/G, RUB, G/G, G/B, G/R,
B/B, B/G, B/R. The optimal ratio to select depends
upon the range of color information expected in the
slide specimen. As noted above, typical stains used
for detecting candidate objects of interest such as
tumor cells are predominantly red, as opposed to
predominantly green or blue. Thus, the pixels of a
cell of interest which has been stained contain a red component which is larger than either the green or
blue components. A ratio of red divided by blue
(R/B) provides a value which is greater than one for
tumor cells but is approximately one for any clear or
white areas on the slide. Since the remaining cells,
i.e., normal cells, typically are stained blue, the
R/B ratio for pixels of these latter cells yields
values of less than one. The R/B ratio is preferred
for clearly separating the color information typical
in these applications .
Fig. 17B illustrates the flow diagram by which
this conversion is performed. In the interest of
processing speed, the conversion is implemented with
a look up table . The use of a look up table for
color conversion accomplishes three functions: (1)
performing a division operation; (2) scaling the
result for processing as an image having pixel values
ranging from 0 to 255; and (3) defining objects which
have low pixel values in each color band (R,G,B) as
"black" to avoid infinite ratios (i.e., dividing by
zero). These "black" objects are typically staining artifacts or can be edges of bubbles caused
by pasting a coverglass over the specimen.
Once the look up table is built at 304 for the
specific color ratio (i.e., choices of tumor and
nucleated cell stains) , each pixel in the original
RGB image is converted at 308 to produce the output.
Since it is of interest to separate the red stained
tumor cells from blue stained normal ones, the ratio
of color values is then scaled by a user specified
factor. As an example, for a factor of 128 and the
ratio of (red pixel value) / (blue pixel value), clear
areas on the slide would have a ratio of 1 scaled by
128 for a final X value of 128. Pixels which lie in
red stained tumor
cells would have X value greater than 128, while blue
stained nuclei of normal cells would have value less
than 128. In this way, the desired objects of
interest can be numerically discriminated. The
resulting 640 x 480 pixel matrix, referred to as the
X-image, is a gray scale image having values ranging
from 0 to 255. Other methods exist for discriminating color
information. One classical method converts the RGB
color information into another color space, such as
HSI (hue, saturation, intensity) space. In such a
space, distinctly different hues such as red, blue,
green, yellow, may be readily separated. In
addition, relatively lightly stained objects may be
distinguished from more intensely stained ones by
virtue of differing saturations. However, converting
from RGB space to HSI space requires more complex
computation. Conversion to a color ratio is faster;
for example, a full image can be converted by the
ratio technique of the present invention in about 30
ms while an HSI conversion can take several seconds.
In yet another approach, one could obtain color
information by taking a single color channel from the
camera. As an example, consider a blue channel, in
which objects that are red are relatively dark.
Objects which are blue, or white, are relatively
light in the blue channel. In principle, one could
take a single color channel, and simply set a threshold wherein everything darker than some
threshold is categorized as a candidate object of
interest, for example, a tumor cell, because it is
red and hence dark in the channel being reviewed.
However, one problem with the single channel approach
occurs where illumination is not uniform. Non-
uniformity of illumination results in non-uniformity
across the pixel values in any color channel, for
example, tending to peak in the middle of the image
and dropping off at the edges where the illumination
falls off. Performing thresholding on this non-
uniform color information runs into problems, as the
edges sometimes fall below the threshold, and
therefore it becomes more difficult to pick the
appropriate threshold level. However, with the ratio
technique, if the values of the red channel fall off
from center to edge, then the values of the blue
channel also fall off center to edge, resulting in a
uniform ratio, non-uniformities. Thus, the ratio
technique is more immune to illumination. As previously described, the color conversion
scheme is relatively insensitive to changes in color
balance, i.e., the relative outputs of the red,
green, and blue channels. However, some control is
necessary to avoid camera saturation, or inadequate
exposures in any one of the color bands. This color
balancing is performed automatically by utilizing a
calibration slide consisting of a clear area, and a
"dark" area having a known optical transmission or
density. The system obtains images from the clear
and "dark" areas, calculates "white" and "black"
adjustments for the image processor 25, and thereby
provides correct color balance.
In addition to the color balance control,
certain mechanical alignments are automated in this
process. The center point in the field of view for
the various microscope objectives as measured on the
slide can vary by several (or several tens of)
microns. This is the result of slight variations in
position of the microscope objectives 44a as
determined by the turret 44 (Fig. 4), small variations in alignment of the objectives with
respect to the system optical axis, and other
factors. Since it is desired that each microscope
objective be centered at the same point, these
mechanical offsets must be measured and automatically
compensated.
This is accomplished by imaging a test slide
which contains a recognizable feature or mark. An
image of this pattern is obtained by the system with
a given objective, and the position of the mark
determined. The system then rotates the turret to
the next lens objective, obtains an image of the test
object, and its position is redetermined. Apparent
changes in position of the test mark are recorded for
this objective. This process is continued for all
objectives. Once these spatial offsets have been
determined, they are automatically compensated for by
moving the stage 38 by an equal (but opposite) amount
of offset during changes in objective. In this way,
as different lens objectives are selected, there is
no apparent shift in center point or area viewed. A low pass filtering process precedes
thresholding. An objective of thresholding is to
obtain a pixel image matrix having only candidate
objects of interest, such as tumor cells above a
threshold level and everything else below it.
However, an actual acquired image will contain noise.
The noise can take several forms, including white
noise and artifacts. The microscope slide can have
small fragments of debris that pick up color in the
staining process and these are known as artifacts.
These artifacts are generally small and scattered
areas, on the order of a few pixels, which are above
the threshold. The purpose of low pass filtering is
to essentially blur or smear the entire color
converted image. The low pass filtering process will
smear artifacts more than larger objects of interest.
Such as tumor cells and thereby eliminate or reduce
the number of artifacts that pass the thresholding
process . The result is a cleaner thresholded image
downstream. In the low pass filter process, a 3 x 3
matrix of coefficients is applied to each pixel in the 640 x 480 x-image. A preferred coefficient
matrix is as follows:
1/9 1/9 1/9
1/9 1/9 1/9
1/9 1/9 1/9
At each pixel location, a 3 x 3 matrix comprising the
pixel of interest and its neighbors is multiplied by
the coefficient matrix and summed to yield a single
value for the pixel of interest . The output of this
spatial convolution process is again a 640 x 480
matrix. As an example, consider a case where the
center pixel and only the center pixel, has a value
of 255 and each of its other neighbors, top left,
top, top right and so forth, have values of 0.
This singular white pixel case corresponds to a
small object. The result of the matrix
multiplication and addition using the coefficient
matrix is a value of 1/9 (255) or 28 for the center
pixel, a value which is below the nominal threshold
of 128. Now consider another case in which all the
pixels have a value of 255 corresponding to a large object. Performing the low pass filtering operation
on a 3 x 3 matrix for this case yields a value of 255
for the center pixel. Thus, large objects retain
their values while small objects are reduced in
amplitude or eliminated. In the preferred method of
operation, the low pass filtering process is
performed on the X image twice in succession.
In order to separate objects of interest, such
as a tumor cell in the x image from other objects and
background, a thresholding operation is performed
designed to set pixels within cells of interest to a
value of 255, and all other areas to 0. Thresholding
ideally yields an image in which cells of interest
are white and the remainder of the image is black. A
problem one faces in thresholding is where to set the
threshold level. One cannot simply assume that cells
of interest are indicated by any pixel value above
the nominal threshold of 128. A typical imaging
system may use an incandescent halogen light bulb as
a light source. As the bulb ages, the relative
amounts of red and blue output can change. The tendency as the bulb ages is for the blue to drop off
more than the red and the green. To accommodate for
this light source variation over time, a dynamic
thresholding process is used whereby the threshold is
adjusted dynamically for each acquired image. Thus,
for each 640 x 480 image, a single threshold value is
derived specific to that image.
As shown in Fig. 18, the basic method is to
calculate, for each field, the mean X value, and the
standard deviation about this mean at 312. The
threshold is then set at 314 to the mean plus an
amount defined by the product of a (user specified)
factor and the standard deviation of the color
converted pixel values. The standard deviation
correlates to the structure and number of objects in
the image. Preferably, the user specified factor is
in the range of approximately 1.5 to 2.5. The factor
is selected to be in the lower end of the range for
slides in which the stain has primarily remained
within cell boundaries and the factor is selected to
be in the upper end of the range for slides in which the stain is pervasively present throughout the
slide. In this way, as areas are encountered on the
slide with greater or lower background intensities,
the threshold may be raised or lowered to help reduce
background objects. With this method, the threshold
changes in step with the aging of the light source
such that the effects of the aging are canceled out.
The image matrix resulting at 316 from the
thresholding step is a binary image of black (0) 5
and white (255) pixels.
As is often the case with thresholding
operations such as that described above, some
undesired areas will lie above the threshold value
due to noise, small stained cell fragments, and other
artifacts. It is desired and possible to eliminate
these artifacts by virtue of their small size
compared with legitimate cells of interest.
Morphological processes are utilized to perform this
function.
Morphological processing is similar to the low
pass filter convolution process described earlier except that it is applied to a binary image. Similar
to spatial convolution, the morphological process
traverses an input image matrix, pixel by pixel, and
places the processed pixels in an output matrix.
Rather than calculating a weighted sum of neighboring
pixels as m the low pass convolution process, the
morphological process uses set theory operations to
combine neighboring pixels in a nonlinear fashion.
Erosion is a process whereby a single pixel
layer is taken away from the edge of an object.
Dilation is the opposite process which adds a single
pixel layer to the edges of an object. The power of
morphological processing is that it provides for
further discrimination to eliminate small objects
that have survived the thresholding process and yet
are not likely tumor cells . The erosion and dilation
processes that make up a morphological "open"
preferably make small objects disappear yet allows
large objects to remain. Morphological processing of
binary images is described in detail in "Digital Image Processing", pages 127-137, G.A. Baxes, John
Wiley & Sons (1994) .
Fig. 19 illustrates the flow diagram for this
process. As shown here, a morphological "open"
process performs this suppression. A single
morphological open consists of a single morphological
erosion 320 followed by a single morphological
dilation 322. Multiple "opens" consist of multiple
erosions followed by multiple dilations. In the
preferred embodiment, one or two morphological opens
are found to be suitable . At this point in the
processing chain, the processed image contains
thresholded objects of interest, such as tumor cells
(if any were present in the original image) , and
possibly some residual artifacts that were too large
to be eliminated by the processes above.
Fig. 20 provides a flow diagram illustrating a
blob analysis performed to determine the number,
size, and location of objects in the thresholded
image. A blob is defined as a region of connected
pixels having the same "color," in this case, a value of 255. Processing is performed over the
entire image to determine the number of such regions
at 324 and to determine the area and x,y coordinates
for each detected blob at 326. Comparison of the
size of each blob to a known minimum area at 328 for
a tumor cell allows a refinement in decisions about
which objects are objects of interest, such as tumor
cells, and which are artifacts. The location
(x,y coordinates) of objects identified as cells of
interest in this stage are saved for the final 40x
reimaging step described below. Objects not passing
the size test are disregarded as artifacts.
The processing chain described above identifies
objects at the scanning magnification as cells of
interest candidates. As illustrated in Fig. 21, at
the completion of scanning, the system switches to
the 4Ox magnification objective at 330, and each
candidate is reimaged to confirm the identification
332. Each 40x image is reprocessed at 334 using the
same steps as described above but with test
parameters suitably modified for the higher magnification (e.g., area). At 336, a region of
interest centered on each confirmed cell is saved to
the hard drive for review by the pathologist .
As noted earlier, a mosaic of saved images is
made available for viewing by the pathologist. As
shown in Fig. 22, a series of images of cells which
have been confirmed by the image analysis is
presented in the mosaic 150. The pathologist can
then visually inspect the images to make a
determination whether to accept (152) or reject (153)
each cell image. Such a 5 determination can be noted
and saved with the mosaic of images for generating a
printed report .
In addition to saving the image of the cell and
its region, the cell coordinates are saved should the
pathologist wish to directly view the cell through
the oculars or on the image monitor. In this case,
the pathologist reloads the slide carrier, selects
the slide and cell for review from a mosaic of cell
images, and the system automatically positions the
cell under the microscope for viewing. It has been found that normal cells whose nuclei
have been stained with hematoxylin are often quite
numerous , numbering in the thousands per lOx image .
Since these cells are so numerous, and since they
tend to clump, counting each individual nucleated
cell would add an excessive processing burden, at the
expense of speed, and would not necessarily provide
an accurate count due to clumping. The apparatus
performs an estimation process in which the total
area of each field that is stained hematoxylin blue
is measured and this area is divided by the average
size of a nucleated cell. Fig. 23 outlines this
process. In this process, a single color band (the
red channel provides the best contrast for blue
stained nucleated cells) is processed by calculating
the average pixel value for each field at 342,
establishing two threshold values (high and low) as
indicated at 344, 346, and counting the number of
pixels between these two values at 348. In the
absence of dirt, or other opaque debris, this
provides a count of the number of predominantly blue pixels . By dividing this value by the average area
for a nucleated cell at 350, and looping over all
fields at 352, an approximate cell count is obtained.
Preliminary testing of this process indicates an
accuracy with +/- 15%. It should be noted that for
some slide preparation techniques, the size of
nucleated cells can be significantly larger than the
typical size. The operator can select the
appropriate nucleated cell size to compensate for
these characteristics.
As with any imaging system, there is some loss
of modulation transfer (i.e., contrast) due to the
modulation transfer function (MTF) characteristics of
the imaging optics, camera, electronics, and other
components. Since it is desired to save "high
quality" images of cells of interest both for
pathologist review and for archival purposes, it is
desired to compensate for these MTF losses. An MTF
compensation, or MTFC, is performed as a digital
process applied to the acquired digital images. A
digital filter is utilized to restore the high spatial frequency content of the images upon storage,
while maintaining low noise levels. With this MTFC
technology, image quality is enhanced, or restored,
through the use of digital processing methods as
opposed to conventional oil-immersion or other
hardware based methods. MTFC is described further in
"The Image Processing Handbook," pages 225 and 337,
J. C. Rues, CRC Press (1995) .
Referring to Fig. 24, the functions available in
a user interface of the apparatus 10 are shown. From
the user interface, which is presented graphically on
computer monitor 26, an operator can select among
apparatus functions which include acquisition 402,
analysts 404, and system configuration 406. At the
acquisition level 402, the operator can select
between manual 408 and automatic 410 modes of
operation. In the manual mode, the operator is
presented with manual operations 409. Patient
information 414 regarding an assay can be entered at
412. In the analysis level 404, review 416 and
report 418 functions are made available. At the review level 416, the operator can select a montage
function 420. At this montage level, a pathologist
can perform diagnostic review functions including
visiting an image 422, accept/reject of cells 424,
nucleated cell counting 426, accept/reject of cell
counts 428, and saving of pages at 430. The report
level 418 allows an operator to generate patient
reports 432. In the configuration level 406, the
operator can select to configure preferences at 434,
input operator information 437 at 436, create a
system log at 438, and toggle a menu panel at 440.
The configuration preferences include scan area
selection functions at 442, 452; montage
specifications at 444, bar code handling at 446,
default cell counting at 448, stain selection at 450,
and scan objective selection at 454.
Computer Implementation
Aspects of the invention may be implemented in
hardware or software, or a combination of both.
However, preferably, the algorithms and processes of
the invention are implemented in one or more computer
programs executing on programmable computers each
comprising at least one processor, at least one data
storage system (including volatile and non-volatile
memory and/or storage elements) , at least one input
device, and at least one output device. Program code
is applied to input data to perform the functions
described herein and generate output information.
The output information is applied to one or more
output devices, in known fashion.
Each program may be implemented in any desired
computer language (including machine, assembly, high
level procedural, or object oriented programming
languages) to communicate with a computer system. In
any case, the language may be a compiled or
interpreted language . Each such computer program is preferably stored
on a storage media or device (e.g., ROM, CD-ROM,
tape, or magnetic diskette) readable by a general or
special purpose programmable computer, for
configuring and operating the computer when the
storage media or device is read by the computer to
perform the procedures described herein. The
inventive system may also be considered to be
implemented as a computer-readable storage medium,
configured with a computer program, where the storage
medium so configured causes a computer to operate in
a specific and predefined manner to perform the
functions described herein.
A number of embodiments of the present invention
have been described. Nevertheless, various
modifications may be made without departing from the
spirit and scope of the invention. Accordingly, the
invention is not to be limited by the specific
illustrated embodiment, but only by the scope of the
appended claims .

Claims

CLAIMSWhat is claimed:
1. A method for automated detection of a cell
proliferative disorder, comprising,
providing a sample on a slide, wherein the sample is
stained with an antibody to a protein associated with
a cell proliferative disorder;
identifying a processing parameter for the
stained sample;
scanning the stained sample at a plurality
of locations at a low magnification on an
optical system;
acquiring a low magnification image at the
low magnification at each location in the
scanned area;
processing each low magnification image to
detect a candidate objects of interest;
storing stage coordinates of each location
for each candidate object of interest; adjusting the optical system to a higher
magnification;
repositioning the stage to the location for
each candidate object of interest;
acquiring a higher magnification image of
each candidate object of interest; and
storing each higher magnification image.
2. The method of claim 1, wherein the cell
proliferative disorder is a neoplasm.
3. The method of claim 1, wherein the cell
proliferative disorder is breast cancer.
4. The method of claim 1, wherein the antibody is
selected from the group consisting of an anti-
HER2/neu antibody, anti-estrogen receptor
antibody, anti-progesterone receptor antibody,
anti-p53 antibody and anti-cyclin Dl antibody.
5. The method of claim 1, wherein the processing
parameter is selected from the group consisting
of a positive control, negative control or test
sample .
6. The method of claim 1, wherein the detection of
the candidate object of interest is by size,
color or shape .
7. An automated system for quantification of a cell
proliferative disorder in a sample, comprising:
a slide configured to accept a sample,
wherein the sample is stained with an antibody
to a protein associated with a cell
proliferative disorder;
a code associated with the slide for
identification of the slide's processing
parameters ;
a lens for magnification of the sample on
the slide; a stage configured to hold the slide,
wherein the stage is optically coupled to the
lens ;
a camera optically coupled to the lens
configured to acquire an image from the lens;
a computer program residing on a computer-
readable medium for
executing the processing parameters of
the code;
selecting a position on the stage
identifying a candidate object of
interest based upon color, shape or size;
obtaining a low magnification image of
the candidate object of interest;
storing the position of the candidate
object of interest;
adjusting the magnification of the
lens ;
repositioning the stage to the position
of the candidate object of interest; obtaining a higher magnification image
of the candidate object of interest;
storing the higher magnification image;
allowing retrieval of the images
acquired; and
a monitor configured to view the
images .
8. The system of claim 7, wherein the code is a bar
code .
9. The system of claim 7, wherein the lens is an
objective of a microscope.
10. The system of claim 7, wherein the camera is a
CCD camera .
11. A computer program, residing on a computer-
readable medium, comprising instructions for
causing a computer to:
execute a processing parameter of a slide; select a position on a stage of an optical
system;
identify a candidate object of interest
based upon color, shape or size;
obtain a low magnification image of the
candidate object of interest ,-
store the position of the candidate object
of interest;
adjust the magnification of the optical
system;
reposition the stage to the position of the
candidate object of interest;
obtain a higher magnification image of the
candidate object of interest;
store the higher magnification image; and
allow retrieval of the images acquired.
12. An apparatus comprising a computer-readable
storage medium tangibly embodying program
instruction for quantifying a cell proliferative
disorder, the program instruction inducing instruction operable for causing an optical
system to:
execute a processing parameter of a slide;
select a position on a stage of an optical
system;
identify a candidate object of interest
based upon color, shape or size;
obtain a low magnification image of the
candidate object of interest;
store the position of the candidate object
of interest;
adjust the magnification of the optical
system;
reposition the stage to the position of the
candidate object of interest;
obtain a higher magnification image of the
candidate object of interest;
store the higher magnification image; and
allow retrieval of the images acquired.
3. The apparatus of claim 12, wherein the image is
a digital image .
PCT/US2000/019046 1999-07-13 2000-07-12 Automated detection of objects in a biological sample WO2001004828A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP2001510159A JP2003504627A (en) 1999-07-13 2000-07-12 Automatic detection of objects in biological samples
AU60934/00A AU6093400A (en) 1999-07-13 2000-07-12 Automated detection of objects in a biological sample
EP00947299A EP1203343A4 (en) 1999-07-13 2000-07-12 Automated detection of objects in a biological sample

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US14382399P 1999-07-13 1999-07-13
US60/143,823 1999-07-13

Publications (1)

Publication Number Publication Date
WO2001004828A1 true WO2001004828A1 (en) 2001-01-18

Family

ID=22505818

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/019046 WO2001004828A1 (en) 1999-07-13 2000-07-12 Automated detection of objects in a biological sample

Country Status (4)

Country Link
EP (1) EP1203343A4 (en)
JP (1) JP2003504627A (en)
AU (1) AU6093400A (en)
WO (1) WO2001004828A1 (en)

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004013658A2 (en) * 2002-07-30 2004-02-12 Deutsches Krebsforschungszentrum Method and apparatus for multiple labeling detection and evaluation of a plurality of particles
WO2004017052A3 (en) * 2002-08-15 2004-06-03 Qinetiq Ltd Histological assessment
WO2004051554A1 (en) 2002-11-27 2004-06-17 3M Innovative Properties Company Biological growth plate scanner with automated image processing profile selection
GB2400205A (en) * 2003-03-05 2004-10-06 Fairfield Imaging Ltd Analysis of pathological samples and provision of digital images relating to the samples
EP1572979A1 (en) * 2002-12-20 2005-09-14 DakoCytomation Denmark A/S A method and apparatus for automatic staining of tissue samples
EP1804046A2 (en) * 2005-12-28 2007-07-04 Sysmex Corporation Sample image obtaining apparatus, sample image obtaining system, and method for obtaining sample image
EP1845359A2 (en) * 2001-10-29 2007-10-17 Pola Chemical Industries, Inc. Method for preparing corneocyte specimens and staining corneocytes, and skin analysis system
JP2010060564A (en) * 2002-08-15 2010-03-18 Qinetiq Ltd Histological assessment
CN102362168A (en) * 2009-03-27 2012-02-22 索尼公司 Observation device
EP1927860A3 (en) * 2006-11-30 2013-03-06 Sysmex Corporation Blood image analyzer
US8922781B2 (en) 2004-11-29 2014-12-30 The General Hospital Corporation Arrangements, devices, endoscopes, catheters and methods for performing optical imaging by simultaneously illuminating and detecting multiple points on a sample
US8928889B2 (en) 2005-09-29 2015-01-06 The General Hospital Corporation Arrangements and methods for providing multimodality microscopic imaging of one or more biological structures
US9060689B2 (en) 2005-06-01 2015-06-23 The General Hospital Corporation Apparatus, method and system for performing phase-resolved optical frequency domain imaging
US9069130B2 (en) 2010-05-03 2015-06-30 The General Hospital Corporation Apparatus, method and system for generating optical radiation from biological gain media
US9186066B2 (en) 2006-02-01 2015-11-17 The General Hospital Corporation Apparatus for applying a plurality of electro-magnetic radiations to a sample
US9282931B2 (en) 2000-10-30 2016-03-15 The General Hospital Corporation Methods for tissue analysis
US9326682B2 (en) 2005-04-28 2016-05-03 The General Hospital Corporation Systems, processes and software arrangements for evaluating information associated with an anatomical structure by an optical coherence ranging technique
US9330092B2 (en) 2011-07-19 2016-05-03 The General Hospital Corporation Systems, methods, apparatus and computer-accessible-medium for providing polarization-mode dispersion compensation in optical coherence tomography
US9341783B2 (en) 2011-10-18 2016-05-17 The General Hospital Corporation Apparatus and methods for producing and/or providing recirculating optical delay(s)
EP2946198A4 (en) * 2013-01-21 2016-07-27 Univ Cornell Smartphone-based apparatus and method for obtaining repeatable, quantitative colorimetric measurement
US9408539B2 (en) 2010-03-05 2016-08-09 The General Hospital Corporation Systems, methods and computer-accessible medium which provide microscopic images of at least one anatomical structure at a particular resolution
US9415550B2 (en) 2012-08-22 2016-08-16 The General Hospital Corporation System, method, and computer-accessible medium for fabrication miniature endoscope using soft lithography
US9441948B2 (en) 2005-08-09 2016-09-13 The General Hospital Corporation Apparatus, methods and storage medium for performing polarization-based quadrature demodulation in optical coherence tomography
US9510758B2 (en) 2010-10-27 2016-12-06 The General Hospital Corporation Apparatus, systems and methods for measuring blood pressure within at least one vessel
US9516997B2 (en) 2006-01-19 2016-12-13 The General Hospital Corporation Spectrally-encoded endoscopy techniques, apparatus and methods
US9541504B2 (en) 2010-08-05 2017-01-10 Cambridge Research & Instrumentation, Inc. Enhancing visual assessment of samples
US9557154B2 (en) 2010-05-25 2017-01-31 The General Hospital Corporation Systems, devices, methods, apparatus and computer-accessible media for providing optical imaging of structures and compositions
US9615748B2 (en) 2009-01-20 2017-04-11 The General Hospital Corporation Endoscopic biopsy apparatus, system and method
US9629528B2 (en) 2012-03-30 2017-04-25 The General Hospital Corporation Imaging system, method and distal attachment for multidirectional field of view endoscopy
USRE46412E1 (en) 2006-02-24 2017-05-23 The General Hospital Corporation Methods and systems for performing angle-resolved Fourier-domain optical coherence tomography
US9664615B2 (en) 2004-07-02 2017-05-30 The General Hospital Corporation Imaging system and related techniques
US9733460B2 (en) 2014-01-08 2017-08-15 The General Hospital Corporation Method and apparatus for microscopic imaging
US9763623B2 (en) 2004-08-24 2017-09-19 The General Hospital Corporation Method and apparatus for imaging of vessel segments
US9784681B2 (en) 2013-05-13 2017-10-10 The General Hospital Corporation System and method for efficient detection of the phase and amplitude of a periodic modulation associated with self-interfering fluorescence
US9795301B2 (en) 2010-05-25 2017-10-24 The General Hospital Corporation Apparatus, systems, methods and computer-accessible medium for spectral analysis of optical coherence tomography images
US9933446B2 (en) 2008-03-04 2018-04-03 3M Innovative Properties Company Processing of biological growth media based on measured manufacturing characteristics
US9968245B2 (en) 2006-10-19 2018-05-15 The General Hospital Corporation Apparatus and method for obtaining and providing imaging information associated with at least one portion of a sample, and effecting such portion(s)
US9968261B2 (en) 2013-01-28 2018-05-15 The General Hospital Corporation Apparatus and method for providing diffuse spectroscopy co-registered with optical frequency domain imaging
US10058250B2 (en) 2013-07-26 2018-08-28 The General Hospital Corporation System, apparatus and method for utilizing optical dispersion for fourier-domain optical coherence tomography
US10117576B2 (en) 2013-07-19 2018-11-06 The General Hospital Corporation System, method and computer accessible medium for determining eye motion by imaging retina and providing feedback for acquisition of signals from the retina
US10169861B2 (en) 2013-12-27 2019-01-01 Fujifilm Corporation Image processing apparatus, non-transitory computer readable medium, and image processing method
US10228556B2 (en) 2014-04-04 2019-03-12 The General Hospital Corporation Apparatus and method for controlling propagation and/or transmission of electromagnetic radiation in flexible waveguide(s)
US10285568B2 (en) 2010-06-03 2019-05-14 The General Hospital Corporation Apparatus and method for devices for imaging structures in or at one or more luminal organs
US10413175B2 (en) 2006-05-10 2019-09-17 The General Hospital Corporation Process, arrangements and systems for providing frequency domain imaging of a sample
US10426548B2 (en) 2006-02-01 2019-10-01 The General Hosppital Corporation Methods and systems for providing electromagnetic radiation to at least one portion of a sample using conformal laser therapy procedures
US10478072B2 (en) 2013-03-15 2019-11-19 The General Hospital Corporation Methods and system for characterizing an object
US10736494B2 (en) 2014-01-31 2020-08-11 The General Hospital Corporation System and method for facilitating manual and/or automatic volumetric imaging with real-time tension or force feedback using a tethered imaging device
US10835110B2 (en) 2008-07-14 2020-11-17 The General Hospital Corporation Apparatus and method for facilitating at least partial overlap of dispersed ration on at least one sample
US10893806B2 (en) 2013-01-29 2021-01-19 The General Hospital Corporation Apparatus, systems and methods for providing information regarding the aortic valve
US10912462B2 (en) 2014-07-25 2021-02-09 The General Hospital Corporation Apparatus, devices and methods for in vivo imaging and diagnosis
US11179028B2 (en) 2013-02-01 2021-11-23 The General Hospital Corporation Objective lens arrangement for confocal endomicroscopy
US11452433B2 (en) 2013-07-19 2022-09-27 The General Hospital Corporation Imaging apparatus and method which utilizes multidirectional field of view endoscopy
US11490826B2 (en) 2009-07-14 2022-11-08 The General Hospital Corporation Apparatus, systems and methods for measuring flow and pressure within a vessel
US11490797B2 (en) 2012-05-21 2022-11-08 The General Hospital Corporation Apparatus, device and method for capsule microscopy
CN115541578A (en) * 2022-09-28 2022-12-30 佐健(上海)生物医疗科技有限公司 High-throughput super-resolution rapid scanning and analyzing system for cervical cell pathological section

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2333521B1 (en) 2001-04-30 2019-12-04 The General Hospital Corporation Method and apparatus for improving image clarity and sensitivity in optical coherence tomography using dynamic feedback to control focal properties and coherence gating
EP1611470B1 (en) 2003-03-31 2015-10-14 The General Hospital Corporation Speckle reduction in optical coherence tomography by path length encoded angular compounding
EP2011434A3 (en) 2003-06-06 2009-03-25 The General Hospital Corporation Process and apparatus for a wavelength tuned light source
DE102004009934A1 (en) * 2004-02-26 2005-09-22 Niendorf, Axel, Prof.Dr.med. Method for examining a tissue sample
WO2006024014A2 (en) 2004-08-24 2006-03-02 The General Hospital Corporation Process, system and software arrangement for measuring a mechanical strain and elastic properties of a sample
WO2006078928A2 (en) * 2005-01-18 2006-07-27 Trestle Corporation System and method for creating variable quality images of a slide
WO2007013551A1 (en) * 2005-07-29 2007-02-01 Olympus Corporation Light intensity measuring method and light intensity measuring device
WO2007084995A2 (en) 2006-01-19 2007-07-26 The General Hospital Corporation Methods and systems for optical imaging of epithelial luminal organs by beam scanning thereof
JP4911172B2 (en) * 2006-07-12 2012-04-04 東洋紡績株式会社 Analytical apparatus and use thereof
JP4938428B2 (en) * 2006-12-01 2012-05-23 シスメックス株式会社 Specimen image creation method and apparatus
WO2008108059A1 (en) * 2007-03-01 2008-09-12 Nec Corporation Breast cancer pathologic image diagnosis supporting system, breast cancer pathologic image diagnosis supporting method, beast cancer pathologic image diagnosis supporting program, and recording medium in which brest cancer pathologic image diagnosis supporting program is recorded
WO2008118781A2 (en) 2007-03-23 2008-10-02 The General Hospital Corporation Methods, arrangements and apparatus for utilizing a wavelength-swept laser using angular scanning and dispersion procedures
US10534129B2 (en) 2007-03-30 2020-01-14 The General Hospital Corporation System and method providing intracoronary laser speckle imaging for the detection of vulnerable plaque
WO2009018456A2 (en) 2007-07-31 2009-02-05 The General Hospital Corporation Systems and methods for providing beam scan patterns for high speed doppler optical frequency domain imaging
CH708797B1 (en) * 2007-08-17 2015-05-15 Tecan Trading Ag Sample part magazine for a slide transport device of a laser scanner device.
WO2009125547A1 (en) * 2008-04-09 2009-10-15 株式会社ニコン Culture apparatus controller and control program
US8937724B2 (en) 2008-12-10 2015-01-20 The General Hospital Corporation Systems and methods for extending imaging depth range of optical coherence tomography through optical sub-sampling
CA2749670A1 (en) 2009-02-04 2010-08-12 The General Hospital Corporation Apparatus and method for utilization of a high-speed optical wavelength tuning source
KR102492083B1 (en) * 2021-10-21 2023-01-27 주식회사 이뮤니스바이오 Cell count method

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5428690A (en) * 1991-09-23 1995-06-27 Becton Dickinson And Company Method and apparatus for automated assay of biological specimens
US5888742A (en) * 1997-10-28 1999-03-30 Incyte Pharmaceuticals, Inc. Human phospholipid binding proteins
US5889881A (en) * 1992-10-14 1999-03-30 Oncometrics Imaging Corp. Method and apparatus for automatically detecting malignancy-associated changes

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW203120B (en) * 1985-10-04 1993-04-01 Abbott Lab
JP2873015B2 (en) * 1989-04-27 1999-03-24 株式会社京都第一科学 Analysis equipment
EP0864082B1 (en) * 1995-11-30 2003-04-02 Chromavision Medical Systems, Inc. Method for automated image analysis of biological specimens

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5428690A (en) * 1991-09-23 1995-06-27 Becton Dickinson And Company Method and apparatus for automated assay of biological specimens
US5889881A (en) * 1992-10-14 1999-03-30 Oncometrics Imaging Corp. Method and apparatus for automatically detecting malignancy-associated changes
US5888742A (en) * 1997-10-28 1999-03-30 Incyte Pharmaceuticals, Inc. Human phospholipid binding proteins

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP1203343A4 *

Cited By (73)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9282931B2 (en) 2000-10-30 2016-03-15 The General Hospital Corporation Methods for tissue analysis
EP1845359A2 (en) * 2001-10-29 2007-10-17 Pola Chemical Industries, Inc. Method for preparing corneocyte specimens and staining corneocytes, and skin analysis system
US7425427B2 (en) 2001-10-29 2008-09-16 Pola Chemical Industries Inc. Method for staining corneocytes, method for preparing corneocytes specimen and skin analysis system
US7421105B2 (en) 2001-10-29 2008-09-02 Pola Chemical Industries Inc. Method for staining corneocytes, method for preparing corneocytes specimen and skin analysis system
EP1845359A3 (en) * 2001-10-29 2007-11-14 Pola Chemical Industries, Inc. Method for preparing corneocyte specimens and staining corneocytes, and skin analysis system
WO2004013658A3 (en) * 2002-07-30 2004-05-21 Steinbeis Transferzentrum Anal Method and apparatus for multiple labeling detection and evaluation of a plurality of particles
WO2004013658A2 (en) * 2002-07-30 2004-02-12 Deutsches Krebsforschungszentrum Method and apparatus for multiple labeling detection and evaluation of a plurality of particles
JP2010060564A (en) * 2002-08-15 2010-03-18 Qinetiq Ltd Histological assessment
US7079675B2 (en) 2002-08-15 2006-07-18 Qinetiq Limited Histological assessment
WO2004017052A3 (en) * 2002-08-15 2004-06-03 Qinetiq Ltd Histological assessment
EP1573654A1 (en) * 2002-11-27 2005-09-14 3M Innovative Properties Company Biological growth plate scanner with automated image processing profile selection
EP1573654A4 (en) * 2002-11-27 2007-12-12 3M Innovative Properties Co Biological growth plate scanner with automated image processing profile selection
US7901933B2 (en) 2002-11-27 2011-03-08 3M Innovative Properties Company Methods of processing a biological growth plate in a biological growth plate scanner
EP2325778A1 (en) * 2002-11-27 2011-05-25 3M Innovative Properties Company Biological growth plate scanner with automated image processing profile selection
WO2004051554A1 (en) 2002-11-27 2004-06-17 3M Innovative Properties Company Biological growth plate scanner with automated image processing profile selection
EP1572979A1 (en) * 2002-12-20 2005-09-14 DakoCytomation Denmark A/S A method and apparatus for automatic staining of tissue samples
EP1572979A4 (en) * 2002-12-20 2011-10-19 Dakocytomation Denmark As A method and apparatus for automatic staining of tissue samples
US9599630B2 (en) 2002-12-20 2017-03-21 Dako Denmark A/S Method and apparatus for automatic staining of tissue samples
GB2400205A (en) * 2003-03-05 2004-10-06 Fairfield Imaging Ltd Analysis of pathological samples and provision of digital images relating to the samples
US9664615B2 (en) 2004-07-02 2017-05-30 The General Hospital Corporation Imaging system and related techniques
US9763623B2 (en) 2004-08-24 2017-09-19 The General Hospital Corporation Method and apparatus for imaging of vessel segments
US8922781B2 (en) 2004-11-29 2014-12-30 The General Hospital Corporation Arrangements, devices, endoscopes, catheters and methods for performing optical imaging by simultaneously illuminating and detecting multiple points on a sample
US9326682B2 (en) 2005-04-28 2016-05-03 The General Hospital Corporation Systems, processes and software arrangements for evaluating information associated with an anatomical structure by an optical coherence ranging technique
US9060689B2 (en) 2005-06-01 2015-06-23 The General Hospital Corporation Apparatus, method and system for performing phase-resolved optical frequency domain imaging
US9441948B2 (en) 2005-08-09 2016-09-13 The General Hospital Corporation Apparatus, methods and storage medium for performing polarization-based quadrature demodulation in optical coherence tomography
US8928889B2 (en) 2005-09-29 2015-01-06 The General Hospital Corporation Arrangements and methods for providing multimodality microscopic imaging of one or more biological structures
US9513276B2 (en) 2005-09-29 2016-12-06 The General Hospital Corporation Method and apparatus for optical imaging via spectral encoding
EP1804046A3 (en) * 2005-12-28 2013-02-20 Sysmex Corporation Sample image obtaining apparatus, sample image obtaining system, and method for obtaining sample image
EP1804046A2 (en) * 2005-12-28 2007-07-04 Sysmex Corporation Sample image obtaining apparatus, sample image obtaining system, and method for obtaining sample image
US9516997B2 (en) 2006-01-19 2016-12-13 The General Hospital Corporation Spectrally-encoded endoscopy techniques, apparatus and methods
US10426548B2 (en) 2006-02-01 2019-10-01 The General Hosppital Corporation Methods and systems for providing electromagnetic radiation to at least one portion of a sample using conformal laser therapy procedures
US9186066B2 (en) 2006-02-01 2015-11-17 The General Hospital Corporation Apparatus for applying a plurality of electro-magnetic radiations to a sample
USRE46412E1 (en) 2006-02-24 2017-05-23 The General Hospital Corporation Methods and systems for performing angle-resolved Fourier-domain optical coherence tomography
US10413175B2 (en) 2006-05-10 2019-09-17 The General Hospital Corporation Process, arrangements and systems for providing frequency domain imaging of a sample
US9968245B2 (en) 2006-10-19 2018-05-15 The General Hospital Corporation Apparatus and method for obtaining and providing imaging information associated with at least one portion of a sample, and effecting such portion(s)
EP1927860A3 (en) * 2006-11-30 2013-03-06 Sysmex Corporation Blood image analyzer
US9933446B2 (en) 2008-03-04 2018-04-03 3M Innovative Properties Company Processing of biological growth media based on measured manufacturing characteristics
US10835110B2 (en) 2008-07-14 2020-11-17 The General Hospital Corporation Apparatus and method for facilitating at least partial overlap of dispersed ration on at least one sample
US9615748B2 (en) 2009-01-20 2017-04-11 The General Hospital Corporation Endoscopic biopsy apparatus, system and method
CN102362168A (en) * 2009-03-27 2012-02-22 索尼公司 Observation device
US11490826B2 (en) 2009-07-14 2022-11-08 The General Hospital Corporation Apparatus, systems and methods for measuring flow and pressure within a vessel
US10463254B2 (en) 2010-03-05 2019-11-05 The General Hospital Corporation Light tunnel and lens which provide extended focal depth of at least one anatomical structure at a particular resolution
US9642531B2 (en) 2010-03-05 2017-05-09 The General Hospital Corporation Systems, methods and computer-accessible medium which provide microscopic images of at least one anatomical structure at a particular resolution
US9408539B2 (en) 2010-03-05 2016-08-09 The General Hospital Corporation Systems, methods and computer-accessible medium which provide microscopic images of at least one anatomical structure at a particular resolution
US9069130B2 (en) 2010-05-03 2015-06-30 The General Hospital Corporation Apparatus, method and system for generating optical radiation from biological gain media
US9951269B2 (en) 2010-05-03 2018-04-24 The General Hospital Corporation Apparatus, method and system for generating optical radiation from biological gain media
US10939825B2 (en) 2010-05-25 2021-03-09 The General Hospital Corporation Systems, devices, methods, apparatus and computer-accessible media for providing optical imaging of structures and compositions
US9557154B2 (en) 2010-05-25 2017-01-31 The General Hospital Corporation Systems, devices, methods, apparatus and computer-accessible media for providing optical imaging of structures and compositions
US9795301B2 (en) 2010-05-25 2017-10-24 The General Hospital Corporation Apparatus, systems, methods and computer-accessible medium for spectral analysis of optical coherence tomography images
US10285568B2 (en) 2010-06-03 2019-05-14 The General Hospital Corporation Apparatus and method for devices for imaging structures in or at one or more luminal organs
US9541504B2 (en) 2010-08-05 2017-01-10 Cambridge Research & Instrumentation, Inc. Enhancing visual assessment of samples
US9510758B2 (en) 2010-10-27 2016-12-06 The General Hospital Corporation Apparatus, systems and methods for measuring blood pressure within at least one vessel
US9330092B2 (en) 2011-07-19 2016-05-03 The General Hospital Corporation Systems, methods, apparatus and computer-accessible-medium for providing polarization-mode dispersion compensation in optical coherence tomography
US9341783B2 (en) 2011-10-18 2016-05-17 The General Hospital Corporation Apparatus and methods for producing and/or providing recirculating optical delay(s)
US9629528B2 (en) 2012-03-30 2017-04-25 The General Hospital Corporation Imaging system, method and distal attachment for multidirectional field of view endoscopy
US11490797B2 (en) 2012-05-21 2022-11-08 The General Hospital Corporation Apparatus, device and method for capsule microscopy
US9415550B2 (en) 2012-08-22 2016-08-16 The General Hospital Corporation System, method, and computer-accessible medium for fabrication miniature endoscope using soft lithography
EP2946198A4 (en) * 2013-01-21 2016-07-27 Univ Cornell Smartphone-based apparatus and method for obtaining repeatable, quantitative colorimetric measurement
US9968261B2 (en) 2013-01-28 2018-05-15 The General Hospital Corporation Apparatus and method for providing diffuse spectroscopy co-registered with optical frequency domain imaging
US10893806B2 (en) 2013-01-29 2021-01-19 The General Hospital Corporation Apparatus, systems and methods for providing information regarding the aortic valve
US11179028B2 (en) 2013-02-01 2021-11-23 The General Hospital Corporation Objective lens arrangement for confocal endomicroscopy
US10478072B2 (en) 2013-03-15 2019-11-19 The General Hospital Corporation Methods and system for characterizing an object
US9784681B2 (en) 2013-05-13 2017-10-10 The General Hospital Corporation System and method for efficient detection of the phase and amplitude of a periodic modulation associated with self-interfering fluorescence
US10117576B2 (en) 2013-07-19 2018-11-06 The General Hospital Corporation System, method and computer accessible medium for determining eye motion by imaging retina and providing feedback for acquisition of signals from the retina
US11452433B2 (en) 2013-07-19 2022-09-27 The General Hospital Corporation Imaging apparatus and method which utilizes multidirectional field of view endoscopy
US10058250B2 (en) 2013-07-26 2018-08-28 The General Hospital Corporation System, apparatus and method for utilizing optical dispersion for fourier-domain optical coherence tomography
US10169861B2 (en) 2013-12-27 2019-01-01 Fujifilm Corporation Image processing apparatus, non-transitory computer readable medium, and image processing method
US9733460B2 (en) 2014-01-08 2017-08-15 The General Hospital Corporation Method and apparatus for microscopic imaging
US10736494B2 (en) 2014-01-31 2020-08-11 The General Hospital Corporation System and method for facilitating manual and/or automatic volumetric imaging with real-time tension or force feedback using a tethered imaging device
US10228556B2 (en) 2014-04-04 2019-03-12 The General Hospital Corporation Apparatus and method for controlling propagation and/or transmission of electromagnetic radiation in flexible waveguide(s)
US10912462B2 (en) 2014-07-25 2021-02-09 The General Hospital Corporation Apparatus, devices and methods for in vivo imaging and diagnosis
CN115541578A (en) * 2022-09-28 2022-12-30 佐健(上海)生物医疗科技有限公司 High-throughput super-resolution rapid scanning and analyzing system for cervical cell pathological section
CN115541578B (en) * 2022-09-28 2023-10-24 佐健(上海)生物医疗科技有限公司 High-flux super-resolution cervical cell pathological section rapid scanning analysis system

Also Published As

Publication number Publication date
AU6093400A (en) 2001-01-30
EP1203343A4 (en) 2006-08-23
EP1203343A1 (en) 2002-05-08
JP2003504627A (en) 2003-02-04

Similar Documents

Publication Publication Date Title
US6546123B1 (en) Automated detection of objects in a biological sample
WO2001004828A1 (en) Automated detection of objects in a biological sample
US7272252B2 (en) Automated system for combining bright field and fluorescent microscopy
US7428325B2 (en) Method and apparatus for automated image analysis of biological specimens
US7226788B2 (en) Automated slide staining apparatus
US8712118B2 (en) Automated measurement of concentration and/or amount in a biological sample
US20050037406A1 (en) Methods and apparatus for analysis of a biological specimen
US6631203B2 (en) Histological reconstruction and automated image analysis
WO2001057785A1 (en) Method and apparatus for automated image analysis of biological specimens
WO2001006446A1 (en) Automated method for image analysis of residual protein

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2000947299

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 2000947299

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 2000947299

Country of ref document: EP