ArticlePDF AvailableLiterature Review

State of the Art. A Structural and Functional Assessment of the Lung via Multidetector-Row Computed Tomography: Phenotyping Chronic Obstructive Pulmonary Disease

Authors:

Abstract and Figures

With advances in multidetector-row computed tomography (MDCT), it is now possible to image the lung in 10 s or less and accurately extract the lungs, lobes, and airway tree to the fifth- through seventh-generation bronchi and to regionally characterize lung density, texture, ventilation, and perfusion. These methods are now being used to phenotype the lung in health and disease and to gain insights into the etiology of pathologic processes. This article outlines the application of these methodologies with specific emphasis on chronic obstructive pulmonary disease. We demonstrate the use of our methods for assessing regional ventilation and perfusion and demonstrate early data that show, in a sheep model, a regionally intact hypoxic pulmonary vasoconstrictor (HPV) response with an apparent inhibition of HPV regionally in the presence of inflammation. We present the hypothesis that, in subjects with pulmonary emphysema, one major contributing factor leading to parenchymal destruction is the lack of a regional blunting of HPV when the regional hypoxia is related to regional inflammatory events (bronchiolitis or alveolar flooding). If maintaining adequate blood flow to inflamed lung regions is critical to the nondestructive resolution of inflammatory events, the pathologic condition whereby HPV is sustained in regions of inflammation would likely have its greatest effect in the lung apices where blood flow is already reduced in the upright body posture.
Content may be subject to copyright.
State of the Art
A Structural and Functional Assessment of the Lung via
Multidetector-Row Computed Tomography
Phenotyping Chronic Obstructive Pulmonary Disease
Eric A. Hoffman, Brett A. Simon, and Geoffrey McLennan
Departments of Radiology, Medicine, and Biomedical Engineering, University of Iowa, Iowa City, Iowa; and Department of Anesthesiology
and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland
With advances in multidetector-row computed tomography (MDCT),
it is now possible to image the lung in 10 s or less and accurately
extract the lungs, lobes, and airway tree to the fifth- through sev-
enth-generation bronchi and to regionally characterize lung den-
sity, texture, ventilation, and perfusion. These methods are now
being used to phenotype the lung in health and disease and to
gain insights into the etiology of pathologic processes. This article
outlines the application of these methodologies with specific em-
phasis on chronic obstructive pulmonary disease. We demonstrate
the use of our methods for assessing regional ventilation and perfu-
sion and demonstrate early data that show, in a sheep model, a
regionally intact hypoxic pulmonary vasoconstrictor (HPV) re-
sponse with an apparent inhibition of HPV regionally in the pres-
ence of inflammation. We present the hypothesis that, in subjects
with pulmonary emphysema, one major contributing factor leading
to parenchymal destruction is the lack of a regional blunting of
HPV when the regional hypoxia is related to regional inflammatory
events (bronchiolitis or alveolar flooding). If maintaining adequate
blood flow to inflamed lung regions is critical to the nondestructive
resolution of inflammatory events, the pathologic condition
whereby HPV is sustained in regions of inflammation would likely
have its greatest effect in the lung apices where blood flow is already
reduced in the upright body posture.
Keywords: airways; computed tomography; emphysema; inflammation;
functional imaging
The goal of this article reflects that of the mandate for guest speak-
ers at the Aspen Lung Conference: to provide a state-of-the-art
overview and to introduce a new and possibly controversial hypoth-
esis. The focus of the conference was chronic obstructive pulmonary
disease (COPD), and the goal of this article is to introduce the
breadth of tools now available through advanced multidetector-
rowcomputedtomography(MDCT)thatcanbeusedtogainnew
insights into lung pathologies associated with COPD.
The pathologic events leading to emphysema are insidious
and include structural and physiologic alterations that are char-
acterized by inflammatory processes within the peripheral pul-
monary parenchyma, thickening of arteriolar walls, and paren-
chymal destruction. A growing body of literature documents
that these changes are likely to be associated with alterations
in blood flow dynamics at a regional, microvascular level, and
(Received in original form March 20, 2006; accepted in final form May 30, 2006 )
Supported in part by NIH grants R01-HL-64368-01 and R01-HL-60158-04.
Correspondence and requests for reprints should be addressed to Eric A. Hoffman,
Ph.D., Department of Radiology, University of Iowa, 200 Hawkins Drive, CC701
GH, Iowa City, IA 52242. E-mail: eric-hoffman@uiowa.edu
Proc Am Thorac Soc Vol 3. pp 519–534, 2006
DOI: 10.1513/pats.200603-086MS
Internet address: www.atsjournals.org
thus may serve as a beacon pointing toward the onset of early
emphysema. Furthermore, recent findings in our laboratory have
led us to believe that regional alterations in blood flow parame-
ters may not only serve as an early marker for inflammatory
processes but may also be a major etiologic component of the
pathologic process, leading to emphysema in a subset of the
smoking population (not all smokers have emphysema). We
present preliminary evidence that leads us to a new hypothesis
relating emphysema to an inherent loss of the ability to blunt
regional hypoxic pulmonary vasoconstriction (HPV) when the
regional hypoxia is derived from inflammatory events.
MDCT provides the ability to image the lung with a theoreti-
cal in vivo resolution of approximately 0.5 mm. The whole lung
can be imaged at this resolution in approximately 10 seconds,
which is well within a single breath-hold. Scanner rotation speeds
are on the order of 300 milliseconds per revolution, and recently
there has been the introduction of dual-source CT (Somatom
Definition; Siemens Medical Systems, Erlangen, Germany)
whereby two X-ray guns are placed on the gantry, serving to
double the temporal resolution of the scanner system, and thus
opening up the possibility of dual energy scanning, which allows
for sensitive discrimination between tissue types and contrast
agents, such as iodine and xenon. Dynamic imaging via CT allows
for regional quantitative assessment of parenchymal perfusion
and ventilation. With advances in image-processing methods,
the lung, lobes, bronchial tree, and vascular trees can be ex-
tracted and quantitatively assessed. Density and texture mea-
sures of the lung parenchyma via MDCT imaging are now pro-
viding tools for establishing regional presence and distribution
of lung pathology, which, when coupled with regional measures
of function, may serve as important phenotypes within a popula-
tion, serving as the starting point for the quest to define associ-
ated genotypes.
From the unique opportunity to link structure and function
via MDCT, we have found evidence that, on a very regional
basis, the lung is able to shut off or blunt HPV in the presence
of local inflammation. This article puts forth the following series
of interrelated hypotheses as follows:
Smoking is associated with regional microinflammatory
events, which in turn cause regional hypoxia.
Smokers without the ability to shut off regional HPV in the
presence of inflammation are susceptible to emphysema.
If perfusion to the inflamed hypoxic regions is important
to the evolution of emphysema, then the HPV in regions
of inflammation will be compounded with reduced flow to
the apical lung due to gravitational effects.
Inflammatory parenchymal lung diseases are common and are
significant causes of disability and premature death. These diseases
520 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 3 2006
are the result of subacute/chronic or chronic inflammatory pro-
cesses, and are linked to cigarette smoking, either as a cause or
as a modifying agent. Current methods for the assessment of these
disorders include measures of lung function, radiologic techniques
such as CT scanning (1), radionuclear-based ventilation/perfu-
sion lung scans (2–5), use of hyperpolarized 3-He gas (2, 6–8)
in conjunction with magnetic resonance imaging (MRI), or direct
assessment of lung pathology. Much of the focus on mechanisms
of improvement has focused on lung mechanics (9–11). Remy-
Jardin and colleagues (1) have provided a unique observation
via CT demonstrating that longitudinal changes leading to an
emphysema-like lung begin with micronodular and ground-glass
appearances in the lung field correlating to bronchiolitis and
parenchymal inflammation. However, although many individu-
als with these regional inflammatory processes progressed to-
ward an increased emphysema burden (in the inflamed regions),
not all subjects with inflammation evolved toward emphysema,
suggesting that there may be important differences in the ways
individuals react to regional inflammatory processes in the lung.
Although respiratory textbooks teach that it is teleologically
advantageous to shunt blood away from regions of poor ventila-
tion, when the poor ventilation is caused by inflammatory
processes (i.e., micronodules and ground glass found by Remy-
Jardin and colleagues [1]), the best hope for resolution of the
inflammation is to maintain an adequate blood supply to that
area. In fact, three of the four components of the classic descrip-
tion of the inflammatory response—calor (warmth), rubor (red-
ness), and tumor (swelling)—reflect increased local blood flow
(dolor, or pain, being the fourth). Schuster and colleagues and
Gust and colleagues, in two key papers using positron emission
tomography (PET) imaging to study mechanisms of hypoxemia
in acute lung injury (12, 13), showed that HPV is shut off when
even a very small dose of endotoxin has been delivered to the
lung. Recent work in our laboratory using MDCT, and outlined
below, provides further evidence of local HPV inhibition in the
presence of regional inflammation concomitant with other areas
of the lung maintaining a healthy HPV response.
Recent conclusions from the National Emphysema Treatment
Trial (NETT) (14) show that the subgroup most likely to respond
to surgery with improvement in exercise and quality of life are
subjects with low exercise capabilities and apical predominance
of the disease (15). If a key feature of the disease is the shunting
of perfusion away from inflamed regions of lung, thus reducing
blood flow to regions needing perfusion to resolve the inflamma-
tion, then the normally reduced perfusion to the lung apices in
upright humans likely contributes significantly to susceptibility of
the parenchyma to adverse responses to inflammatory processes
in the confounding presence of unconstrained HPV.
Measures based on airflow or other measures of global lung
function have reached their limits in their ability to provide new
insights into the etiology of the disease, or even in leading us
to an understanding of how lung volume reduction, in late stages
of the disease, provides patient improvements. A number of
articles have been written in which attempts are made to explain
improvements of physiologic status post-LVRS (9, 16) on the
basis of lung mechanics, and we find it difficult to understand how
these relate to the observations from the NETT (15) showing that
subjects with apical but not basal prevalence of disease receive
the greatest benefit from surgery. However, if regional pulmo-
nary perfusion is again brought into consideration, it makes
sense that, if one removes apical lung that is not contributing
well to gas exchange and blood is shunted to less diseased basal
lung, gas exchange will be improved. Furthermore, by removing
a diseased portion of the basal lung when the disease is predomi-
nantly basal, then it is likely that blood will be preferentially
shunted to the contralateral basal lung. Using scintigraphy to
assess regional V
˙
/Q
˙
, Moonen and colleagues (2) have recently
concluded that an important mechanism for improvement in
functional status post-LVRS relates to the reduction of regional
shunt (i.e., blood flow may be directed toward regions of im-
proved ventilation whereas regions receiving blood flow but that
have poor ventilation are removed).
A recent international consensus statement on the diagnosis
and therapy of COPD—the Global Strategy for the Diagnosis,
Management, and Prevention of Chronic Obstructive Lung Dis-
ease (GOLD [Global Initiative for Chronic Obstructive Lung
Disease])—has established diagnostic criteria that currently
do not include CT findings (17). This is not surprising given that
the consensus statement has been developed in part for the
World Health Organization. It is notable that the summary
makes the observation that different inflammatory events occur
“in various parts of the lung,” a reference to the marked hetero-
geneity of the disease which cannot be defined without imaging.
Of interest also are the future recommended research directions,
which include identifying better defining characteristics of COPD,
developing other measures to assess and monitor COPD, and
recognizing the increasing need to identify earlier cases of the
disease, all potential outcomes of improvements to quantitative
lung imaging.
High-resolution volumetric MDCT with parenchymal structural
analysis, bolus contrast–based measurement of pulmonary perfu-
sion parameters, and xenon-enhanced measurement of regional
ventilation can provide objective and reproducible measures to
phenotypically describe the normal and the inflamed lung and
can provide important information regarding regional physiologic
status of the lung before and after interventional procedures such
as LVRS, endobronchial valve insertions to limit gas flow to
proximal lung, and grommet placements to relieve trapped gas.
PHENOTYPING EMPHYSEMA
Evaluation of Disease Progression
It has been well demonstrated that lung function declines with
age (18–21) and perhaps also as a result of inflammation (22).
This has confounded research related to the effects of smoking
cessation on lung health. There are mixed results as to whether
or not smoking cessation halts the progression of emphysema-
tous lung disease (20, 23–27). Work by Bosse and colleagues
(28) attempted to take into account the aging process and sug-
gested that the disease process is slowed if one stops smoking.
However, tests were not sensitive enough to conclude this defini-
tively. There appear to be important sex differences in the effects
of cigarette smoking and cessation (29). No reliable specific
biochemical markers of disease presence or progression have
been identified (30), in part perhaps because of the lack of a
sensitive standard to diagnose and follow the diseases. More
recently, CT parameters have been shown to be likely more
sensitive to disease progression (31). Furthermore, the long time
course of these diseases means that clinical trials using only
whole lung function as primary outcome measures require huge
numbers of subjects for extremely long periods of time.
Anatomic–Physiologic Correlates of Emphysema
The lack of a direct marker for emphysema has meant that
epidemiologic studies have been limited to COPD, and these
perhaps give a limited view as to the epidemiology of emphy-
sema, a specific subset of COPD but not identified as such by
spirometry. The direct effect of cigarette smoking on lung func-
tion has been widely studied, with differences in relative changes
in FEV
1
and the effects of smoking noted (28, 32–35). Although
some studies show an increased rate of loss of FEV
1
for current
smokers, there is a less significant decrease in FEV
1
for reformed
Hoffman, Simon, and McLennan: CT-Based Lung Structure and Function 521
smokers (28, 32). However, more recent studies have found similar
FEV
1
declines with age in both smokers and never-smokers (34,
35). It must be emphasized that these changes are likely related
to bronchial hyperreactivity (36, 37) rather than to emphysema,
highlighting again the need for objective measurement tools to
assess emphysema.
As indicated, chronic airflow limitation (COPD) is commonly
seen in emphysema, but it is not essential. Measurements of
lung physiology are not always able to distinguish the abnormali-
ties that result from emphysema from those which result from
the other causes of COPD, such as chronic bronchitis or asthma
(38). The strongest positive association between an index of
airflow limitation, FEV
1
(% predicted) and a pathologically
derived emphysema score comes from the National Institutes
of Health Intermittent Positive-pressure Breathing Trial (39).
There were only 48 subjects in this study, as autopsies were
required for the pathologic assessment to be performed. Pulmo-
nary function tests were performed every 3 months during the
study, and were therefore available at some point before death.
However, these subjects were highly selected; to enter the study,
they were required to have very significant airflow obstruction,
and could not be severely hypoxic; and to complete the study,
they had to die during the observation period. In contrast, a
study examining pathologic lung specimens taken during sur-
gery, and appropriately fixed, showed no relationship between
the pathologic emphysema rating and indices of airflow (40).
Furthermore, an autopsy study enrolling 242 subjects over 6
years demonstrated that, although those subjects with greater
pulmonary disability tended to have a greater degree of patho-
logic emphysema, 17 subjects with greater than 30% pathologic
emphysema had no evidence for clinical COPD (41). Other
pulmonary function tests—namely, diffusing capacity for carbon
monoxide (Dl
CO
) and the exponential description of the defla-
tion pressure/volume curve (K)—have been used to identify,
and to obtain a measure of, severity for pulmonary emphysema.
A number of studies have found that measurement of Dl
CO
has a very weak correlation with the pathologic assessment of
emphysema (42–44). Measurements of elastic recoil pressure
curves in life compared with pathologic assessment of emphy-
sema at subsequent lung resection or postmortem have yielded
conflicting results on the value of static compliance and K as a
measure of emphysema (45–49). More recent studies show a
weak but significant correlation between K and macroscopic
emphysema (r 0.49) (47, 48), with K believed to be a measure
of alveolar distensibility. This background highlights the continu-
ing search for a marker for emphysema presence and severity.
Vascular and Intravascular Alterations in Early Stages of
Parenchymal Lung Disease
Increased margination of the neutrophils in the small blood
vessels in the lung has been demonstrated in smokers (50–55).
One of the earliest abnormalities may therefore be regional
change in perfusion of affected lung regions. As the emphysema-
tous lesions develop in the peripheral lung, there is not only
destruction of the terminal air units but also gross destruction
of the microvasculature of the lung (55–57). These changes, in
the vascular bed and alveolar wall, may occur at the same time;
however, it is probable that the changes in vascular perfusion
occur slightly earlier than the alveolar wall changes, in keeping
with a primary role played by the blood neutrophils. In an elastase-
induced emphysema pig model, it has been shown that vascular
perfusion is reduced in early emphysematous lesions using single
photon emission computed tomography (SPECT) scanning (58).
In this model in which the elastase is injected into the lung,
there are initially areas of increased lung density, the result
of edema and alveolar hemorrhage. In a guinea pig model of
emphysema, muscularization of the pulmonary arterioles occurs
well before any evidence of emphysema, suggesting another
potential mechanism for alterations in blood flow as an early
feature in emphysema development (59). Hyde and colleagues
(60) have recently demonstrated that, in the adult rabbit, blood
flow is significantly diverted from inflamed lung. The presence
and magnitude of both pulmonary and bronchial blood flow
have been shown to effect the recovery of the lung from broncho-
constriction and the clearance of aerosol or noxious particles
(61–64). Thus, although there is the notion that blood flow alter-
ations serve as a tag of inflammation, the physiology remains
largely unknown, and its elucidation in the in vivo system will
provide important new information as well as a tool for further
investigations. As we suggest in the first part of this article,
pulmonary perfusion abnormalities may be a primary cause of
emphysema, rather than a secondary phenomenon.
THE SCANNERS
X-Ray CT
Volumetric physiologic imaging had its beginning in the mid-
1970s with the Dynamic Spatial Reconstructor (DSR), which
supported 14 X-ray guns on a continuous rotating gantry and
was purpose-built to provide dynamic volumetric images of the
heart and lungs (65). Much of the work establishing the accuracy
and precision of volumetric lung imaging was performed on the
DSR (65, 66). Commercial imaging technology lagged signifi-
cantly behind this early work. The electron beam CT (EBCT)
(67) emerged in the early to mid-1980s as another purpose-
built scanner that entered the commercial arena. EBCT had no
moving parts and swept an electron beam along parallel X-ray
targets to achieve improved scan speeds of up to 50 milliseconds
per slice pair and eight stacked slices in approximately 224 milli-
seconds. There have been rapid advances in speed and resolution
with the advent of MDCT (68). Its cone-beam spiral CT uses a
two-dimensional (2D) detector array, allowing larger scanning
range in shorter time with higher image resolution (69, 70). The
ability to acquire multiple image slices per rotation with rotation
speeds as short as 0.33 seconds allows for a significant reduction
in acquisition time. Faster scan times will significantly impact
functional imaging protocols where the rate of perfusion of a
contrast agent is measured over time or where gated imaging is
needed. We believe the future of lung assessment resides with
true dynamic low-dose volumetric CT scanners that image at
least one-third of the thorax with 0.5-mm isotropic voxels and
a full rotation scan aperture of 150 milliseconds and that have
superior contrast resolution for radiopaque gas and injected
contrast detection. The system will be likely coupled with a
low-Tesla MR scanner that will be used to complement the
information available from the CT image. Patients will be
scanned frequently by low-Tesla MR and less frequently over
time by use of the CT component. To this end, MDCT has
evolved to where typical scanners now acquire 64 slices in a
single rotation spanning up to 4 cm of the z-axis of the chest,
and manufacturers have shown prototypes supporting up to 256-
slice scanning (Toshiba, Tochigi, Japan) in a single rotation.
A dual X-ray source scanner (Definition; Siemens, Erlangen,
Germany) provides significant increases in speed and opens the
possibility of dual energy scanning. With dual energy scanning
(setting the two X-ray sources at different kV, such as 80 and
140 kV), there is a density shift in regions with interposed con-
trast agents, such as inhaled xenon gas or injected iodinated
contrast agents, without a concomitant shift in normal body
tissues. As such, one can subtract away the tissues from the
images derived from the two imaging chains while leaving behind
the xenon or iodine signal as an index of regional lung function.
522 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 3 2006
Customized compounds (71) can now be designed to selectively
target tissues based on particular characteristics of the tissue.
MRI
Over the past 10 years, there has been renewed interest in
applying MRI to the lung. Of particular interest has been the
use of hyperpolarized helium (HP 3-He) MRI. Methods include
the following: measures of peripheral airspace size (72–75),
visualization of ventilation distribution at high spatial resolution
(76–79), and assessment of gas flow patterns within the lobar
and segmental airways (80–82). Furthermore, the speed of depo-
larization of HP 3-He enables direct measurement of regional
partial pressure of oxygen and thus allows for an inference of
regional gas exchange (83–85). Critical to HP 3-He, and its more
recent counterpart, HP Xe, is the need to quantitate the resultant
images. We believe that MDCT may serve as a gold standard
against which these quantitative measures can be developed.
SCANNING PROTOCOLS
Within the Appendix, we provide the scanning protocols associ-
ated with our anatomic and functional imaging studies. These
protocols include the radiation estimations. Radiation dose re-
mains the limiting factor in determining the benefit of the tech-
nology relative to the risk when applied to a particular study.
Hoffman and coworkers have discussed radiation dose consider-
ations more fully elsewhere (86).
QUANTITATIVE IMAGE ANALYSIS
Critical to taking full advantage of MDCT (and MRI) is the
ability to objectively evaluate the information content of the
images. In the case of the lung, the starting point is reliable
detection of the lungs, lobes, airways, and blood vessels, followed
by an analysis of parenchymal density and texture, and finally
a regional quantification of regional ventilation and perfusion
parameters.
Over the past 25 to 30 years, quantitative imaging has been
the focus in our laboratory to study the lung, and has included
biplane fluoroscopy methods for estimating lung stress–strain
(pleural pressure) (87, 88) and X-ray CT methods for the purpose
of evaluating the normal physiology of the lung. CT has been
validated as a tool for assessing lung volume (89), regional air
content (90, 91), regional lung expansion (91–93), airway seg-
mentation (93–95), and vessel segmentation (96, 97). These stud-
ies were focused on the use of purpose-built scanner systems
(the DSR [98]) and the electron beam CT scanner (67). More
recently, with the emergence of MDCT scanners, a Bioengineer-
ing Research Partnership Grant from the NIH (HL-064368,
E.A.H. and G.M.) has served to bring together investigators
from multiple institutions from around the world to establish
MDCT as a comprehensive imaging modality to assess both
structure and function of the human lung, to establish the normal
range of airway and vascular structure, parenchymal density,
and texture together with regional characteristics of ventilation
and perfusion. Perfusion is assessed via dynamic imaging of first-
pass kinetics of a bolus injection of iodinated contrast agent and
ventilation via the wash-in and washout characteristics of stable,
radio-dense xenon gas. A cohort of normal human subjects are
being imaged over 4 decades of age range, both male and female,
across a broad spectrum of racial and ethnic backgrounds to
establish an atlas of the normal lung, against which an unknown
lung can be statistically compared for abnormality.
Respiratory Gating
To detect early pathology and small, incremental progression of
disease, one must take great care to appropriately calibrate the
scanner on a regular basis, taking into account the imaging char-
acteristics of the scanner and image reconstruction algorithms.
Perhaps even more important, however, is to also take great
care that the lung is imaged at standardized volumes, just as one
coaches a patient in the pulmonary function laboratory. To this
end, we have established a respiratory gating methodology that
allows us to accurately gate image acquisition to lung volume
in human subjects, using either a pneumotachograph, an induc-
tance plethysmograph (Respitrace; Research Instrumentation
Associates, Inc., Chesterland, OH), or turbine flowmeter signal.
With modified scanner software, one is able to reduce the scanner
pitch (table increment per 360 gantry rotation divided by beam
collimation) down to 0.1 for retrospective respiratory-gated
spiral imaging. Within our laboratory, we have built a fully inte-
grated software/hardware solution using the pneumotachometer
and inductance plethysmograph and are currently building a
second system based on the turbine for Xe imaging in humans.
We use software written in LabView (National Instruments,
Austin, TX) to record patient physiology (including airway pres-
sure, ECG, blood pressures, etc.) and then we are able to gate
the scanner on and off according to the physiologic parameters of
interest. Scanner manufacturers are currently providing simple
pneumatic belts for respiratory gating. Little work has currently
been done to verify the accuracy of these belts under various
conditions, such as shifts from abdominal to ribcage breathing
and prone versus supine scanning.
The segmentation of the lung, lobes, airway, and pulmonary
vascular bed is described together with methods for assessing
lung texture (parenchymal pathologies), perfusion, and ventila-
tion in the following sections.
Lung Segmentation
Automated segmentation of the lungs from a 3D set of CT
images is a crucial first step in the quantitative analysis of pulmo-
nary physiology or pathophysiology. With large 3D image vol-
umes becoming commonplace, routine manual segmentation to
identify regions of interest (ROIs) is too cumbersome and time-
consuming. In addition, manual analysis has significant interob-
server and intraobserver variability.
We have developed and validated a segmentation method to
accurately extract the lungs from CT images (99) (Figure 1). This
approach, which can be used automatically or semiautomatically,
relies on thresholding to obtain approximate initial lung masks.
These lung masks are refined using topologic analysis (e.g., to
delete cavities and small disconnected pieces) and specialized
processing to enforce anatomic constraints (e.g., using a graph
search to find the most likely location of the line separating the
left and right lung). Experimental studies using images acquired
from humans have shown our method to be very accurate:
computer-generated and manually defined lung areas (in pixels)
correlated very well in individual slices (r 0.99).
Lobe Segmentation
Zhang and colleagues (100) have developed a semiautomatic
method for identifying the fissures in CT images (Figure 1). This
method uses a combination of anatomic features and CT image
features to identify the fissures on 2D transverse slices. These
features are combined into a cost function that reflects the likeli-
hood that a pixel lays on the fissure. A graph search, which is
a heuristic cost-based search technique, is used to find a path
between the endpoints. Graph searching finds the minimum cost
path between the two endpoints, where the cost function defini-
tion reflects the problem of interest. The user must initialize the
process once for each fissure of interest, but once the procedure
has been initialized, the entire 3D surface can be automatically
identified. The overall root mean square error between manual
Hoffman, Simon, and McLennan: CT-Based Lung Structure and Function 523
Figure 1. Results of vascular
(upper left), lobe (middle), and
airway (lower right) segmenta-
tion. After the airways are
identified (segmented) and the
centerline and branchpoints
are identified, then the airway
tree is automatically labeled.
The lobar fissures are identified
by the geometry of the seg-
mented blood vessels as shown
by the red and green arrow-
heads shown in the upper left
panel. L left, R right, B
broncus, UL upper lobe, M
middle lobe, LL lower lobe.
tracing of the fissure and our semiautomatic method is about
2 pixels. Under development are methods to automatically initi-
ate the lobe segmentation process through the development of
a standard lung atlas representing the average shape of the
normal human lung. The individual is then matched to the atlas
and the location of the fissures in the atlas serve as the initial
guess for the search initiation. More recent work from the labora-
tory has used an anatomic pulmonary atlas with a priori knowl-
edge about lobar fissure shapes from a set of presegmented
training datasets to achieve a fully automatic lobe segmentation
(101).
Airway Lumen and Wall Segmentation
Airways of interest range in size from 1- to 15-mm inside diame-
ter, and the software determines the borders of the inner and
outer airway walls (102). The small airways have very thin walls,
typically on the order of 10 to 15% of the inner diameter. The
established full-width at half-maximum method for measure-
ment can give very inaccurate results for these small thin-walled
structures. To address this problem, we use a new method of
estimating the airway wall locations. We first assess the point
spread function of the particular scanner/slice selection/recon-
struction algorithm of interest and then use a model-based de-
convolution to account for blur introduced in the scanning pro-
cess. This approach was shown to be more accurate than
previously used wall detection methods, especially for thin-
walled structures. Phantom studies have demonstrated the new
method to be applicable across a wide variety of airway sizes
(102, 103). As shown in Figure 2, once the airway tree has
been identified in 3D, airway paths can be “straightened” into
a pathway “pipe” view to allow for assessment of the local geo-
metry perpendicular to the airway centerline.
To identify the airway tree structure, Tschirren and colleagues
(104, 105) have developed an automated segmentation, skele-
tonization, and branchpoint matching method. The airway tree
is identified using a seeded region growing algorithm, starting
from an automatically identified seed point within the trachea.
The algorithm is designed so that it can overcome subtle gray-
level changes (e.g., those caused by beam hardening). On the
other hand, a “leaking” into the surrounding lung tissue can be
avoided. The implementation of the algorithm uses graph algo-
rithms that make it fast and memory friendly. The method reli-
ably segments the first five to six airway generations. The binary
airway tree is then skeletonized to identify the 3D centerlines
524 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 3 2006
Figure 2. Once the
three-dimensional air-
way tree has been iden-
tified and labeled, paths
can be identified and
straightened so as to
provide luminal and
wall dimensions mea-
sured as a function of
the distance along the
path and perpendicular
to the local long axis.
The airway cross-section
corresponding to the
yellow vertical line in the
upper panel is shown in
the lower left panel. The
green arrow in the lower
left panel can be rotated
about the centerline of
the airway to alter the
cut plane shown in the
straightened airway pre-
sented in the upper panel.
of individual branches and to determine the branchpoint loca-
tions. A sequential 3D thinning algorithm reported by Palagyi
and colleagues (106) was customized for our application. False
branches are pruned, and the resulting skeleton is guaranteed
to lie in the middle of the cylindrically shaped airway segments.
Branchpoints are used to define airway tree segments, which
are then automatically labeled with a modified standardized
nomenclature that we have established that takes into account
the most common variability between individuals. This nomen-
clature (shown in Figure 1) can be applied to images of multiple
lung volumes of the same individual to allow us to track the
change in airway dimensions along an airway path as well as the
change in airway dimensions with change in lung volume. Our
airway segmentation methods have been shown to be robust
in the presence of significant emphysema and when applied to
images acquired using low-dose scanning protocols. In Figure 3,
we demonstrate the ability to extract an airway tree of a subject
with interstitial lung disease in which there is considerable mixed
pathology, including emphysema, honeycombing, traction bron-
chiectasis, and fibrosis.
Parenchymal Analysis
Computer-based methods for objective quantitation of MDCT
datasets to compare normal and diseased lung parenchyma are
increasingly being used in conjunction with 2D datasets. A cor-
nerstone of lung assessment for emphysema by MDCT scanning
has become known as the density mask. The basis of the density
mask is that a CT scanner, if properly calibrated, reconstructs
air with a Hounsfield unit (HU standardized unit of X-ray
attenuation) of 1,000, water as 0, and blood/tissue as approxi-
mately 55. Because the lung is composed of only air or blood/
tissue densities and because the HU is linear between these two
values, one is able to assess the percentage of air and percentage
of blood/tissue in each reconstructed voxel. Because emphysema
is defined as an enlargement of the peripheral airspaces associ-
ated with parenchymal destruction, the HU of a voxel becomes
an index of presence and severity of the disease. By empirically
defining a given lung density at full inspiration as emphysema,
one can set a density threshold (HU) below which all voxels are
considered to be emphysema (107–114). This is the so-called
density mask. It has been observed that the density mask for
severe emphysema in 5-mm-thin or thinner slices falls at approxi-
mately 950 HU, moderate emphysema at approximately 910
HU, and mild at approximately 850 HU. By identifying where
the lung is in the image and then dividing the lung into left and
right, apical, mid, and basal regions, and then dividing these
regions into the “core” and “peel,” we are able to begin to es-
tablish phenotypes for populations (distinguished, for instance,
by sex, ethnicity,
1
-antitrypsin deficiency, and now possibly sub-
populations of smokers), differentiating populations based on
Hoffman, Simon, and McLennan: CT-Based Lung Structure and Function 525
Figure 3. Demonstration of the ability to
extract a detailed airway tree from com-
puted tomography (CT) scans of a patient
with significant mixed-lung pathology.
The images show emphysema, honey-
combing, traction bronchiectasis, and
fibrosis.
characteristics of the pattern and severity of emphysema. Adams
and coworkers have pointed out the importance of imaging with-
out contrast agent when using HU as a measure of emphysema
(107). A density-masking approach alone is not sufficient to
accurately distinguish normal from diseased lung (115–117), es-
pecially in the case of early or mixed pathologic processes. The
density mask is, however, particularly useful in characterizing
mild/moderate and severe emphysema and has been used in the
NETT to identify subgroups of patients who show benefit from
LVRS (15). With the increased use of CT to screen for lung
cancer (118) and coronary calcium (119), Reddy and colleagues
have demonstrated the utility of using these same scans to char-
acterize the presence and distribution of emphysema (120, 121).
Care must be taken when one uses CT to quantitate parenchymal
characteristics because scanner miscalibration and reconstruc-
tion kernels can cause some variations in the measurements
(122–124). Furthermore, because the X-ray is not a single energy,
beam-hardening artifacts, if not well corrected for by the manu-
facturers, can cause additional errors.
As our image analysis methods have evolved, we have inte-
grated the tools into a PC-based comprehensive lung image
analysis package called Pulmonary Analysis Software Suite
(PASS). In addition to the traditional density mask discussed
above, we have incorporated an additional measure, one pro-
posed by Mishima and colleagues (125), which has been termed
the “fractal dimensions” or “alpha.” Alpha is the slope of the
log–log relationship of hole size versus percentage of holes at
that size. The notion is that initially a random set of holes evolves
in lung regions and, as such, the log–log plot of hole size versus
percentage of holes is linear. However, once the initial holes
have evolved, there is a greater likelihood that these holes will
have destabilized the lung mechanically and the small holes will
combine to form bigger holes as opposed to more small holes
appearing. Thus, the slope of the log–log plot diminishes. In this
way, alpha becomes an index of disease severity.
Texture (Adaptive Multiple-Feature Method)
High-resolution CT (HRCT) enhances the resolving power of
the image (126–130), allowing detection of less severe emphy-
sema. Various computer-assisted texture-based methods have
successfully been used for tissue characterization. Traditional
methods of texture analysis can be grouped into statistical, struc-
tural, and hybrid methods (131). Methods for tissue classification
typically rely on region gray-scale statistical measures (i.e., mean,
variance, frequency histogram) or textural measures (autocorre-
lation, co-occurrence matrices, run-length matrices, etc.) (107–
111, 132–141).
Although simple density measures are adequate for the as-
sessment of moderate to severe emphysema, this simple measure
is inadequate in assessing early pathologic changes, detecting
changes where the pathology is mixed, or detecting more com-
plex patterns such as ground glass. We have developed and
patented a unique method of texture analysis of the lung for the
objective assessment of pathologic processes in which simple
lung density measures are inadequate for detection or differenti-
ation of processes. Our Adaptive Multiple-Feature Method
(AMFM) assesses as many as 22 independent textural features
from HRCT scans to classify a tissue pattern (116, 142, 143).
The AMFM is 100% reproducible and performs as well as experi-
enced human observers who have been told the patient diagnosis.
Recently, a goal of extending the AMFM feature set from 2D
to 3D (144) has been motivated by the emergence of MDCT
scanners with the ability to acquire volumetric image datasets
526 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 3 2006
with near isotropic (near equal dimensions in all three orthogo-
nal directions) voxels. We have used images from the normal
population studied through our Bioengineering Research Part-
nership (BRP) grant together with heavy smokers studied at the
University of Iowa in the National Lung Screening Trial. The
3D ROIs from lung regions (50 subjects total) of normal never-
smokers were identified together with normal lung from normal
smokers, mild emphysema from smokers with mild COPD, and
moderate to severe emphysema regions from smokers with
moderate to severe COPD. Lung status was defined based on
American Thoracic Society GOLD criteria for characterizing
COPD derived from spirometry-based FEV
1
/FVC measures. Al-
though 2D performed as well as 3D for GOLD 2 and GOLD
3 emphysema, the 3D feature set provides a highly significant
improvement in differentiating normal-appearing lung sampled
from nonsmokers versus GOLD class 0 smokers.
Functional Imaging
Numerous methods have been developed to assess ventilation
and perfusion, or their functional outcome, gas exchange. Al-
though clearly useful, pulmonary function tests are global mea-
surements of airflow, lung volumes, and gas exchange from which
are inferred primary structural and functional alterations. Im-
aging techniques such as PET (145) and the newly emerging
hyperpolarized gas imaging via MRI (146–152) offer unique,
complementary regional information to X-ray CT and, as they
develop, are expected to offer enhancements to the knowledge
base that we propose to build into the lung atlas via MDCT.
Ventilation assessed by CT. Regional ventilation is measured
from time course of CT density change during a multibreath
wash-in and washout of radio-dense Xe gas (153). Studies to
date have demonstrated that the optimal imaging time is at end
expiration when conducting airways are filled with alveolar gas
(154). Average regional time constants are similar for repeat
runs reducing inspired Xe gas concentrations from 55 to 30%,
but the coefficient of variation at 30% Xe is significantly greater
than at 40% and higher concentrations. The addition of 30%
krypton gas to 30% Xe gas provides the same contrast enhance-
ment and signal-to-noise ratio as 40% Xe (155). Krypton has
none of the unwanted side effects of higher concentrations of
Xe gas. Of particular note is the observation that wash-in and
washout time constants are not equal, as previously assumed.
Washout is longer, specifically at higher Xe concentrations and
in dependent basal lung regions (154).
Perfusion assessed by CT. Dynamic imaging methods have
been used to estimate arterial, venous, and capillary transit times
and capillary flow distributions (156–163). These methods in-
volve two types of image data collection regimes: inlet–outlet
detection is typically used for conducting vessels and whole
organ analysis; the other data collection regime is referred to
as residue detection. Residue detection is typically used alone or
in conjunction with inlet detection for analysis of microvascular
regions wherein the individual vessels are below the resolution
of the imaging system. Various approaches for determining
blood flow and/or mean transit time have been described (157,
160–170).
To assess regional parenchymal perfusion, we place a catheter
in the right ventricular outflow tract in animals and in the supe-
rior vena cava in humans. A sharp (0.5 cc/kg over 2 s) bolus of
iodinated contrast agent (Visipaque; GE Healthcare, Milwau-
kee, WI) is delivered during ECG gated axial scanning. Scanning
commences one to two heartbeats before contrast injection, with
lungs held at functional residual capacity. By sampling the recon-
structed time-attenuation curves within the region of a pulmo-
nary artery and the lung parenchyma, we are able to calculate
regional mean transit times as well as blood flow normalized to
air or tissue content (171). We are able to deconvolve the signals
such that we can estimate the timing of flow within the microvas-
cular bed (162, 172).
We have begun imaging normal human subjects to establish
the image-based atlas of blood flow in the normal human lung.
As part of our work to determine the differences between the
normal lung and the lung of smokers, we have imaged a series
of never-smokers and smokers, both falling within a GOLD
category 0. As shown in Figure 4, in preliminary studies we have
found that smokers, even if defined as normal by pulmonary
function tests, have increased heterogeneity (coefficient of varia-
tion) of local mean transit times of the contrast agent. With
voxels on the order of 0.4 0.4 0.4 mm, the increased coeffi-
cient of variation shows up only when sampling of regional blood
flow occurs in regions no larger than 3 3 voxels, indicating
that the level of early disruption of blood flow is at the level of
the microvasculature.
HPV AND ITS FAILURE IN THE PRESENCE OF
INFLAMMATION
By combining the ability to assess regional lung density, ventila-
tion, and perfusion, we now provide the initial evidence for our
hypothesis introduced at the beginning of this article: in subjects
with pulmonary emphysema, one major contributing factor lead-
ing to parenchymal destruction is the lack of a regional blunting
of HPV when the regional hypoxia is related to regional inflam-
matory events (bronchiolitis or alveolar flooding).
One of the fundamental homeostatic mechanisms by which
the lung preserves oxygenation in the face of injury is HPV.
HPV causes pulmonary arterial blood vessels to constrict in
response to local hypoxia, thus redistributing blood flow away
from poorly ventilated regions and toward lung regions that are
well ventilated. This response optimizes local V
˙
/Q
˙
matching and
minimizes shunt as a mechanism of hypoxemia.
Both ventilation and perfusion were measured in units of
ml/min in sheep being evaluated under a protocol, approved by
the University of Iowa Animal Care and Use Committee, to
develop imaging methods to evaluate the functional status of
the lung after placement of endobronchial valves used in humans
as an alternative to LVRS.
Studies reported here were performed at the Iowa Compre-
hensive Lung Imaging Center using a Siemens Sensation 64
MDCT scanner, modified to allow for external scan gating to the
respiratory signal, and anesthetized, supine sheep were studied.
Figure 5 shows data from one sheep with the original gray-scale
images shown in the left column, the ventilation coded image in
the middle column, and the perfusion image shown in the right
column. Images taken before valve placement are in the upper
row and post–valve placement data are shown in the lower row.
Of interest are the following: (1) the sheep arrived in the labora-
tory with pneumonia, (2) loss of ventilation post–valve place-
ment is evident in the lower middle panel,(3 ) reflex loss of blood
flow in response to the loss of ventilation due to the valve is
evident in the lower right panel, and (4 ) perfusion shunted away
from the region of the valve served to preferentially enhance
perfusion in the unventilated region in the dependent lung where
there was evidence of pneumonia. Other data in the laboratory
have shown that when a sheep is exposed to inspired hypoxia
(15% O
2
), perfusion is reduced everywhere except in the region
of pneumonia, and in the region of pneumonia, perfusion is
enhanced.
These studies reflect earlier findings of Gust and colleagues
and Schuster and colleagues (13, 173) in which endotoxin was
shown to block HPV when assessing regional perfusion via use
of PET. These data, coupled with the findings of perfusion
Hoffman, Simon, and McLennan: CT-Based Lung Structure and Function 527
Figu r e 4. Anormalnon-
smoker and GOLD 0
sm oke r showi n g consi d-
erable increase in paren-
chymal perfusion hetero-
geneity. When sampling
at increasingly coarse
sample sizes, the differ-
ence in the coefficients of
variation (COV) between
the two groups disap-
pears when the region-
of-interest size is larger
that 3 3voxels( 1.2
mm on a side , about one-
fifth to one-tenth the size
of an adult ascinus) (174).
Color coding ranges
from 0 (blue)to15(red)
ml/min.
disruption in smokers shown in Figure 4, indicating likely perfusion
disturbance in response to inflammation, have led us to hypothesize
that the normal response to hypoxia caused by inflammatory pro-
cesses is to block HPV, by a process not here stipulated, so as to
ensure that the cascade of events serving to fight inflammation and
Figure 5. Before (base-
line; top row) and after
(post valve; bottom row)
endobronchial valve
placement. The left col-
umn provides a view of
one CT section at the
level of the diaphragm
dome. Note the signifi-
cant dependent pneu-
monia (A ). The center
column demonstrates
ventilation overlayed
in color as assessed by
xenon-CT. Note that
there is little ventilation
in either the baseline or
the post valve in the re-
gion of the dependent
pneumonia. By design,
there is a large region
where ventilation was
eliminated by the valve
placement (B ). The right
column shows a color
overlay of the perfusion
measurements. Note
that in the region (C )
coinciding with region B
from the central column
(no ventilation due to
valve placement), there
is a regional loss of perfusion, indicating an intact hypoxic pulmonary vasoconstrictor (HPV) response. At the same time, in the region of pneumonia
(D ) where there is little or no ventilation, blood flow is enhanced after valve placement. Presumably, blood flow shunted away from the valve-
based HPV is shunted straight toward the region of inflammation, presumably because the inflammation has served to blunt HPV in this region,
leaving this region as the path of least resistance for the blood flow diverted from the region effected by HPV. Thus, this image demonstrates that
lung has the ability to locally modulate HPV based on local inflammation.
infection can manifest itself. We further hypothesize that, in a
subset of smokers, there is an inability to block HPV in the face
of inflammation and this failure sets the individual up for the
evolution of the tissue destruction, which is a hallmark of emphy-
sema. Indeed, Remy-Jardin and colleagues (1) have demonstrated,
528 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 3 2006
as discussed earlier, in a small cohort of subjects, that, in a subset
of smoker who show decline over time in pulmonary function tests,
this decline is correlated with the development of emphysema in
regions showing signs of ground glass and micronodules at an
earlier time point. Not all smokers with ground glass and mi-
cronodules developed emphysema in these regions but these
subjects also did not show a decline in pulmonary function tests.
CONCLUSIONS
We have demonstrated that MDCT provides for a comprehen-
sive means of imaging the lung. It provides for a sensitive and
objective method of assessing the lung parenchyma, airway, and
functional status at the lung periphery, including measures of
both ventilation and perfusion. With a combination of highly
detailed anatomic information together with function measures,
we are able to evaluate regional pathologic processes for the
refined assessment of COPD phenotypes. We note that with the
growing set of powerful tools available in the clinical setting,
it becomes important to carefully establish new questions and
apply appropriate imaging modalities. One must take care to
apply these modalities appropriately, which brings a critical need
for interdisciplinary interactions among pulmonologists, physiol-
ogists, radiologists, physicists, engineers, and many more.
Conflict of Interest Statement : E.A.H. is the founder and co-owner of VIDA Diag-
nostics, which is commercializing some of the software that has evolved out of
his research group. He is also a member of the CT Medical Advisory Board for
Siemens Medical Solutions. B.A.S. does not have a financial relationship with a
commercial entity that has an interest in the subject of this manuscript. G.M. is
co-owner of VIDA Diagnostics, which is commercializing some software develop-
ments coming out of his laboratory. In addition, he is a clinical investigator for
Asthmatx and Emphasys.
Acknowledgment : The authors thank the entire research team of their Bioengin-
eering Research Partnership grant whose combined efforts have made possible
the findings reported within this paper. Of particular note are the contributions of
Drs. Joseph Reinhardt and Milan Sonka and Juerg Tschirren, who made significant
contributions to the image processing, and Drs. Deokiee Chon, Osama Saba, and
David Riker, who have made significant contributions to the experimental side of
the studies reported here. Abby Russi helped in the preparation of this manuscript.
References
1. Remy-Jardin M, Edme JL, Boulenguez C, Remy J, Mastora I, Sobaszek
A. Longitudinal follow-up study of smoker’s lung with thin-section
CT in correlation with pulmonary function tests. Radiology 2002;222:
261–270.
2. Moonen M, Xu J, Johansson A, Thylen A, Bake B. Effects of lung
volume reduction surgery on distribution of ventilation and perfusion.
Clin Physiol Funct Imaging 2005;25:152–157.
3. Xu JH, Moonen M, Johansson A, Bake B. Distribution of ventilation-
to-perfusion ratios analysed in planar scintigrams of emphysematous
patients. Clin Physiol 2000;20:89–94.
4. Cederlund K, Hogberg S, Jorfeldt L, Larsen F, Norman M, Rasmussen
E, Tylen U. Lung perfusion scintigraphy prior to lung volume reduc-
tion surgery. Acta Radiol 2003;44:246–251.
5. Hunsaker AR, Ingenito EP, Reilly JJ, Costello P. Lung volume reduc-
tion surgery for emphysema: correlation of CT and V
˙
/Q
˙
imaging with
physiologic mechanisms of improvement in lung function. Radiology
2002;222:491–498.
6. Spector ZZ, Emami K, Fischer MC, Zhu J, Ishii M, Vahdat V, Yu J,
Kadlecek S, Driehuys B, Lipson DA, et al. Quantitative assessment of
emphysema using hyperpolarized 3He magnetic resonance imaging.
Magn Reson Med 2005;53:1341–1346.
7. Dupuich D, Berthezene Y, Clouet PL, Stupar V, Canet E, Cremillieux Y.
Dynamic 3He imaging for quantification of regional lung ventilation
parameters. Magn Reson Med 2003;50:777–783.
8. van Beek E, Wild J, Schreiber W, Kauczor H, Mugler JP III, de Lange
EE. Functional MRI of the lungs using hyperpolarized 3-helium gas.
J Magn Reson Imaging 2004;20:540–554.
9. Fessler HE, Scharf SM, Permutt S. Improvement in spirometry following
lung volume reduction surgery: application of a physiologic model.
Am J Respir Crit Care Med 2002;165:34–40.
10. Criner G, Cordova FC, Leyenson V, Roy B, Travaline J, Sudarshan S,
O’Brien G, Kuzma AM, Furukawa S. Effect of lung volume reduction
surgery on diaphragm strength. Am J Respir Crit Care Med 1998;157:
1578–1585.
11. Lewis TA, Tzeng YS, McKinstry EL, Tooker AC, Hong K, Sun Y,
Mansour J, Handler Z, Albert MS. Quantification of airway diameters
and 3D airway tree rendering from dynamic hyperpolarized 3He
magnetic resonance imaging. Magn Reson Med 2005;53:474–478.
12. Schuster DP, Marklin GF. Effect of changes in inflation and blood
volume on regional lung density: a PET study: 2. J Comput Assist
Tomogr 1986;10:730–735.
13. Gust R, Kozlowski J, Stephenson AH, Schuster DP. Synergistic hemody-
namic effects of low-dose endotoxin and acute lung injury. Am J
Respir Crit Care Med 1998;157:1919–1926.
14. National Emphysema Treatment Trial Research Group. Rationale and
design of the National Emphysema Treatment Trial (NETT): a pro-
spective randomized trial of lung volume reduction surgery. J Thorac
Cardiovasc Surg 1999;118:518–528.
15. Fishman A, Martinez F, Naunheim K, Piantadosi S, Wise R, Ries A,
Weinmann G, Wood DE; National Emphysema Treatment Trial Re-
search Group. A randomized trial comparing lung-volume-reduction
surgery with medical therapy for severe emphysema. N Engl J Med
2003;348:2059–2073.
16. Fessler HE, Permutt S. Lung volume reduction surgery and airflow
limitation. Am J Respir Crit Care Med 1998;157:715–722.
17. Pauwels RA, Buist AS, Calverley PM, Jenkins CR, Hurd SS. Global
strategy for the diagnosis, management, and prevention of chronic
obstructive pulmonary disease. NHLBI/WHO Global Initiative for
Chronic Obstructive Lung Disease (GOLD) workshop summary. Am
J Respir Crit Care Med 2001;163:1256–1276.
18. Knudson RJ, Clark DF, Kennedy TC, Knudson DE. Effect of aging
alone on mechanical properties of the normal adult lung. J Appl
Physiol 1977;43:1054–1062.
19. Muieson G, Sorbini CA, Grassi V. Respiratory function in the aged.
Bull Physiopathol Respir (Nancy) 1971;7:973–1009.
20. Fletcher C, Petro R. The natural history of chronic airflow obstruction.
Br Med J 1977;1:1645–1648.
21. Bosse R, Sparrow D, Garvey AJ, Costa PT, Weiss ST, Rowe JW.
Cigarette smoking, aging, and decline in pulmonary function: a longi-
tudinal study. Arch Environ Health 1980;35:247–252.
22. Meyer KC, Ershler W, Rosenthal NS, Lu XG, Peterson K. Immune
dysregulation in the aging human lung. Am J Respir Crit Care Med
1996;153:1072–1079.
23. Buist AS, Nagy JM, Sexton GJ. Effect of smoking cessation on pulmo-
nary function: a 30-month follow-up to two smoking cessation clinics.
Am Rev Respir Dis 1979;120:953–957.
24. McCarthy DS, Craig DB, Cherniack RM. The effect of modification of
smoking habit on lung function. Am Rev Respir Dis 1976;114:103–113.
25. Comstock GW, Brownlow WJ, Stone RW, Sartwell PE. Cigarette smok-
ing and changes in respiratory findings. Arch Environ Health 1970;21:
50–57.
26. Bake B, Oxhoj H, Sixt R, Wilhelmsen L. Ventilatory lung function
following two years of tobacco abstinence. Scand J Respir Dis 1977;
58:311–328.
27. Wilhelmsen L, Orha I, Tibblin G. Decrease in ventilatory capacity
between ages of 50 and 54 in representative sample of Swedish men.
BMJ 1969;3:553–556.
28. Bosse R, Sparrow D, Rose CL, Weiss ST. Longitudinal effect of age
and smoking cessation on pulmonary function. Am Rev Respir Dis
1981;123:378–381.
29. Xu X, Weiss ST, Rijcken B, Schouten JP. Smoking, changes in smoking
habits, and rate of decline in FEV1: new insight into gender differ-
ences. Eur Respir J 1994;7:1056–1061.
30. Dillon TJ, Walsh RL, Scicchitano R, Eckert B, Cleary EG, McLennan
G. Plasma elastin-derived peptide levels in normal adults, children,
and emphysematous subjects. Physiologic and computed tomographic
scan correlates. Am Rev Respir Dis 1992;146:1143–1148.
31. Dirksen A, Dijkman JH, Madsen F, Stoel B, Hutchison DC, Ulrik CS,
Skovgaard LT, Kok-Jensen A, Rudolphus A, Seersholm N, et al. A
randomized clinical trial of (1)-antitrypsin augmentation therapy.
Am J Respir Crit Care Med 1999;160:1468–1472.
32. Beaty TH, Menkes HA, Cohen BH, Newill CA. Risk factors associated
with longitudinal change in pulmonary function. Am Rev Respir Dis
1984;129:660–667.
33. Camilli AE, Burrows B, Knudson RJ, Lyle SK, Lebowitz MD. Longitudinal
changes in forced expiratory volume in one second in adults: effects
Hoffman, Simon, and McLennan: CT-Based Lung Structure and Function 529
of smoking and smoking cessation. Am Rev Respir Dis 1987;
135:794–799.
34. Tager IB, Segal MR, Speizer FE, Weiss ST. The natural history of
forced expiratory volumes. Am Rev Respir Dis 1988;138:837–849.
35. Knudson RJ, Knudson DE, Kattenborn WT, Bloom JW. Subclinical
effects of cigarette smoking: a five year follow up of physiological
comparisons of healthy middle aged smokers and non-smokers. Chest
1989;95:512–518.
36. Sherman CB. The health consequences of cigarette smoking: pulmonary
diseases. Med Clin North Am 1992;76:355–375.
37. Vollmer WM, Johnson LR, Buist AS. Relationship of response to a
bronchodilator and decline in forced expiratory volume in one second
in population studies. Am Rev Respir Dis 1985;132:1186–1193.
38. Macklem PT. The pathophysiology of chronic bronchitis and emphy-
sema. Med Clin North Am 1973;57:669–679.
39. Campbell DA, McLennan G, Coates JR, Frith PA, Gluyas PA, Latimer
KM, Luke CG, Martin AJ, Roder DM, Ruffin RE, et al. A comparison
of asthma deaths and near-fatal asthma attacks in South Australia.
Eur Respir J 1994;7:490–497.
40. Martin AJ, Campbell DA, Gluyas PA, Coates JR, Ruffin RE, Roder
DM, Latimer KM, Luke CG, Frith PA, Yellowlees PM, et al. Charac-
teristics of near-fatal asthma in childhood. Pediatr Pulmonol 1995;20:
1–8.
41. Campbell DA, Yellowlees PM, McLennan G, Coates JR, Frith PA,
Gluyas PA, Latimer KM, Luke CG, Martin AJ, Ruffin RE. Psychiatric
and medical features of near fatal asthma. Thorax 1995;50:254–259.
42. West WW, Nagai A, Hodgkin JE, Thurlbeck WM. The National Insti-
tutes of Health Intermittent Positive Pressure Breathing trial: pathol-
ogy studies. III. The diagnosis of emphysema. Am Rev Respir Dis
1987;135:123–129.
43. Morrison NJ, Abboud RJ, Ramadan F, Miller RR, Gibson NN, Evans
KG, Nelems B, Muller NL. Comparison of single breath carbon
monoxide diffusing and pressure-volume curves in detecting emphy-
sema. Am Rev Respir Dis 1989;139:1179–1187.
44. Klein JS, Gamsu G, Webb WR, Golden J, Muller NL. High-resolution
CT diagnosis of emphysema in symptomatic patients with normal
chest radiographs and isolated low diffusing capacity. Radiology 1992;
182:817–821.
45. Boushy SF, Aboumrad MH, North LB, Helgason AH. Lung recoil
pressure, airway resistance, and forced flows related to morphologic
emphysema. Am Rev Respir Dis 1971;104:551–561.
46.GelbAF,GoldWM.WrightRr,BruchMR,NakelJA.Physiologic
diagnosis of subclinical emphysema. Am Rev Respir Dis 1973;107:50–63.
47. Berend N, Skoog C, Thurlbeck WM. Pressure-volume characteristics
of excised human lungs: effects of sex, age and emphysema. J Appl
Physiol 1980;49:558–565.
48. Pare PD, Brooks LA, Bates J, Lawson LM, Nelems JM, Wright JL,
Hogg JC. Exponential analysis of the lung pressure-volume curve as
a predictor of pulmonary emphysema. Am Rev Respir Dis 1982;126:
54–61.
49. Bergin C, Muller N, Nichols DM, Lillington G, Hogg JC, Mullen B,
Grymaloski MR, Osbourne S, Pare PD. The diagnosis of emphysema:
a computed tomography–pathologic correlation. Am Rev Respir Dis
1986;133:541–546.
50. Hubbard RC, Brantly ML, Crystal RG. Proteases. In: Crystal RG, West
JB, editors. The lung: scientific foundations. New York: Raven Press;
1991. pp. 1763–1773.
51. Taylor RG, Gross E, Joyce H, Holland F, Pride NB. Smoking, allergy,
and the differential white blood cell count. Thorax 1985;40:17–22.
52. Hunninghake G, Gadek J, Kawanami O, Ferrans V, Crystal R. Inflamma-
tory and immune processes in the human lung in health and disease:
evaluation by bronchoalveolar lavage. Am J Pat hol 1979;97:149–206.
53. Niewoehner DE. Cigarette smoking, lung inflammation, and the devel-
opment of emphysema. J Lab Clin Med 1988;111:15–27.
54. Cohen AB, Rossi M. Neutrophils in normal lungs. Am Rev Respir Dis
1983;127:S3–S9.
55. Malech HL, Gallin JI. Current concepts: immunology neutrophils in
human diseases. N Engl J Med 1987;317:687–694.
56. Janoff A. Biochemical links between cigarette smoking and emphysema.
J Appl Physiol 1983;55:285–293.
57. Murphy KR, Marsh WR, Glezen LS, Irvin CG, Wilson MC, Larsen
GL. Inflammation and the late phase reaction in asthma: the effect
of polymorphonuclear leukocyte depletion on airways obstruction
and bronchial hyperreactivity in an animal model. Bull Eur Physiopa-
thol Respir 1986;22:48–53.
58. Noma S, Moskowitz GW, Herman PG, Khan A, Rojas KA. Pulmonary
scintigraphy in elastase-induced emphysema in pigs: correlation with
high-resolution computed tomography and histology. Invest Radiol
1992;27:429–435.
59. Wright JL, Churg A. Effect of long-term cigarette smoke exposure on
pulmonary vascular structure and function in the guinea pig. Exp
Lung Res 1991;17:997–1009.
60. Hyde DM, Downey GP, Tablin F, Rosengren S, Giclas PC, Henson
PM, Worthen GS. Age-dependent neutrophil and blood flow respon-
siveness in acute pulmonary inflammation in rabbits. Am J Physiol
1997;272:L471–L478.
61. Kelly LJ, Mitzner W, Spannhake EW, Bromberger-Barnea B, Menkes
HA. Pulmonary blood flow affects recovery from constriction in dog
lung periphery. J Appl Physiol 1986;60:1554–1560.
62. Kelly L, Kolbe J, Mitzner W, Spannhake EW, Bromberger-Barnea B,
Menkes H. Bronchial blood flow affects recovery from constriction
in dog lung periphery. J Appl Physiol 1986;60:1954–1959.
63. Wagner EM, Foster WM. Importance of airway blood flow on particle
clearance from the lung. J Appl Physiol 1996;81:1878–1883.
64. Wagner EM. Aerosol clearance by the bronchial circulation. J Aerosol
Med 1996;9:7–10.
65. Ritman EL, Robb RA, Harris LD. Imaging physiological functions:
experience with the DSR. Philadelphia: Praeger; 1985.
66. Hoffman EA, Larsen RL. Regional pulmonary blood flow via X-ray
CT and biodegradable radiopaque microspheres. Circulation 1990;4:
111–124.
67. Boyd DP, Lipton MJ. Cardiac computed tomography. Proc IEEE 1983;
71:298–307.
68. Saito T, Misaki M, Shirato K, Takishima T. Three-dimensional quantita-
tive coronary angiography. IEEE Trans Biomed Eng 1990;37:768–777.
69. Saito Y. Multislice X-ray CT scanner. Med Rev 1998;98:1–8.
70. Wang G, Lin TH, Cheng PC, Shinozaki DM. A general cone-beam
reconstruction algorithm. IEEE Trans Med Imaging 1993;12:486–496.
71. Kao CY, Hoffman EA, Beck KC, Bellamkonda RV, Annapragada AV.
Long-residence-time nano-scale liposomal iohexol for X-ray-based
blood pool imaging. Acad Radiol 2003;10:475–483.
72. Saam BT, Yablonskiy DA, Kodibagkar VD, Leawoods JC, Gierada
DS, Cooper JD, Lefrak SS, Conradi MS. MR imaging of diffusion
of (3)He gas in healthy and diseased lungs. Magn Reson Med 2000;
44:174–179.
73. Salerno M, de Lange EE, Altes TA, Truwit JD, Brookeman JR, Mugler
JP III. Emphysema: hyperpolarized helium 3 diffusion MR imaging
of the lungs compared with spirometric indexes—initial experience.
Radiology 2002;222:252–260.
74. Yablonskiy DA, Sukstanskii AL, Leawoods JC, Gierada DS, Bretthorst
GL, Lefrak SS, Cooper JD, Conradi MS. Quantitative in vivo assess-
ment of lung microstructure at the alveolar level with hyperpolarized
3He diffusion MRI. Proc Natl Acad Sci USA 2002;99:3111–3116.
75. Owers-Bradley JR, Fichele S, Bennattayalah A, McGloin CJ, Bowtell
RW, Morgan PS, Moody AR. MR tagging of human lungs using
hyperpolarized 3He gas. J Magn Reson Imaging 2003;17:142–146.
76. MacFall JR, Charles HC, Black RD, Middleton H, Swartz JC, Saam B,
Driehuys B, Erickson C, Happer W, Cates GD, et al. Human lung
air spaces: potential for MR imaging with hyperpolarized He-3.
Radiology 1996;200:553–558.
77. de Lange EE, Mugler JP III, Brookeman JR, Knight-Scott J, Truwit
JD, Teates CD, Daniel TM, Bogorad PL, Cates GD. Lung air spaces:
MR imaging evaluation with hyperpolarized 3He gas. Radiology
1999;210:851–857.
78. Kauczor HU, Ebert M, Kreitner KF, Nilgens H, Surkau R, Heil W,
Hofmann D, Otten EW, Thelen M. Imaging of the lungs using 3He
MRI: preliminary clinical experience in 18 patients with and without
lung disease. J Magn Reson Imaging 1997;7:538–543.
79. Altes TA, Powers PL, Knight-Scott J, Rakes G, Platts-Mills TA, de
Lange EE, Alford BA, Mugler JP III, Brookeman JR. Hyperpolarized
3He MR lung ventilation imaging in asthmatics: preliminary findings.
J Magn Reson Imaging 2001;13:378–384.
80. Viallon M, Berthezene Y, Callot V, Bourgeois M, Humblot H, Briguet
A, Cremillieux Y. Dynamic imaging of hyperpolarized (3)He distribu-
tion in rat lungs using interleaved-spiral scans. NMR Biomed 2000;
13:207–213.
81. Salerno M, Altes TA, Brookeman JR, de Lange EE, Mugler JP III.
Dynamic spiral MRI of pulmonary gas flow using hyperpolarized
(3)He: preliminary studies in healthy and diseased lungs. Magn Reson
Med 2001;46:667–677.
530 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 3 2006
82. Wild JM, Paley MN, Kasuboski L, Swift A, Fichele S, Woodhouse N,
Griffiths PD, van Beek EJ. Dynamic radial projection MRI of inhaled
hyperpolarized 3He gas. Magn Reson Med 2003;49:991–997.
83. Eberle B, Weiler N, Markstaller K, Kauczor H, Deninger A, Ebert M,
Grossmann T, Heil W, Lauer LO, Roberts TP, et al. Analysis of
intrapulmonary O(2) concentration by MR imaging of inhaled hyper-
polarized helium-3. J Appl Physiol 1999;87:2043–2052.
84. Deninger AJ, Eberle B, Ebert M, Grossmann T, Heil W, Kauczor H,
Lauer L, Markstaller K, Otten E, Schmiedeskamp J, et al. Quantifica-
tion of regional intrapulmonary oxygen partial pressure evolution
during apnea by (3)He MRI. J Magn Reson 1999;141:207–216.
85. Rizi RR, Baumgardner JE, Ishii M, Spector ZZ, Edvinsson JM, Jalali
A, Yu J, Itkin M, Lipson DA, Gefter W. Determination of regional
VA/Q by hyperpolarized 3He MRI. Magn Reson Med 2004;52:65–72.
86. Hoffman EA, Clough AV, Christensen GE, Lin CL, McLennan G,
Reinhardt JM, Simon BA, Sonka M, Tawhai MH, van Beek EJ,
et al. The comprehensive imaging-based analysis of the lung: a forum
for team science. Acad Radiol 2004;11:1370–1380.
87. Hoffman EA, Behrenbeck T, Chevalier PA, Wood EH. Estimation of
regional pleural surface expansile forces in intact dogs. J Appl Physiol
1983;55:935–948.
88. Hoffman EA, Lai-Fook SJ, Wei J, Wood EH. Regional pleural surface
expansile forces in intact dogs by wick catheters. J Appl Physiol 1983;
55:1523–1529.
89. Hoffman EA, Sinak LJ, Robb RA, Ritman EL. Noninvasive quantita-
tive imaging of shape and volume of lungs. J Appl Physiol 1983;54:
1414–1421.
90. Hoffman EA. Effect of body orientation on regional lung expansion: a
computed tomographic approach. J Appl Physiol 1985;59:468–480.
91. Hoffman EA, Ritman EL. Effect of body orientation on regional lung
expansion in dog and sloth. J Appl Physiol 1985;59:481–491.
92. Hoffman EA, Acharya RS, Wollins JA. Computer-aided analysis of
regional lung air content using three-dimensional computed tomo-
graphic images and multinomial models. Int J Math Model 1986;7:
1099–1116.
93. Reinhardt JM, D’Souza ND, Hoffman EA. Accurate measurement of
intra-thoracic airways. IEEE Trans Med Imaging 1998;16:820–827.
94. Tschirren J, Hoffman EA, McLennan G, Sonka M. Intrathoracic airway
trees: segmentation and airway morphology analysis from low dose
CT scans. IEEE Trans Med Imaging 2005;24:1529–1539.
95. Tschirren J, McLennan G, Palagyi K, Hoffman E, Sonka M. Matching
and anatomical labeling of human airway tree. IEEE Trans Med
Imaging (In press)
96. Liu YH, Hoffman EA, Hagler DJ, Seward JB, Julsrud PR, Mair DD,
Ritman EL. Accuracy of pulmonary vascular dimensions estimated
with the dynamic spatial reconstructor. Am J Physiol Imaging 1986;1:
201–207.
97. Shikata H. EA H, Sonka M. Automated segmentation of pulmonary
vascular tree from 3D CT images. Prog Biomed Optics Imaging 2004;
5:107–116.
98. RobbRA,SinakLJ,HoffmanEA,KinseyJH,HarrisLD,RitmanEL.
Dynamic volume imaging of moving organs. JMedSyst1982;6:539–554.
99. Hu S, Hoffman EA, Reinhardt JM. Automatic lung segmentation for
accurate quantitation of volumetric X-ray CT images. IEEE Trans
Med Imaging 2001;20:490–498.
100. Zhang L, Hoffman EA, Reinhardt JM. Lung lobar segmentation by
graph search with 3D shape constraints. In: Chen C-T, Clough AV,
editors. Medical imaging 2001. Physiology and function from multidi-
mensional images; Feb 18–20, 2001, San Diego. Bellingham, WA:
SPEI. Proceedings of SPIE; vol. 4321.
101. Zhang L, Hoffman EA, Reinhardt JM. Atlas-driven lung lobe seg-
mentation in volumetric x-ray CT images. In: Clough AV, Amini AA,
editors. Medical imaging 2003. Physiology and function––methods,
systems, and applications; Feb 16–18, 2003, San Diego. Bellingham,
WA: SPEI. Proceedings of SPIE; vol. 5031.
102. Reinhardt JM, Raab SA, D’Souza ND, Hoffman E. Intrathoracic airway
measurement: ex-vivo validation. In: Hoffman EA, editor. Medical
imaging 1997. Physiology and function from multidimensional images;
Feb 23–25. 1997. Newport Beach, CA. Bellingham, WA: SPEI. Pro-
ceedings of SPIE; vol. 3033.
103. Saba O, Beck K, Chon D, Nixon E, Hoffman E. Lung volume measure-
ment reproducibility using gated-axial and spiral multi-row detector
CT (MDCT) [abstract]. Am J Respir Crit Care Med 2003;167:A844.
104. Tschirren J, Pala
´
gyi K, Hoffman EA, Sonka M. Segmentation, skeletoni-
zation, and branchpoint matching: a fully automated quantitative
evaluation of human intrathoracic airway trees. In: Dohi T, Kikinis
R, editors. Proceedings of the Fifth International Conference on
Medical Image Computing and Computer Assisted Intervention,
2002, Tokyo, Japan. Berlin: Springer-Verlag; 2002. pp. 12–19.
105. Tschirren J, Hoffman EA, McLennan G, Sonka M. Branchpoint labeling
and matching in human airway trees. In: Clough AVA, Amir A.,
editors. Proceedings of the SPIE Medical Imaging, 2003, San Diego,
CA; 2003. pp. 187–194.
106. Palagyi K, Tschirren J, Hoffman EA, Sonka M. Quantitative analysis
of pulmonary airway tree structures. Comput Biol Med 2005.
107. Adams H, Bernard M, McConnochie K. An appraisal of CT pulmonary
density mapping in normal subjects. Clin Radiol 1991;43:238–242.
108. Kinsella M, Muller NL, Abboud RT, Morrison NJ. DyBuncio A. Quanti-
tation of emphysema by computed tomography using a “density
mask” program and correlation with pulmonary function tests. Chest
1990;97:315–321.
109. Muller NL, Staples CA, Miller RR, Abboud RT. “Density mask”: an
objective method to quantitate emphysema using computed tomogra-
phy. Chest 1988;94:782–787.
110. Gould GA, Macnee W, McLean A, Warren PM, Redpath A, Best JJK,
Lamb D, Flenley DC. CT measurements of lung density in life can
quantitate distal airspace enlargement: an essential defining feature
of human emphysema. Am Rev Respir Dis 1988;137:380–392.
111. Hartley PG, Galvin JR, Hunninghake GW, Merchant JW, Yagla SJ,
Speakman SB, Schwartz DA. High-resolution CT-derived measures
of lung density are valid indexes of interstitial lung disease. J Appl
Physiol 1994;76:271–277.
112. Coxson HO, Rogers RM. Quantitative computed tomography of chronic
obstructive pulmonary disease. Acad Radiol 2005;12:1457–1463.
113. Newell JD Jr, Hogg JC, Snider GL. Report of a workshop: quantitative
computed tomography scanning in longitudinal studies of emphy-
sema. Eur Respir J 2004;23:769–775.
114. Stolk J, Ng WH, Bakker ME, Reiber JH, Rabe KF, Putter H, Stoel
BC. Correlation between annual change in health status and computer
tomography derived lung density in subjects with alpha1-antitrypsin
deficiency. Thorax 2003;58:1027–1030.
115. Uppaluri R, Hoffman E, Schwartz D, McLennan G, Hunninghake G,
Dayton C, Hartley P. Quantitative analysis of the chest CT in
asbestos-exposed subjects using an adaptive multiple feature method
[abstract]. Am J Respir Crit Care Med 1998;157:A276.
116. Uppaluri R, Mitsa T, Sonka M, Hoffman EA, McLennan G. Quantifica-
tion of pulmonary emphysema from lung CT images using texture
analysis. Am J Respir Crit Care Med 1997;156:248–254.
117. Uppaluri R, McLennan G, Enright P, Hoffman E. AMFM: a quantitative
assessment of early parenchymal changes in smokers [abstract]. Am
J Respir Crit Care Med 1998;157:A788.
118. Aberle DR, Gamsu G, Henschke CI, Naidich DP, Swensen SJ. A con-
sensus statement of the Society of Thoracic Radiology: screening for
lung cancer with helical computed tomography. J Thorac Imaging
2001;16:65–68.
119. Bild DE, Detrano R, Peterson D, Guerci A, Liu K, Shahar E, Ouyang
P, Jackson S, Saad MF. Ethnic differences in coronary calcification:
the Multi-Ethnic Study of Atherosclerosis (MESA). Circulation 2005;
111:1313–1320.
120. Reddy S, Berbaum K, Sprenger K, McLennan G, Hoffman E. Evalua-
tion of emphysema-like lung parenchyma in high risk smokers:
quantitation of low-dose multidetector-row computed tomography
[abstract]. Proc Am Thorac Soc 2005;2;A259.
121. Reddy S, Barr G, Berbaum K, McLennan G, van Beek E, Hoffman E.
Quantitative evaluation of emphysema: is coronary calcium screening
CT a surrogate for a full lung study? Radiological Society of North
America 91st Annual Meeting; Chicago, IL; 2005. p. 638.
122. Stoel BC, Bakker ME, Stolk J, Dirksen A, Stockley RA, Piitulainen E,
Russi EW, Reiber JH. Comparison of the sensitivities of 5 different
computed tomography scanners for the assessment of the progression
of pulmonary emphysema: a phantom study. Invest Radiol 2004;39:1–7.
123. Shaker SB, Dirksen A, Laursen LC, Skovgaard LT, Holstein-Rathlou
NH. Volume adjustment of lung density by computed tomography
scans in patients with emphysema. Acta Radiol 2004;45:417–423.
124. Boedeker KL, McNitt-Gray MF, Rogers SR, Truong DA, Brown MS,
Gjertson DW, Goldin JG. Emphysema: effect of reconstruction algo-
rithm on CT imaging measures. Radiology 2004;232:295–301.
125. Mishima M, Hirai T, Itoh H, Nakano Y, Sakai H, Muro S, Nishimura
K, Oku Y, Chin K, Ohi M, et al. Complexity of terminal airspace
geometry assessed by lung computed tomography in normal subjects
and patients with chronic obstructive pulmonary disease. Proc Natl
Acad Sci USA 1999;96:8829–8834.
126. Hruban RH, Meziane MA, Zerhouni EA, Khouri NF, Fishman EK,
Wheeler PS, Dumler JS, Hutchins GM. High resolution computed
Hoffman, Simon, and McLennan: CT-Based Lung Structure and Function 531
tomography of inflation fixed lungs: pathologic-radiologic correlation
of centrilobular emphysema. Am Rev Respir Dis 1987;136:935–940.
127. Miller RA, Muller NL, Vedal S, Morrison NJ, Staples CA. Limitations
of computed tomography in the assessment of emphysema. Am Rev
Respir Dis 1989;139:980–983.
128. Murata K, Itoh H, Senda M, Yonekura Y, Nishimura K, Izumi T, Oshima
S, Torizuka K. Stratified impairment of pulmonary ventilation in
diffuse panbronchiolitis: PET and CT studies. J Comput Assist
Tomogr 1989;13:48–53.
129. Webb WR. High-resolution computed tomography of the lung: normal
and abnormal anatomy. Semin Roentgenol 1991;26:110–117.
130. Sanders C. Imaging of emphysema. Semin Respir Crit Care Med 1992;
13:318–330.
131. Sonka M, Hlavac V, Boyle R. Image processing, analysis, and machine
vision. London: Chapman and Hall; 1993.
132. Schad LR, Bluml S, Zuna I. MR tissue characterization of intracranial
tumors by means of texture analysis. Magn Reson Imaging 1993;11:
889–896.
133. Miller P, Astley S. Classification of breast tissue by texture analysis.
Image Vis Comput 1992;10:277–282.
134. Wu C, Chen Y. Multi-threshold dimension vector for texture analysis
and its application to liver tissue classification. Pattern Recognit 1993;
1:137–144.
135. McPherson D, Aylward P, Knosp B, Bean J, Kerber R, Collins S,
Skorton D. Ultrasound chracterization of acute myocardial ischemia
by quantitative texture analysis. Ultrason Imaging 1986;8:227–240.
136. Skorton DJ, Collins SM, Nichols J, Pandian NG, Bean JA, Kerber RE.
Quantitative texture analysis in two-dimensional echocardiography:
application to the diagnosis of experimental myocardial contusion.
Circulation 1983;68:217–223.
137. Chandrasekaran K, Aylward PE, Fleagle SR, Burns TL, Seward JB,
Tajik AJ, Collins SM, Skorton DJ. Feasibility of identifying amyloid
and hypertrophic cardiomyopathy with the use of computerized quan-
titative texture analysis of clinical echocardiographic data. J Am Coll
Cardiol 1989;13:832–840.
138. Uppaluri R, Mitsa T, Galvin JR. Fractal analysis of high-resolution CT
images as a tool for quantification of lung diseases. In: Hoffman EA,
editor. Proceedings SPIE Medical Imaging: Physiology and Function
from Multidimensional Images; 1995. pp. 133–142.
139. Tully RJ, Conners RW, Harlow CA, Lodwick GS. Towards computer
analysis of pulmonary infiltration. Invest Radiol 1978;13:298–305.
140. Katsuragawa S, Kunio D, MacMahon H. Image feature analysis and
computer-aided diagnosis in digital radiography: classification of nor-
mal and abnormal lungs with interstitial disease in chest images. Med
Phys 1989;16:38–44.
141. Fleagle S, Stanford W, Burns T, Skorton D. Feasibility of quantitative
texture analysis of cardiac magnetic resonance imagery: preliminary
results. SPIE Medical Imaging 1994;2168:23–32.
142. Uppaluri R, Hoffman EA, Sonka M, Hartley P, Hunninghake GW,
McLennan G. Computer recognition of regional lung disease patterns.
Am J Respir Crit Care Med 1999;160:648–654.
143. Uppaluri R, Hoffman EA, Sonka M, Hunninghake GW, McLennan G.
Interstitial lung disease: a quantitative study using the adoptive multiple
feature method. Am J Respir Crit Care Med 1999;159:519–525.
144. Xu Y, Sonka M, McLennan G, Guo J, Hoffman E. Sensitivity and
specificity of 3-D texture analysis of lung parenchymal is better than
2-D for discrimination of lung pathology in stage 0 COPD. In: Amini
A, Manduca A, editors. Proceedings SPIE Medical Imaging. 2005.
pp. 474–485.
145. Jones HA. Inflammation imaging. Proc Am Thorac Soc 2005;2:513–514.
146. Mills GH, Wild JM, Eberle B, Van Beek EJ. Functional magnetic
resonance imaging of the lung. Br J Anaesth 2003;91:16–30.
147. Peces-Barba G, Ruiz-Cabello J, Cremillieux Y, Rodriguez I, Dupuich
D, Callot V, Ortega M, Rubio Arbo ML, Cortijo M, Gonzalez-
Mangado N. Helium-3 MRI diffusion coefficient: correlation to mor-
phometry in a model of mild emphysema. Eur Respir J 2003;22:14–19.
148. Rizi RR, Lipson DA, Dimitrov IE, Ishii M, Roberts DA. Operating
characteristics of hyperpolarized 3He and arterial spin tagging in MR
imaging of ventilation and perfusion in healthy subjects. Acad Radiol
2003;10:502–508.
149. Kauczor HU, Hanke A, Van Beek EJ. Assessment of lung ventilation
by MR imaging: current status and future perspectives. Eur Radiol
2002;12:1962–1970.
150. Deninger AJ, Mansson S, Petersson JS, Pettersson G, Magnusson P,
Svensson J, Fridlund B, Hansson G, Erjefeldt I, Wollmer P, et al.
Quantitative measurement of regional lung ventilation using 3He
MRI. Magn Reson Med 2002;48:223–232.
151. Lipson DA, Roberts DA, Hansen-Flaschen J, Gentile TR, Jones G,
Thompson A, Dimitrov IE, Palevsky HI, Leigh JS, Schnall M, et al.
Pulmonary ventilation and perfusion scanning using hyperpolarized
helium-3 MRI and arterial spin tagging in healthy normal subjects
and in pulmonary embolism and orthotopic lung transplant patients.
Magn Reson Med 2002;47:1073–1076.
152. Moller HE, Chen XJ, Saam B, Hagspiel KD, Johnson GA, Altes TA,
de Lange EE, Kauczor HU. MRI of the lungs using hyperpolarized
noble gases. Magn Reson Med 2002;47:1029–1051.
153. Tajik JK, Chon D, Won C, Tran BQ, Hoffman EA. Subsecondmultisection
CT of regional pulmonary ventilation. Acad Radiol 2002;9:130–146.
154. Chon D, Simon BA, Beck KC, Shikata H, Saba OI, Won C, Hoffman
EA. Differences in regional wash-in and wash-out time constants for
xenon-CT ventilation studies. Respir Physiol Neurobiol 2005;148:65–83.
155. Chon D, Beck K, Shikata H, Saba O, Simon B, Hoffman E. Effect of
reduce inhaled [xenon] and [krypton] supplementation of signal noise
of regional CT-based ventilation measurements [abstract]. Am J
Respir Crit Care Med 2004;167:A547.
156. Ayappa I, Brown LV, Wang PM, Lai-Fook SJ. Arterial, capillary, and
venous transit times and dispersion measured in isolated rabbit lungs.
J Appl Physiol 1995;79:261–269.
157. Capen RL, Latham LP, Wagner WW Jr. Comparison of direct and
indirect measurements of pulmonary capillary transit times. J Appl
Physiol 1987;62:1150–1154.
158. Clough AV, Linehan JH, Dawson C. Regional perfusion parameters
from pulmonary microfocal angiograms. Am J Physiol 1997;272:
H1537–H1548.
159. Clough AV, Haworth ST, Hanger CC, Wang J, Roerig DL, Linehan
JH, Dawson CA. Transit time dispersion in the pulmonary arterial
tree. J Appl Physiol 1998;85:565–574.
160. Hoffman EA, Tajik JK. Dynamic and high resolution CT assessment
of pulmonary blood flow distributions. Am Rev Respir Dis 1993;
147:A201.
161. Mintun MA, Ter-Pergossian MM, Green MA, Lich LL, Schuster DP.
Quantitative measurement of regional pulmonary blood flow with
positron emission tomography. J Appl Physiol 1986;60:317–326.
162. Tajik JK, Tan BQ, Hoffman EA. CT-based assessment of regional
pulmonary blood flow parameters: an update. In: Chen C-T, Clough
AV, editors. Medical imaging 1999: physiology and function from
multidimensional images; 1999 05/99. San Diego, CA: SPIE; 1999.
pp. 181–187.
163. Wolfkiel CJ, Rich S. Analysis of regional pulmonary enhancement in
dogs by ultrafast computed tomography. Invest Radiol 1992;27:211–
216.
164. Bassingthwaighte JB, Raymond GR, Chan JIS. Principles of tracer
kinetics. In: Zaret BL, Beller GA, editors. Nuclear cardiology: state
of the art and future directions. Mosby: St. Louis, MO; 1993. pp.
3–23.
165. Bentley MD, Lerman LO, Hoffman EA, Fiksen-Olsen MJ, Ritman EL,
Romero JC. Measurement of renal perfusion and blood flow with
fast computed tomography. Circ Res 1994;74:945–951.
166. Eigler NL, Schuhlen H, Whiting JS, Pfaff JM, Zeiher A, Gu S. Digital
angiographic impulse response analysis of regional myocardial perfu-
sion: estimation of coronary flow, flow reserve, and distribution vol-
ume by compartmental transit time measurement in a canine model.
Circ Res 1991;68:870–880.
167. Ritman EL. Temporospatial heterogeneity of myocardial perfusion
and blood volume in the porcine heart wall. Ann Biomed Eng 1998;
26:519–525.
168. Musch G, Layfield JD, Harris RS, Melo MF, Winkler T, Callahan RJ,
Fischman AJ, Venegas JG. Topographical distribution of pulmonary
perfusion and ventilation, assessed by PET in supine and prone hu-
mans. J Appl Physiol 2002;93:1841–1851.
169. Levin DL, Chen Q, Zhang M, Edelman RR, Hatabu H. Evaluation
of regional pulmonary perfusion using ultrafast magnetic resonance
imaging. Magn Reson Med 2001;46:166–171.
170. Hatabu H, Tadamura E, Levin DL, Chen Q, Li W, Kim D, Prasad PV,
Edelman RR. Quantitative assessment of pulmonary perfusion with
dynamic contrast-enhanced MRI. Magn Reson Med 1999;42:1033–
1038.
171. Hoffman EA, Tajik JK, Kugelmass SD. Matching pulmonary structure
and perfusion via combined dynamic multislice CT and thin-slice
high-resolution CT. Comput Med Imaging Graph 1995;19:101–112.
172. Won C, Chon D, Tajik J, Tran B, Robinswood G, Beck K, Hoffman
E. CT-based assessment of regional pulmonary microvascular blood
flow parameters. J Appl Physiol 2003;94:2483–2493.
532 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 3 2006
173. Schuster DP, Marklin GF. The effect of regional lung injury or alveolar
hypoxia on pulmonary blood flow and lung water measured by posi-
tron emission tomography. Am Rev Respir Dis 1986;133:1037–1042.
174. Osborne DR, Effmann EL, Hedlund LW. Postnatal growth and size
of the pulmonary acinus and secondary lobule in man. AJR Am J
Roentgenol 1983;140:449–454.
APPENDIX
Scan Protocols
Scan protocols together with estimated dose calculations used
for these studies are provided in Tables 1 through 3.
Volume Scans
Our current volumetric protocol consists of 100 milliampere-
seconds (mAs), 120 kV, and 1-mm collimation, with an effective
slice thickness of 1.3 mm, overlap of 0.65 mm, and pitch of 1.2
mm. The slice parameter mode is 32 0.6 mm. We will use
512 512 slice matrices. The subject is apneic at a controlled
lung volume (40 and 95% VC). We carefully check the calibra-
tion of the scanner on a weekly basis. To estimate the effective
dose, we have used the WinDOSE program developed by Profes-
sor Willi Kalender (University of Erlangen, Germany) and the
CT dose index (CTDI) for the Siemens Sensation 64. The total
effective dose (He) is the primary measure that our radiation
safety committee evaluates. The radiation dose, as outlined in
Table 1, from the procedures is equal to the risk that the average
American experiences from exposure to 40 months of natural
background radiation.
Xenon Regional Ventilation
Reference whole lung scans obtained at static inflations of 40
and 95% VC are used for axial scan locations. A ventilation
study is performed at 20 time points with 80 kVp and 150 mAs.
The slice parameter mode is 20 1.2 mm so that a 2.4 cm (or
greater depending on the axial extent of the field-of-view on
future scanner configurations) z-axis coverage is achieved. To
estimate the effective dose, we have used the WinDOSE pro-
gram and the CTDI for the Siemens Sensation 64. The total
effective dose (He) is the primary measure that the radiation
safety committee evaluates. The radiation dose, as outlined in
Table 2, from the procedures is equal to the risk that the average
American experiences from exposure to 14 months of natural
background radiation.
The ECG signal is replaced by a signal from our custom data
acquisition and control program to trigger the scanner at specific
points during the ventilatory cycle. The subject breathes sponta-
neously with a mouthpiece connected to our lung volume con-
troller and two-way switching valve (room air and the Xe En-
hancer set to provide 30% Xe/30% O
2
). The subject is instructed
to maintain a constant breathing pattern by watching a graphical
TABLE 1. RADIATION DOSE ESTIMATES FOR TWO
VOLUME SCANS
Volume scans 64 slice
Two scans Male Female
Organs, dose (mrad)
Lung 2,060 2,100
Breast 0 1,920
Skeleton 1,040 1,200
Esophagus 1,430 1,600
Red marrow 630 680
Skin 3,900 3,900
H
E,
mrem 690 1,060
TABLE 2. RADIATION DOSE ESTIMATES FOR
VENTILATION STUDY
Ventilation 150 mAs 80 kV
15 scans Male Female
Organs, dose (mrad)
Lung 830 848
Breast 0 900
Skeleton 330 382.5
Esophagus 406 410
Red marrow 150 180
Skin 18,000 18,000
H
E,
mrem 236.25 360
display with target lines. To deliver xenon gas, we use an En-
hancer 9000, which allows for xenon recycling. CO
2
is scrubbed
from the exhalate and xenon and oxygen are sensed and replaced
to maintain a constant concentration of the inspired gas. The
scanner is activated via our PC software programmed in the
LabView (National Instruments) environment and three gated
images are taken as the pre-Xe baseline. The switching valve
connects the subject to 30% Xe gas. The subject inhales nine
breaths of Xe.
Bolus Contrast Regional Perfusion
Scanning is in the axial mode at the same slice locations as in
the ventilation study. To obtain regional perfusion (Q) with
contrast injection, the scanner is set up as in the Xe protocols
described above, with an ECG trigger signal, and the subject
remains apneic during scanning. A Medrad power injector sys-
tem (Mark V Power Injector; Medrad, Indianola, PA) is used
to give a 2-second bolus of contrast (0.5 ml/kg, up to a total
volume of 50 ml). The lung volume controller is used to start
breath-hold at normal functional residual capacity. Two to three
baseline images are obtained followed by dye injection. A total
of 12 stacked image sets, one per heartbeat, are obtained to
follow the contrast agent (Visipaque; GE Healthcare, Milwau-
kee, WI) through the lung fields. The scanner is setup in axial,
ECG triggering mode, using 80 kVp, 150 mAs, 360 rotations,
0.5-second scan time, 512 512 matrix, and the field of view
adjusted to fit the lung field of interest. The slice parameter
mode is 20 1.2 mm so that a 2.4-cm portion of the lung field
will be examined. To estimate the effective dose, we used the
WinDOSE program and the CTDI for the Siemens Sensation
64. The total effective dose (He) is the primary measure that
our radiation safety committee evaluates. The radiation dose
from the procedures, as outlined in Table 3, is equal to the risk
that the average American experiences from exposure to 19
months of natural background radiation.
TABLE 3. RADIATION DOSE ESTIMATES FOR
VENTILATION STUDY
Blood flow 150 mAs 80 kV
20 scans Male Female
Organs, dose (mrad)
Lung 1,320 1,140
Breast 0 960
Skeleton 660 580
Esophagus 540 620
Red marrow 200 240
Skin 24,200 24,200
H
E,
mrem 315 480
... These drawbacks can be addressed by the development of objective quantitative scoring methods, but to date quantitative assessment is mainly restricted to CT density-based quantification of emphysema [7]. Several new quantitative approaches have been proposed, especially for idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease outside emphysema [4,8]. These approaches mainly use histogram analysis, airway segmentation or texture analysis [4,[8][9][10]. ...
... Several new quantitative approaches have been proposed, especially for idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease outside emphysema [4,8]. These approaches mainly use histogram analysis, airway segmentation or texture analysis [4,[8][9][10]. However, their use is limited by the heterogeneity of the acquisition protocols, the CT manufacturer dependence of image characteristics and by the influence of physiological variables such as the level of inspiration on lung attenuation [4]. ...
Thesis
Disease staging and monitoring of chronic lung diseases are two major challenges for patient care and evaluation of new therapies. Monitoring mainly relies on pulmonary function testing but morphological assessment is a key point for diagnosis and staging In the first part, we propose different models to score bronchial disease severity on computed tomography (CT) scan. A simple thresholding approach using adapted thresholds and a more sophisticated machine learning approach with radiomics are evaluated In the second part, we evaluate deep learning methods to segment lung fibrosis on chest CT scans in patients with systemic sclerosis. We combine elastic registration to atlases of different thoracic morphology and deep learning to produce a model performing as well as radiologists In the last part of the thesis, we demonstrate that lung deformation assessment between inspiratory and expiratory magnetic resonance images can be used to depict fibrotic lung areas, which show less deformation during respiration and that CT assessment of lung deformation on serial CT scans can be used to diagnose lung fibrosis worsening
... The advent of quantitative computed tomography (CT) imaging techniques has enabled better evaluation of pulmonary disease. Multidetector CT has previously been used for detection of emphysema [1,2] and the development of the dual-energy CT has improved the differentiation between different contrast materials (i.e. iodine or xenon) and lung parenchyma for assessment of regional perfusion or ventilation [3,4]. ...
... Anatomical landmarks were used as a guide for correspondence between excised tissue samples and digital segmentation. Blood vessels were excluded by excluding voxels greater than 100 HU [1,28]. The average CT perfusion within each digital segment was compared to the corresponding reference fluorescent microsphere perfusion measurement. ...
Article
Full-text available
Purpose To evaluate the accuracy of a low-dose first-pass analysis (FPA) CT pulmonary perfusion technique in comparison to fluorescent microsphere measurement as the reference standard. Method The first-pass analysis CT perfusion technique was validated in six swine (41.7 ± 10.2 kg) for a total of 39 successful perfusion measurements. Different perfusion conditions were generated in each animal using serial balloon occlusions in the pulmonary artery. For each occlusion, over 20 contrast-enhanced CT images were acquired within one breath (320 x 0.5mm collimation, 100kVp, 200mA or 400mA, 350ms gantry rotation time). All volume scans were used for maximum slope model (MSM) perfusion measurement, but only two volume scans were used for the FPA measurement. Both MSM and FPA perfusion measurements were then compared to the reference fluorescent microsphere measurements. Results The mean lung perfusion of MSM, FPA, and microsphere measurements were 6.21 ± 3.08 (p = 0.008), 6.59 ± 3.41 (p = 0.44) and 6.68 ± 3.89 ml/min/g, respectively. The MSM (PMSM) and FPA (PFPA) perfusion measurements were related to the corresponding reference microsphere measurement (PMIC) by PMSM = 0.51PMIC + 2.78 (r = 0.64) and PFPA = 0.79PMIC + 1.32 (r = 0.90). The root-mean-square-error for the MSM and FPA techniques were 3.09 and 1.72 ml/min/g, respectively. The root-mean-square-deviation for the MSM and FPA techniques were 2.38 and 1.50 ml/min/g, respectively. The CT dose index for MSM and FPA techniques were 138.7 and 8.4mGy, respectively. Conclusions The first-pass analysis technique can accurately measure regional pulmonary perfusion and has the potential to reduce the radiation dose associated with dynamic CT perfusion for assessment of pulmonary disease.
... This allows the accurate characterization of attributes, such as density, texture, aeration, and Hafez et al. Egypt J Radiol Nucl Med (2022) 53:263 distribution of the lungs [3]. MDCT can cover large volumes during simple breath-holding, with good longitudinal, intrinsic spatial resolution and temporal resolution. ...
Article
Full-text available
Background Pediatric chronic lung disease (CLD) represents a heterogeneous group of several distinct clinical entities, with its prevalence increasing over the last decade. The current study aimed to identify the role of chest multidetector computed tomography (MDCT) using modified Bhalla scoring for the early diagnosis of CLD in pediatric patients and determine the most common chest MDCT findings. This prospective study involved 45 pediatric patients with chronic respiratory symptoms, all of whom underwent MDCT. Thereafter, data were analyzed using the modified Bhalla score. Results Chronic lung diseases were classified according to their radiological and clinical criteria. The total CT score, which was the most significant factor for chronic inhalation and chronic recurrent inflammatory lung diseases, varied between 2 and 21 points, with those having autoimmune diseases exhibiting the largest value for the mean CT score. The clinical severity of symptoms was not correlated with CT score. Conclusion Our findings showed that MDCT was a useful tool for diagnosing pediatric CLD and assessing disease extent, severity, and superimposed complications. The modified Bhalla CT scoring system allowed for systematic primary and follow-up assessments of various lung lesions in cases with varying CLD etiologies.
... 10 In addition, FOOTPRINTS is conducting MRI assessments, which could be valuable in improving our understanding of changes in vasculature and perfusion in the lung that may precede changes in emphysema. 51 Due to the small number of patients included in the subset analysis and the challenges associated with obtaining meaningful MRI data, these findings are expected to be limited. ...
Article
Full-text available
Introduction A better understanding is needed of the different phenotypes that exist for patients with chronic obstructive pulmonary disease (COPD), their relationship with the pathogenesis of COPD and how they may affect disease progression. Biomarkers, including those associated with emphysema, may assist in characterising patients and in predicting and monitoring the course of disease. The FOOTPRINTS study (study 352.2069) aims to identify biomarkers associated with emphysema, over a 3-year period. Methods and analysis The FOOTPRINTS study is a prospective, longitudinal, multinational (12 countries), multicentre (51 sites) biomarker study, which has enrolled a total of 463 ex-smokers, including subjects without airflow limitation (as defined by the 2015 Global Initiative for Chronic Obstructive Lung Disease (GOLD) strategy report), patients with COPD across the GOLD stages 1–3 and patients with COPD and alpha1-antitrypsin deficiency. The study has an observational period lasting 156 weeks that includes seven site visits and additional phone interviews. Biomarkers in blood and sputum, imaging data (CT and magnetic resonance), clinical parameters, medical events of special interest and safety are being assessed at regular visits. Disease progression based on biomarker values and COPD phenotypes are being assessed using multivariate statistical prediction models. Ethics and dissemination The study protocol was approved by the authorities and ethics committees/institutional review boards of the respective institutions where applicable, which included study sites in Belgium, Canada, Denmark, Finland, Germany, Japan, Korea, Poland, Spain, Sweden, UK and USA; written informed consent has been obtained from all study participants. Ethics committee approval was obtained for all participating sites prior to enrolment of the study participants. The study results will be reported in peer-reviewed publications. Trial registration number NCT02719184 .
... First, vascular pruning may impede the augmentation of pulmonary blood flow usually seen in infection, resulting in fewer cellular mediators to fight infection and aid in resolving lung injury. 11,35 Second, pulmonary vascular pruning may reflect endothelial dysfunction or activation, leading to increased recruitment of inflammatory cells. 36 Third, arterial pruning may impact capillary blood flow directly, leading to a slower transit of neutrophils or activated platelets through the pulmonary vasculature, leading to more inflammation. ...
Article
Background Pulmonary endothelial damage has been shown to preceed the development of emphysema in animals, and vascular changes in humans have been observed in COPD and emphysema. Research Question Is intraparenchymal vascular pruning associated with longitudinal progression of emphysema on CT or decline in lung function over 5 years? Study Design and Methods The COPDGene Study enrolled ever-smokers with and without COPD in 2008-2011. The percent of emphysema-like lung was assessed at baseline and after 5 years on non-contrast CT as the percentage of lung voxels <-950 Hounsfield units. An automated CT-based tool assessed and classified intrapulmonary arteries and veins. Spirometry measures are post-bronchodilator. Pulmonary arterial pruning was defined as a lower ratio of small artery volume (<5mm² cross sectional area) to total lung artery volume. Mixed linear models included demographics, anthropomorphics, smoking and COPD; with emphysema models also adjusting for CT scanner and lung function models adjusting for clinical center and baseline percent emphysema. Results At baseline the 4,227 participants were 60±9 years old, 50% female, 28% black, 47% current smokers and 41% had COPD. Median percent emphysema was 2.1 (IQR: 0.6, 6.3) and progressed 0.24 percentage points/year (95% CI: 0.22, 0.26) over 5.6 years. Mean FEV1/FVC was 68.5±14.2% and declined 0.26%/year (95% CI: -0.30, -0.23). Greater pulmonary arterial pruning was associated with more rapid progression of percent emphysema (0.11 percentage points/year per SD arterial pruning, 95% CI: 0.09, 0.16), including after adjusting for baseline percent emphysema and FEV1. Arterial pruning was also associated with a faster decline in FEV1/FVC (-0.04%/year per SD arterial pruning, 95% CI: -0.008, -0.001). Interpretation Pulmonary arterial pruning was associated with faster progression of percent emphysema and more rapid decline in FEV1/FVC over 5 years in ever-smokers, suggesting pulmonary vascular differences may be relevant in disease progression.
... 5,39,43 In this work, we will focus on established qCT methodologies that have been validated and utilized in various multicenter studies in collaboration with our research laboratory over last two decades. 31,32,[44][45][46][47][48][49][50][51] These methodologies have been combined with various statistical and other machine-learning analyses, imparting added insights into lung diseases. If appropriately used, these methods can provide direct insights into the disease severity, distribution, progression, and regression as well as identify underlying variations (sub-phenotypes such as parenchyma vs vasculature) in pathology, which may not be otherwise apparent. ...
Article
Full-text available
Increasingly, quantitative lung computed tomography (qCT)-derived metrics are providing novel insights into chronic inflammatory lung diseases, including chronic obstructive pulmonary disease (COPD), asthma, interstitial lung disease (ILD), and more. Metrics related to parenchymal, airway, and vascular anatomy together with various measures associated with lung function including regional parenchymal mechanics, air trapping associated with functional small airways disease (fSAD), and dual-energy derived measures of perfused blood volume are offering the ability to characterize disease phenotypes associated with the chronic inflammatory pulmonary diseases. With the emergence of COVID-19, together with its widely varying degrees of severity, its rapid progression in some cases, and the potential for lengthy post-COVID-19 morbidity, there is a new role in applying well-established qCT-based metrics. Based on the utility of qCT tools in other lung diseases, previously validated supervised classical machine learning (ML) methods, and emerging unsupervised ML and deep-learning approaches, we are now able to provide desperately needed insight into the acute and the chronic phases of this inflammatory lung disease. The potential areas in which qCT imaging can be beneficial include improved accuracy of diagnosis, identification of clinically distinct phenotypes, improvement of disease prognosis, stratification of care, and early objective evaluation of intervention response. There is also a potential role for qCT in evaluating an increasing population of post-COVID-19 lung parenchymal changes such as fibrosis. In this work, we discuss the basis of various lung qCT methods, using case-examples to highlight their potential application as a tool for the exploration and characterization of COVID-19, and offer scanning protocols to serve as templates for imaging the lung such that these established qCT analyses have the best chance at yielding the much needed new insights.
... To date, clinical thoracic CT scans have been validated as a non-invasive imaging technique to correlate with regional lung function [79] and to quantify emphysema (<−950 Hounsfield Units) [80,81]. However, the spatial resolution of clinical CT scans, 800-1000 μm, does not permit the analysis of the smallest conducting airways, or parenchymal structures [82][83][84]. ...
Article
Full-text available
Chronic obstructive pulmonary disease (COPD) is a devastating lung disease with a high personal and societal burden. Exposure to toxic particles and gases, including cigarette smoke, is the main risk factor for COPD. Next to smoking cessation, current treatment strategies of COPD aim to improve symptoms and prevent exacerbations, yet there is no disease modifying treatment. The biggest drawback of today's COPD treatment regime is the ‘one size fits all' pharmacological intervention, mainly based on disease severity and symptoms and not the individual's disease pathology. To halt the worrying increase in the burden of COPD, disease management needs to be advanced with a focus on personalized treatment. The main pathological feature of COPD includes a chronic and abnormal inflammatory response within the lungs which results in airway and alveolar changes in the lung as reflected by (small) airways disease and emphysema. Here we discuss recent developments related to the abnormal inflammatory response, extracellular matrix and age‐related changes, structural changes in the small airways, and the role of sex‐related differences, that are all relevant to explain the individual differences in disease pathology of COPD and improve disease endotyping. Furthermore, we will discuss the most recent developments of new treatment strategies using biologicals to target specific pathological features or disease endotypes of COPD. This article is protected by copyright. All rights reserved.
... volume at maximal inspiration with attenuation less than 2950 HU. Gas trapping was quantified as the percentage of lung volume at end expiration with attenuation less than 2856 HU (29). The square root of wall area for a hypothetical airway with an internal perimeter of 10 mm (Pi10) was derived (30). ...
Article
Rationale: The American Thoracic Society (ATS)/European Respiratory Society defines a positive bronchodilator response (BDR) by a composite of BDR in either forced expiratory volume in 1 second (FEV1) and/or forced vital capacity (FVC) greater than or equal to 12% and 200 ml (ATS-BDR). We hypothesized that ATS-BDR components would be differentially associated with important chronic obstructive pulmonary disease (COPD) outcomes. Objectives: To examine whether ATS-BDR components are differentially associated with clinical, functional, and radiographic features in COPD. Methods: We included subjects with COPD enrolled in the COPDGene study. In the main analysis, we excluded subjects with self-reported asthma. We categorized BDR into the following: 1) No-BDR, no BDR in either FEV1 or FVC; 2) FEV1-BDR, BDR in FEV1 but no BDR in FVC; 3) FVC-BDR, BDR in FVC but no BDR in FEV1; and 4) Combined-BDR, BDR in both FEV1 and FVC. We constructed multivariable logistic, linear, zero-inflated negative binomial, and Cox hazards models to examine the association of BDR categories with symptoms, computed tomography findings, change in FEV1 over time, respiratory exacerbations, and mortality. We also created models using the ATS BDR definition (ATS-BDR) as the main independent variable. Results: Of 3,340 COPD subjects included in the analysis, 1,083 (32.43%) had ATS-BDR, 182 (5.45%) had FEV1-BDR, 522 (15.63%) had FVC-BDR, and 379 (11.34%) had Combined-BDR. All BDR categories were associated with FEV1 decline compared with No-BDR. Compared with No-BDR, both ATS-BDR and Combined-BDR were associated with higher functional residual capacity %predicted, greater internal perimeter of 10 mm, and greater 6-minute-walk distance. In contrast to ATS-BDR, Combined-BDR was independently associated with less emphysema (adjusted beta regression coefficient, -1.67; 95% confidence interval [CI], -2.68 to -0.65; P = 0.001), more frequent respiratory exacerbations (incidence rate ratio, 1.25; 95% CI, 1.03-1.50; P = 0.02) and severe exacerbations (incidence rate ratio, 1.34; 95% CI, 1.05-1.71; P = 0.02), and lower mortality (adjusted hazards ratio, 0.76; 95% CI, 0.58-0.99; P = 0.046). Sensitivity analysis that included subjects with self-reported history of asthma showed similar findings. Conclusions: BDR in both FEV1 and FVC indicates a COPD phenotype with asthma-like characteristics, and provides clinically more meaningful information than current definitions of BDR.
... The pulmonary circulation is regulated by conditions within the lung and directly impacted by pulmonary disease. Within the lung, hypoxia causes localised pulmonary vasoconstriction [29], a response that optimises gas exchange but can be blunted in the setting of pulmonary infection [30,31]. We have found a lower pulmonary perfusion in those with chronic obstructive pulmonary disease and emphysema [32], and a higher TPVVCT in current and ex-smokers (with and without airflow limitation) [13]. ...
Article
Background: Air pollution alters small pulmonary vessels in animal models. We hypothesised that long-term ambient air pollution exposure would be associated with differences in pulmonary vascular volumes in a population-based study. Methods: The Multi-Ethnic Study of Atherosclerosis recruited adults in six US cities. Personalised long-term exposures to ambient black carbon, nitrogen dioxide (NO2), oxides of nitrogen (NO x ), particulate matter with a 50% cut-off aerodynamic diameter of <2.5 μm (PM2.5) and ozone were estimated using spatiotemporal models. In 2010-2012, total pulmonary vascular volume was measured as the volume of detectable pulmonary arteries and veins, including vessel walls and luminal blood volume, on noncontrast chest computed tomography (TPVVCT). Peripheral TPVVCT was limited to the peripheral 2 cm to isolate smaller vessels. Linear regression adjusted for demographics, anthropometrics, smoking, second-hand smoke, renal function and scanner manufacturer. Results: The mean±sd age of the 3023 participants was 69.3±9.3 years; 46% were never-smokers. Mean exposures were 0.80 μg·m-3 black carbon, 14.6 ppb NO2 and 11.0 μg·m-3 ambient PM2.5. Mean±sd peripheral TPVVCT was 79.2±18.2 cm3 and TPVVCT was 129.3±35.1 cm3. Greater black carbon exposure was associated with a larger peripheral TPVVCT, including after adjustment for city (mean difference 0.41 (95% CI 0.03-0.79) cm3 per interquartile range; p=0.036). Associations for peripheral TPVVCT with NO2 were similar but nonsignificant after city adjustment, while those for PM2.5 were of similar magnitude but nonsignificant after full adjustment. There were no associations for NO x or ozone, or between any pollutant and TPVVCT. Conclusions: Long-term black carbon exposure was associated with a larger peripheral TPVVCT, suggesting diesel exhaust may contribute to remodelling of small pulmonary vessels in the general population.
Article
In this study we tested the efficacy of quantitative texture analysis in the identification of acute myocardial ischemia using an ultrasound data acquisition system that digitizes and stores echocardiographic data in polar format. In nine closed-chest dogs, data were acquired before and after coronary occlusion using a 2.4 MHz echocardiographic system. Regions of interest were analyzed at end-diastole and end-systole from the ischemic area and normal area at the same depth of field. Ultrasound data were evaluated using previously reported quantitative gray level texture measures. After occlusion, texture changes indicative of ischemia were found in systolic images. The directional component of the data analysis was important; analysis in the azimuthal direction was more accurate than in the axial direction. Six texture measures exhibited significant changes in the ischemic region from control to occlusion when analyzing data in the azimuthal direction. One false positive result occurred (significant texture change in the normal region from control to occlusion) in the azimuthal direction. Several false positive alterations in the normal regions from control to occlusion were found when the texture was evaluated in the axial direction. For accurate assessment of ischemic changes, preocclusion image data were required. We conclude that quantitative echocardiographic texture analysis using polar format data can identify subtle changes in myocardial texture such as that due to acute ischemia, using data acquired through the Chest Wall.
Article
Knowledge of the contributions of arterial and venous transit time dispersion to the pulmonary vascular transit time distribution is important for understanding lung function and for interpreting various kinds of data containing information about pulmonary function. Thus, to determine the dispersion of blood transit times occurring within the pulmonary arterial and venous trees, images of a bolus of contrast medium passing through the vasculature of pump-perfused dog lung lobes were acquired by using an X-ray microfocal angiography system. Time-absorbance curves from the lobar artery and vein and from selected locations within the intrapulmonary arterial tree were measured from the images. Overall dispersion within the lung lobe was determined from the difference in the first and second moments (mean transit time and variance, respectively) of the inlet arterial and outlet venous time-absorbance curves. Moments at selected locations within the arterial tree were also calculated and compared with those of the lobar artery curve. Transit times for the arterial pathways upstream from the smallest measured arteries (200-mu m diameter) were less than similar to 20% of the total lung lobe mean transit time. Transit time variance among these arterial pathways (interpathway dispersion) was less than similar to 5% of the total variance imparted on the bolus as it passed through the lung lobe. On average, the dispersion that occurred along a given pathway (intrapathway dispersion) was negligible. Similar results were obtained for the venous tree. Taken together, the results suggest that most of the variation in transit time in the intrapulmonary vasculature occurs within the pulmonary capillary bed rather than in conducting arteries or veins.
Article
A prospective epidemiological study of the early stages of the development of chronic obstructive pulmonary disease was performed on London working men. The findings showed that forced expiratory volume in one second (FEV1) falls gradually over a lifetime, but in most non-smokers and many smokers clinically significant airflow obstruction never develops. In susceptible people, however, smoking causes irreversible obstructive changes. If a susceptible smoker stops smoking he will not recover his lung function, but the average further rates of loss of FEV1 will revert to normal. Therefore, severe or fatal obstructive lung disease could be prevented by screening smokers' lung function in early middle age if those with reduced function could be induced to stop smoking. Infective processes and chronic mucus hyper-secretion do not cause chronic airflow obstruction to progress more rapidly. There are thus two largely unrelated disease processes, chronic airflow obstruction and the hypersecretory disorder (including infective processes).
Article
We used a computed tomography (CT) scanner program (“density mask”) that highlights voxels within a given density range to quantitate emphysema by defining areas of abnormally low attenuation. We compared different density masks, mean lung attenuation, visual assessment of emphysema and the pathologic grade of emphysema in 28 patients undergoing lung resection for tumor. In each patient, a single representative CT image was compared with corresponding pathologic specimens of tissue. There was good correlation between the extent of emphysema as assessed by the density mask and the pathologic grade of emphysema. The optimal attenuation level to define areas of emphysema may vary in different scanners, but, once determined for a particular scanner, the density mask accurately assesses the extent of emphysema and eliminates interobserver and intraobserver variability. It has the added advantage of determining the exact percentage of lung parenchyma showing changes consistent with emphysema. (Chest 1988; 94:782-87)
Conference Paper
PURPOSE MDCT is used to screen subjects at risk for lung cancer and coronary atherosclerosis. We have previously shown that quantitative analysis of full lung scans provides extent and distribution of emphysema and is closely correlated with PFT-based COPD categories. An important question is whether a CT performed to screen for coronary calcium (CCS-CT) can also be used to assess presence and distribution of emphysema. METHOD AND MATERIALS We analyzed 386 full lung CTs of heavy smokers imaged via the American College of Radiology Imaging Network-sponsored National Lung Screening Trial. To match CCS-CT studies, we limited our analysis to slices from the pulmonary artery bifurcation to the heart apex. We compared % emphysema from just the cardiac region with that computed from the whole lung. Next, the cardiac region was divided into three sections along the length of the lung (1/8-3/4-1/8) and cumulative histograms of lung densities were computed for each. The apical-basal difference in % emphysema between the top and bottom 1/8 of the cardiac region was compared to the apical-basal difference in % emphysema for the whole lung. RESULTS Defining emphysema at -910 HU cut-off, % emphysema from partial lung data sets was correlated with the % emphysema from full lung data sets. Emphysema Presence: The following linear regression equation was generated: full lung % = 0.94 (partial lung %) + 1.33. 98% of the variation in emphysema from analyzing the full lung was accounted for by variation in emphysema from analyzing the cardiac region only (R2= 0.98). Emphysema Distribution (apical-basal difference): The regression equation was: full lung apical-basal difference = 1.02 (partial lung apical-basal difference) + 1.33. This time 68% of the variation in apical-basal difference from full lung data was accounted for by variation in apical-basal difference from CCS-CT data (R2= 0.68). CONCLUSION While full lung imaging is preferred, the reduced coverage associated with coronary calcium screening provides almost identical information on emphysema presence and reasonable information on distribution without the need for additional radiation exposure. Funded in part by:, HL-064368, HL-077612 and a Doris Duke Fellowship DISCLOSURE G.M.,E.A.H.: Partner, VIDA Diagnostics
Article
Lung parenchyma evaluation via multidetector-row CT (MDCT), has significantly altered clinical practice in the early detection of lung disease. Our goal is to enhance our texture-based tissue classification ability to differentiate early pathologic processes by extending our 2-D Adaptive Multiple Feature Method (AMFM) to 3-D AMFM. We performed MDCT on 34 human volunteers in five categories: emphysema in severe Chronic Obstructive Pulmonary Disease (COPD) as EC, emphysema in mild COPD (MC), normal appearing lung in COPD (NC), non-smokers with normal lung function (NN), smokers with normal function (NS). We volumetrically excluded the airway and vessel regions, calculated 24 volumetric texture features for each Volume of Interest (VOI); and used Bayesian rules for discrimination. Leave-one-out and half-half methods were used for testing. Sensitivity, specificity and accuracy were calculated. The accuracy of the leave-one-out method for the four-class classification in the form of 3-D/2-D is: EC: 84.9%/70.7%, MC: 89.8%/82.7%; NC: 87.5.0%/49.6%; NN: 100.0%/60.0%. The accuracy of the leave-one-out method for the two-class classification in the form of 3-D/2-D is: NN: 99.3%/71.6%; NS: 99.7%/74.5%. We conclude that 3-D AMFM analysis of the lung parenchyma improves discrimination compared to 2-D analysis of the same images.