ArticlePDF Available

Imaging of the pancreas: Recent advances

Authors:
  • Postgraduate Institute of Medical Education and Research, Dr. Ram Manohar Lohia Hospital

Abstract and Figures

A wide spectrum of anomalies of pancreas and the pancreatic duct system are commonly encountered at radiological evaluation. Diagnosing pancreatic lesions generally requires a multimodality approach. This review highlights the new advances in pancreatic imaging and their applications in the diagnosis and management of pancreatic pathologies. The mainstay techniques include computed tomography (CT), magnetic resonance imaging (MRI), endoscopic ultrasound (EUS), radionuclide imaging (RNI) and optical coherence tomography (OCT).
Content may be subject to copyright.
Indian Journal of Endocrinology and Metabolism / 2011 / Vol 15 / Supplement 1 S25
Imaging of the pancreas: Recent advances
Vikas Chaudhary, Shahina Bano1
Department of Radiodiagnosis, Employees’ State Insurance Corporation (ESIC) Model Hospital, Gurgaon – 122001, Haryana, 1Department of
Radiodiagnosis, Govind Ballabh (GB) Pant Hospital and Maulana Azad Medical College, New Delhi – 110 002, India
ABSTRACT
A wide spectrum of anomalies of pancreas and the pancreatic duct system are commonly encountered at radiological evaluation.
Diagnosing pancreatic lesions generally requires a multimodality approach. This review highlights the new advances in pancreatic
imaging and their applications in the diagnosis and management of pancreatic pathologies. The mainstay techniques include computed
tomography (CT), magnetic resonance imaging (MRI), endoscopic ultrasound (EUS), radionuclide imaging (RNI) and optical coherence
tomography (OCT).
Key words: Computed tomography, endoscopic ultrasound, magnetic resonance imaging, optical coherence tomography, pancreatic
imaging, radionuclide imaging, recent advances
Review Article
Access this article online
Quick Response Code:
Website:
www.ijem.in
DOI:
10.4103/2230-8210.83060
IntRoductIon
Pancreatic imaging is an essential tool in the early diagnosis
and staging of pancreatic disease. The diagnosis of
pancreatic diseases generally requires the combined use
of different imaging modalities, allowing the evaluation of
pancreatic ducts, the pancreatic parenchyma and adjacent
soft tissues. This review analyzes the most recent advances
in pancreatic imaging.
technIques of PancReatIc IMagIng
Since the introduction of computed tomography (CT)
scan in late 1970s, there has been dramatic improvement in
pancreatic imaging. With early conventional CT scanners,
only 10-mm thick slices with a large acquisition time of
1 minute/slice were obtained; this resulted in motion
artifacts and limited resolution. In addition, only ionic
intravenous contrast agent was administered slowly over
time. Helical (spiral) CT scanners, introduced in late 1980s,
allowed much faster data acquisition with a slice thickness
of 1–2 mm and a volume data set for three-dimensional
imaging. Power injectors were introduced now, allowing
bolus contrast administration for fast dynamic scanning.
The better spatial resolution and dedicated pancreatic
and portal venous phase (dual-phase helical CT) dynamic
scanning increased the tumor conspicuity and allowed
better detection and staging of pancreatic neoplasms.
However, the multiplanar imaging still suffered from
stair-stepping artifacts. This drawback was overcome with
the introduction of multidetector computed tomography
(MDCT) in late 1990s. In contrast to single-detector
helical CT scanners, these scanners use multiple detector
rows, are 10 times faster, and can obtain 16–64 slices per
rotation at a slice thickness of 0.5 mm. The MDCT has
improved volume coverage speed and spatial resolution
along z-axis, and allows three-dimensional reformatting due
to isotropic voxels and exquisite multiplanar reconstruction
of pancreatic anatomy. High speed of MDCT also allows
organ imaging in clearly dened perfusion phase.[1]
MDCT permits the acquisition in the arterial phase,
pancreatic (parenchymal) phase and portal venous (hepatic)
phase with a delay of 20, 40 and 70 sec, respectively, using
120 ml of iodinated contrast medium injected intravenously
Corresponding Author: Dr. Vikas Chaudhary, Department of Radiodiagnosis, Employees’ State Insurance Corporation (ESIC) Model Hospital,
Gurgaon – 122 001, Haryana, India. E-mail: dr_vikaschaudhary@yahoo.com
[Downloaded free from http://www.ijem.in on Wednesday, September 28, 2016, IP: 83.84.166.247]
Indian Journal of Endocrinology and Metabolism / 2011 / Vol 15 / Supplement 1
S26
at a rate of 3 ml/sec. Maximum enhancement of pancreas
and the maximum tumor-to-parenchymal attenuation
difference is achieved during pancreatic phase followed by
portal venous phase and the arterial phase. Therefore, for
tumor detection, particularly adenocarcinoma [Figure 1],
pancreatic and portal venous phases are superior to those
obtained in the arterial phase. However, for detection
of vascular invasion and liver metastases, the sensitivity
of images obtained in the portal venous phase is better
than those obtained in the pancreatic and arterial phases.
Images of the pancreas obtained in the arterial phase
are helpful in good visualization of the peripancreatic
arterial supply. Using this image acquisition, it is possible
to diagnose and characterize a small pancreatic lesion
(less than 2 cm in diameter) more accurately, establish
the level of peripancreatic vascular invasion and detect
liver metastasis. Most of the authors are of opinion that
the pancreatic parenchymal phase and the portal venous
phase (dual phase) are sufcient for the detection of the
pancreatic adenocarcinoma and the arterial phase may be
reserved for those patients who require CT angiography
(CTA). Thus, the biphasic contrast-enhanced MDCT
is a very effective diagnostic tool in the detection and
accurate preoperative staging of pancreatic malignancies,
which remain a challenge for radiologists. The addition of
multidetector CTA improves the accuracy of diagnosing
unresectable pancreatic carcinoma. Features indicating
vascular involvement include: tumor involvement for one
half of the vessel’s circumference, focal narrowing of the
vessels and dilatation of peripancreatic veins. Criteria for
unresectability include involvement of superior mesenteric
artery or celiac trunk, involvement of superior mesenteric
vein–portal vein conuence, and hepatic, peritoneal or
lymph nodal metastases. The major limitation with the use
of CT is that it cannot accurately differentiate between
benign and malignant lymph nodal enlargement.[1-5]
Recently, a 320-detector CT scan has been introduced.
Comparison of 320-detector volumetric and 64-detector
helical CT images of the pancreas revealed no signicant
difference in CT numbers of pancreas. Signal-to-noise
ratio (SNR) of the pancreas on biphasic images was
signicantly lower in the 320-detector group than in the
64-detector group. Image quality was acceptable in both the
groups and was slightly better in the 64-detector group for
pancreatic phase axial images and arterial phase multiplanar
reformatted images. No signicant difference was found in
the depiction of pancreatic parenchyma, main pancreatic
duct and focal pancreatic lesions.[6]
Majority of pancreatic endocrine tumors [Figures 2 and 3]
are small and very vascular and will be best seen in arterial
phase images; however, in some cases, portal venous phase
imaging best demonstrates the tumor. Thus, dual-phase
MDCT imaging, at 20 and 70 sec following intravenous
contrast infusion, is recommended to optimize the detection
of both the primary tumor and liver metastases.[7]
MDCT is the modality of choice for the diagnosis and
staging of acute pancreatitis. It is highly sensitive in detecting
the necrosis, the hallmark of severe acute pancreatitis
and peripancreatic uid collections. CT outclasses all
imaging modalities in detecting calcications [Figure 4],
a specific sign of advanced chronic pancreatitis, and
complications associated with chronic pancreatitis such
as pseudocyst, intraductal calculi, inammatory masses or
Figure 1: Pancreatic adenocarcinoma. Axial plain (a) and triple-phase
contrast-enhanced CT upper abdomen obtained in arterial (b), pancreatic
parenchymal (c) and portal venous (d) phase, in a 70-year-old male
who presented with severe abdominal pain, demonstrates an ill-dened
nonenhancing hypodense mass involving the head and proximal part of
body of pancreas (thick vertical white arrow). Peripancreatic extension
with encasement/attenuation of celiac axis branches (thin black arrow),
inltration and dilatation of main pancreatic duct (thin white arrow) and
liver metastases (black arrow head) are also evident which make the tumor
unt for resection. Note that the pancreatic and portal venous phases are
best for tumor detection, the hypovascular metastases stand out best in
portal venous phase, while the arterial phase is good for demonstration of
celiac axis encasement
a
b
c
d
Chaudhary and Bano: Pancreatic imaging
[Downloaded free from http://www.ijem.in on Wednesday, September 28, 2016, IP: 83.84.166.247]
Indian Journal of Endocrinology and Metabolism / 2011 / Vol 15 / Supplement 1 S27
Figure 2: Functioning islet cell tumor: Gastrinoma in a 25-year-old male
patient with Zollinger–Ellison syndrome (ZES). Axial plain (a) and dual-
phase contrast-enhanced CT abdomen obtained in arterial (b) and portal
venous (c, d) phase shows a small, well-dened mass of 1 × 0.5 cm size in
the gastric triangle (thick vertical white arrow). The lesion is homogenous
and hypodense on non-contrast CT, shows marked homogenous contrast
enhancement in arterial phase image (suggesting hypervascular nature) and
washout of contrast in portal venous phase. Majority of liver metastases from
the tumor are hypervascular, appearing as homogenous hyperenhancing
foci (thin white arrow) in arterial phase image, while a few are hypovascular
appearing as non-enhancing foci both on arterial and portal venous phase
images (thin black arrow). Due to hypervascular nature, both the tumor (thick
vertical white arrow) and metastases (thick vertical black arrow) appear
echogenic on ultrasound (e). Double-contrast barium meal (f) of the same
patient demonstrates thickened gastric folds. Endoscopy revealed peptic
ulceration of upper gastrointestinal tract. Patient also had hypergastrinemia
a
e
f
b
c d Figure 3: Functioning islet cell tumor: Insulinoma in a 55-year-old male
patient who presented with hypoglycemic symptoms. Axial plain (a) and
dual-phase contrast-enhanced CT abdomen obtained in portal venous
phase (b) shows a well-dened intrapancreatic subcentimeter size mass
(thin white arrow). The lesion is inconspicuous on non-contrast CT,
however shows marked homogenous contrast enhancement in portal
venous phase image
a b
Figure 4: Chronic pancreatitis. Axial non-contrast CT abdomen shows
features of chronic pancreatitis evidenced as atrophic pancreas, dilated
main pancreatic duct (thin black arrow) and dense calcications (thin white
arrow) in pancreatic head region and within the dilated main pancreatic duct
a b
pseudoaneurysm. However, its sensitivity to detect early
chronic pancreatitis is poor. Unenhanced CT has negative
attenuation value of pancreatic tissue replaced by the
fat, therefore can reliably diagnose diffuse fatty change
involving the pancreas.[8] MDCT is also the noninvasive
modality of choice for characterizing pancreatic cystic
lesions more accurately.[9]
MDCT perfusion study is an evolving and promising
technique having various applications. Perfusion CT
involves dynamic scanning after administration of
iodinated contrast material, followed by mathematical
modeling to study contrast material kinetics in the tissue.
The CT perfusion data set derived from kinetic model
allows assessment of physiological parameters such as
tumor and normal pancreatic blood ow (BF), blood
volume (BV), mean transit time (MTT) and permeability–
surface area product (PS). The normal pancreas displays
symmetrical BF, BV, MTT and PS. Comparing with normal
value, CT perfusion imaging helps in diagnosing various
pancreatic diseases (e.g. necrotizing acute pancreatitis, mass
Chaudhary and Bano: Pancreatic imaging
[Downloaded free from http://www.ijem.in on Wednesday, September 28, 2016, IP: 83.84.166.247]
Indian Journal of Endocrinology and Metabolism / 2011 / Vol 15 / Supplement 1
S28
forming chronic pancreatitis) and angiogenesis in different
pancreatic neoplasms (e.g. pancreatic and ampullary
adenocarcinoma, cystadenoma, endocrine tumors, solid
pseudopapillary neoplasm and pancreatic metastasis).[10-13]
It aids in differential diagnosis of pancreatic tumors by
detecting change in their perfusion pattern.[14] It may
help in early detection of small (<2 cm size) pancreatic
adenocarcinomas, when these masses are still resectable,
thereby improving the prognosis of the patients.[15] It has a
crucial role in efcient management in the eld of oncology
as it provides physiological information about tumor neo-
angiogenesis. New blood vessel growth is critical for tumor
growth and metastasis, and newer generation oncologic
treatment regimens target the neo-angiogenesis and the
growth factors that stimulate neo-angiogenesis.[16] Perfusion
CT can be used to predict tumor response to concurrent
chemotherapy and radiotherapy (CCRT).[17]
Dynamic contrast-enhanced CT is the mainstay of imaging
in suspected pancreatic injury, but is frequently normal in
the acute phase. Repeat CT is indicated after 24–48 hours
if there are persistent unexplained symptoms or elevated
amylase.[18]
Although CT remains the most effective imaging modality
for evaluation of the pancreas, magnetic resonance imaging
(MRI) is increasingly used for further identification
and characterization of pancreatic diseases. Technical
innovation in MRI, such as use of phased-array coils,
allows improved spatial resolution and faster T1- and T2-
weighted sequences for imaging the entire upper abdomen
in a single breathhold and providing cross-sectional images
of pancreatic parenchyma analogous to CT images. The
use of fat saturation pulses and dynamic studies following
gadolinium injection increases the sensitivity of MR in
detecting pancreatic lesions. MR angiography (MRA)
is useful in noninvasive evaluation of splanchnic blood
vessels. Half-Fourier T2-weighted pulse sequences for
magnetic resonance cholangiopancreaticography (MRCP)
allow pancreatic duct and side branch delineation
[Figure 5] and detection of anatomic variants such as
pancreatic divisum. Administration of secretin further
improves the conspicuity of the ductal system, allows
monitoring of pancreatic ow dynamics, helps in evaluation
of pancreatic exocrine function, and planning surgery or
therapeutic endoscopic and follow-up study after therapy.
Although MRI is accurate in local staging of the pancreatic
malignancies owing to high soft tissue contrast resolution
(for assessment of peripancreatic fat inltration), for
evaluation of vascular encasement, peritoneal deposits and
lymph nodal involvement, it has limitations as compared
to CT. However, for identifying the liver metastases,
MRI has high sensitivity and specicity when compared
Figure 5: Chronic pancreatitis. Axial fat suppression T2-weighted MR image
(a) and MRCP (b) shows atrophic pancreas with gross dilatation of main
pancreatic duct and side branches (thin white arrow)
a b
to CT. The use of liver-specic contrast agents further
improves the diagnostic value of MRI for detecting liver
metastases. Thus, MRI in combination with secretin-
enhanced MRCP and MRA is useful in the diagnosis and
management of pancreatic malignancies.[19,20] Focal fatty
replacement of the pancreas may appear as hypodense
mass on contrast-enhanced CT and thus mimic an ill-
dened neoplasm. MRI establishes the correct diagnosis
of focal fatty replacement of pancreas using dual-echo
(in-phase and opposed phase) chemical shift imaging
and avoids invasive diagnostic procedures and surgery.[21]
Diffusion-weighted MRI helps to differentiate the subtypes
of pancreatic endocrine neoplasms on the basis of tumor
cellularity and/or extracellular brosis that may account
for various apparent diffusion coefcient (ADC) values
in these tumors.[22] MRI is as sensitive as CT for the
depiction of necrosis and peripancreatic uid collection
in the case of acute pancreatitis [Figure 6], but is less
sensitive than CT for detection of calcications associated
with chronic pancreatitis. However, fat-suppressed T1-
weighted MRI is more sensitive for the detection of
early chronic pancreatitis, prior to the development of
calcications. MRI is also useful in differentiating pancreatic
pseudocysts [Figure 7] from pancreatic cystic neoplasms
[Figures 8–11]. Presence of internal dependant debris is a
highly specic MR nding for diagnosing pseudopancreatic
cyst.[23] Magnetic resonance spectroscopy (MRS) is a
promising clinical tool for oncologic management of
patients. MRS can differentiate chronic focal pancreatitis
from pancreatic cancer. In proton MRS, chronic focal
pancreatitis shows less lipid than pancreatic carcinoma
due to difference in brous tissue content in the two
conditions.[24] MRI also has an important role in the
pancreatic transplantation. Standard MRI, MRCP (secretin-
induced MRCP) and MRA demonstrate the pancreatic
Chaudhary and Bano: Pancreatic imaging
[Downloaded free from http://www.ijem.in on Wednesday, September 28, 2016, IP: 83.84.166.247]
Indian Journal of Endocrinology and Metabolism / 2011 / Vol 15 / Supplement 1 S29
Figure 6: Acute necrotizing pancreatitis. Axial T1W (a) and spectrally
selective inversion recovery (SPIR; fat-suppressed T2W) (b) MR images
through upper abdomen show severe acute necrotizing pancreatitis. The
body of pancreas is replaced by a large cystic appearing lesion (asterisk)
showing internal debris in the dependant portion of the cyst. Extensive
peripancreatic inammatory changes are also present, best appreciated
on SPAIR image. Pancreatic tissue in the tail region shows heterogeneous
signal intensity (thin white arrow) consistent with intraparenchymal
inammatory change
a b
Figure 7: Pancreatic pseudocyst in a 50-year-old male who presented
after an episode of pancreatitis. Axial T1W (a) and T2W (b) MR images
of abdomen show a large, walled-off cystic lesion localized to lesser sac
(asterisk). The lesion shows internal debris along with hemorrhage (arrow).
Presence of internal debris is a highly specic MR nding for diagnosing
pseudopancreatic cyst
a b
Figure 8: Solid and Papillary Epithelial Neoplasm in a 35-year-old female
who presented with epigastric pain. Axial non-contrast (a) and contrast-
enhanced (b) CT abdomen shows a mixed solid and cystic mass in the
pancreatic head (arrow). The diagnosis of SPEN was conrmed after surgery
on histopathologic examination
a b
Figure 9: Branch duct type Intraductal Papillary Mucinous Tumor (IPMT)
in a 55-year-old male who presented with vague abdominal pain. Axial
T2-weighted MR image (a) of upper abdomen shows a well-dened, thin-
walled cystic lesion in the pancreatic head (asterisk). Associated dilatation
of main pancreatic duct is also evident (thin white arrow), which appears
inseparable from the cystic mass lesion on MRCP image (b). ERCP revealed
communication between the cystic mass and the dilated proximal pancreatic
duct. The diagnosis of IPMT was conrmed on histopathologic examination
of the resected tumor
a b
anatomy well before and after transplantation. Serial
contrast-enhanced MRI may demonstrate diminished
perfusion in case of graft rejection, and the vascular
complications are assessed by MRA.[25]
Although high-resolution MDCT with 3D image
reconstruction remains the prime imaging modality for
diagnosing and staging pancreatic cancers, endoscopic
ultrasound (EUS) can be a valuable adjunct to MDCT for
diagnostic evaluation of patients with suspected pancreatic
tumors. EUS with ne needle aspiration cytology (EUS-
FNA) is a highly accurate method for preoperative staging
of pancreatic cancer, as it has the ability to obtain the
tissue conrmation and permit accurate nodal staging
without relying on lymph node size.[26] The intraoperative
ultrasound (IOUS) and laparoscopic ultrasound (LUS) are
highly sensitive methods to assess tumor resectability during
surgery as they permit accurate assessment of location and
number of lesions, locoregional tumor extension, vascular
Chaudhary and Bano: Pancreatic imaging
[Downloaded free from http://www.ijem.in on Wednesday, September 28, 2016, IP: 83.84.166.247]
Indian Journal of Endocrinology and Metabolism / 2011 / Vol 15 / Supplement 1
S30
Figure 10: Mucinous cystadenocarcinoma in a 42-year-old female who
presented with recurrent upper abdominal pain. Axial T2-weighted MR (a)
and MRCP (b) images of abdomen show multilocular macrocystic lesion
(thick white arrow) involving pancreatic head. Note ne internal septations
within the lesion and associated dilatation of MPD and side branches
(thin white arrow). The diagnosis of mucinous cystadenocarcinoma was
conrmed after surgery and histopathologic examination
a b
Figure 11: Serous cystadenocarcinoma in a 72-year-old male who presented with jaundice and recurrent abdominal pain. T2-weighted MRI (a) and MRCP
(b) show multilocular microcystic lesion involving pancreatic head (thick white arrow). The lesion shows very thin internal septations. Large pseudocyst
(asterisk), dilated common bile duct/intrahepatic biliary radicles (CBD/IHBR) (thin black arrow) and normal MPD (white arrow head) are well seen on
MRCP. Cystogastrostomy and choledochoduodenostomy were performed to drain the pseudocyst and relieve the jaundice. Three months later, follow-
up CECT abdomen (c) revealed increase in size of tumor (thick white arrow), multiple liver metastases (thick black arrow) and encasement of superior
mesenteric vessels (thin white arrow) by the cystic mass. The diagnosis of serous cystadenocarcinoma made on imaging was conrmed after surgery
and histopathologic examination
a b c
involvement and lymph nodal or liver metastases.[27] EUS,
particularly the intraductal endoscopic ultrasonography
(IDUS), accurately localizes the pancreatic endocrine tumors,
especially those which are too small to be characterized by
CT or MRI. EUS with color Doppler further improves
the detection of small pancreatic endocrine tumors and
adenocarcinomas. Endocrine tumors, being hypervascular,
demonstrate abundant color Doppler signal having pulsatile
and/or continuous waveform pattern, while majority of
adenocarcinomas demonstrate low vascularity.[28] Contrast-
enhanced EUS using microbubbles has also shown to
improve the detection and characterization of pancreatic
lesions and liver metastasis.[29] EUS-FNA for cytology and
cyst uid analysis aids in the differential diagnosis of cystic
lesions of pancreas that are indeterminate at cross-sectional
imaging.[30] Finally, EUS may also be used therapeutically
in image-guided drainage such as gastrocystostomy in
pancreatic pseudocyst and celiac plexus neurolysis for pain
control in patients of pancreatic cancer or pancreatitis.[31]
Ultrasound elastography, a new technique, evaluates the
relative stiffness of the tissues. EUS real-time elastography
distinguishes normal pancreas from the abnormal pancreas
affected by inammatory or focal disease. However, it
cannot differentiate chronic pancreatitis from malignant
tumor because of their similar brous architecture. This
technique is also useful to select lymph nodes suitable for
biopsy as it can differentiate between benign and malignant
lymph node involvement.[32]
Radionuclide imaging (RNI) aids in improving the diagnosis
and staging of the pancreatic tumors, identifying and
localizing disseminated disease, differentiating post-
treatment recurrent and residual disease from brosis,
and planning and monitoring response to the therapy.
Detection of pancreatic cancer at early stage improves
the long-term survival of the patient. The diagnosis
of early stage pancreatic cancer (small in size, free of
peripancreatic extension and without lymph nodal/liver
metastases) is often difcult with the structural imaging
techniques. [18F]-uorodeoxyglucose positron emission
tomography (FDG-PET) scanning has been found to be
more accurate than other imaging modalities for diagnosing
early pancreatic cancer. The sensitivity of PET is superior
to CT in detecting lesions less than 2 cm in diameter, but
CT scanning is superior to PET for diagnosing cancers
larger than 4 cm in diameter because of lower metabolic
Chaudhary and Bano: Pancreatic imaging
[Downloaded free from http://www.ijem.in on Wednesday, September 28, 2016, IP: 83.84.166.247]
Indian Journal of Endocrinology and Metabolism / 2011 / Vol 15 / Supplement 1 S31
rates in larger tumors.[33] The sensitivity of FDG-PET
for detecting lymph node metastasis in patients with
pancreatic cancer and differentiating pancreatic cancer
from chronic pancreatitis is more than that of CT or
MRI.[34] FDG-PET can also alter the management of
pancreatic cancer by revealing unsuspected metastases
to liver, lung and bone, thereby avoiding the mortality
and morbidity associated with unnecessary surgical
intervention.[35] FDG-PET has the advantage of
differentiating residual and recurrent tumor from
postoperative inammation or brosis.[36] It is also useful
in the follow-up of patients undergoing chemoradiation
therapy or surgical resection.[37] FDG-PET is less sensitive
for detecting pancreatic endocrine tumors, particularly
those which are non-functional and have small size at
presentation. However, two PET radiopharmaceutical
agents such as C-11 labeled 5-hydroxytryptophan (5-HPT)
and l-Dopa are known to detect endocrine pancreatic
tumors.[38] FDG-PET has been found to be more accurate
than CT in characterizing cystic pancreatic lesions as
malignant.[39] The main drawback of PET is its relative
low spatial resolution which limits its role in detecting
direct invasion of adjacent structures or encasement of
blood vessels; these factors are important in planning
surgery. 111In-octreotide single-photon emission computed
tomography (SPECT) has also shown to improve the
localization of pancreatic endocrine tumors. It has been
established that the anatomical–functional image fusion
techniques such as hybrid PET/CT and SPECT/CT
improve the localization and characterization of pancreatic
endocrine tumors and therefore alter the treatment plan.[40]
Optical coherence tomography (OCT) is a new optical
imaging modality introduced in 1991. It uses infrared light
to produce high-resolution, cross-sectional, subsurface
imaging of the microstructure. It has a promising role
in evaluating pancreaticobiliary ductal system, as it can
recognize different patterns of the duct wall structure
in neoplastic and non-neoplastic conditions. OCT has
high diagnostic accuracy, better than brush cytology, for
distinguishing neoplastic from a non-neoplastic MPD
stricture.[41]
Thus, we conclude that MDCT, MRI, EUS and RNI are
excellent modalities for both detection and characterization
of pancreatic lesions. Structural imaging techniques such
as CT and MR provide superior information regarding
local tumor invasion and surgical resectability, whereas
FDG-PET offers a noninvasive and accurate method
for detection of early pancreatic cancer, unsuspected
metastases, differentiation between benign and malignant
pancreatic lesions (such as inammatory or scar tissue
from recurrent or residual tumor), and evaluation of
pancreatic masses with equivocal CT/MRI diagnosis.
EUS-guided aspiration and biopsy is useful in cases that are
indeterminate at cross-sectional imaging. OCT has emerged
as a new technique that differentiates between neoplastic
and non-neoplastic pancreatic duct stricture.
RefeRences
1. Fenchel S, Boll DT, Fleiter TR, Brambs HJ, Merkle EM. Multisclice
helical CT of the pancreas and spleen. Eur J Radiol 2003;45 Suppl
1:S59-72.
2. Lu DS, Vedantham S, Krasny RM, Kadell B, Berger WL, Reber HA.
Two-phase helical CT for pancreatic tumors: Pancreatic versus hepatic
phase enhancement of tumor, pancreas and vascular structures.
Radiology 1996;199:697-701.
3. Boland GW, O’Malley ME, Saez M, Fernandez-del-Castillo C,
Warshaw AL, Mueller PR. Pancreatic-phase versus portal vein-
phase helical CT of the pancreas: Optimal temporal window for
evaluation of pancreatic adenocarcinoma. AJR Am J Roentgenol
1999;172:605-8.
4. McNulty NJ, Francis IR, Platt JF, Cohan RH, Korobkin M,
Gebremariam A. Multi-detector row helical CT of the pancreas:
effect of contrast-enhanced multiphasic imaging on imaging of the
pancreas, peripancreatic vasculature and pancreatic adenocarcinoma.
Radiology 2001;220:97-102.
5. Zamboni GA, Kruskal JB, Vollmer CM, Baptista J, Callery
MP, Raptopoulos VD. Pancreatic Adenocarcinoma: Value of
Multidetector CT Angiography in preoperative evaluation. Radiology
2007;245:770-8.
6. Goshima S, Kanematsu M, Nishibori H, Sakurai K, Miyazawa D,
Watanabe H, et al. CT of the Pancreas: Comparison of Anatomic
Structure Depiction, Image Quality, and Radiation Exposure between
320-Detector Volumetric Images and 64-Detector Helical Images.
Radiology 2011;260:139-47.
7. Ichikawa T, Peterson MS, Federle MP, Baron RL, Haradome H,
Kawamori Y, et al. Islet cell tumor of the pancreas: Biphasic CT
versus MR imaging in tumor detection. Radiology 2000;216:163-71.
8. Anand R, Narula MK, Chaudhary V, Agrawal R. Total pancreatic
lipomatosis with malabsorption syndrome. Indian J Endocrinol
Metab 2011;15:51-3.
9. Kim YH, Saini S, Sahani D, Hahn PF, Mueller PR, Auh YH.
Imaging Diagnosis of cystic Pancreatic Lesions: Pseudocyst versus
Nonpseudocyst. Radiographics 2005;25:671-85.
10. Tsuji Y, Hamaguchi K, Watanabe Y, Okumura A, Isoda H, Yamamato
N, et al. Perfusion CT is superior to angiography in predicting
pancreatic necrosis in patients with severe acute pancreatitis. J
Gastroenterol 2010;45:1155-62.
11. Lu N, Feng XY, Hao SJ, Liang ZH, Jin C, Qiang JW, et al. 64-Slice CT
Perfusion Imaging of Pancreatic Adenocarcinoma and Mass-Forming
Chronic Pancreatitis. Acad Radiol 2011;18:81-8.
12. Kandel S, Kloeters C, Meyer H, Hein P, Hilbig A, Rogalla P. Whole-
organ perfusion of the pancreas using dnamic volume CT in patients
with primary pancreas carcinoma: Acquisition technique, post-
processing and initial results. Eur Radiol 2009;19:2641-6.
13. Technology-Articles.Org. Study of Multisection Helical CT Perfusion
Imaging for Pancreatic Diseases and Angiogenesis in Pancreatic
Neoplasm. Free Online Science and Technology Articles, 2010.
14. d‘Assignies G, Couvelard A, Bahrami S, Vullierme MP, Hammel
P, Hentic O, et al. Pancreatic endocrine tumors: Tumor blood flow
assessed with perfusion CT reflects angiogenesis and correlates with
prognostic factors. Radiology 2009:250:407-16.
Chaudhary and Bano: Pancreatic imaging
[Downloaded free from http://www.ijem.in on Wednesday, September 28, 2016, IP: 83.84.166.247]
Indian Journal of Endocrinology and Metabolism / 2011 / Vol 15 / Supplement 1
S32
15. Fukushima H, Itoh S, Takada A, Mori Y, Suzuki K, Iwano S,
et al. Diagnostic value of curved multiplanar reformatted images
in multislice CT for the detection of resectable pancreatic ductal
adenocarcinoma. Eur Radiol 2006;16:1709-18.
16. Miles KA, Charnsangavej C, Lee FT, Fishman EK, Horton K, Lee TY.
Application of CT in the investigation of angiogenesis in oncology.
Acad Radiol 2000;7:840-50.
17. Park MS, Klotz E, Kim MJ, Song SY, Park SW, Cha SW, et al.
Perfusion CT: Noninvasive surrogate marker for stratification of
pancreatic cancer response to concurrent chemo-and radiation
therapy. Radiology 2009;250:110-7.
18. Patel SV, Spencer JA, El-Hasani S, Sheridan MB. Imaging of
Pancreatic Trauma: Pictoral Review. Br J Radiol 1998;71:985-90.
19. Pavone P, Laghi A, Catalano C, Panebianco V, Pediconi F, Fabiano
S, et al. MR imaging of pancreatic neoplasms. Tumori 1999;85(1
Suppl 1):S6-10.
20. Matos C, Cappeliez O, Winant C, Coppens E, Deviere J, Metens
T. MR imaging of the pancreas: A pictorial tour. RadioGraphics
2002;22:e2.
21. Fung HS, Lau S, Tse KS, Wai JW, Wong WK, Tang KW, et al. The
Role of Chemical Shift Magnetic Resonance Imaging in the Diagnosis
of Focal Fatty Replacement of the Pancreas. J Hong Kong Coll Radiol
2009;11:176-8.
22. Wang Y, Chen ZE, Yaghmai V, Nikolaidis P, McCarthy RJ, Merrick
L, et al. Diffusion-weighted MR imaging in pancreatic endocrine
tumors correlated with histopathologic characteristics. J Magn Reson
Imaging 2011;33:1071-9.
23. Macari M, Finn ME, Bennett GL, Cho KC, Newman E, Hajdu CH,
et al. differentiating pancreatic cystic neoplasms from pancreatic
pseudocysts at MR imaging: Value of perceived internal debries.
Radiology 2009;251:77-84.
24. Cho SG, Lee DH, Lee KY, Ji H, Lee KH, Ros PR, et al. Differentiation
of chronic focal pancreatitis from pancreatic carcinoma by in vivo
proton magnetic resonance spectroscopy. J Comput Assist Tomogr
2005;29:163-9.
25. Fattahi R, Modanlou KA, Bieneman BK, Soydan N, Balci NC, Burton
FR. Magnetic resonance imaging in pancreatic transplantation. Top
Magn Reson Imaging 2009;20:49-55.
26. Varadarajulu S, Wallace MB. Application of endoscopic
ultrsonography in pancreatic cancer. Cancer Control 2004;11:15-22.
27. Long EE, Van Dam J, Weinstein S, Jeffrey B, Desser T, Norton JA.
Computed tomography, endoscopic, laproscopic, and intra-operative
sonography for assessing respectability of pancreatic cancer. Surg
Oncol 2005;14:105-13.
28. Rosch T, Lightdale CJ, Botet JF, Boyce GA, Sivak MV Jr, Yasuda
K, et al. Localization of pancreatic endocrine tumors by endoscopic
ultrasonography. N Engl J Med 1992;326:1721-6.
29. Ozawa Y, Numata K, Tanaka K, Ueno N, Kiba T, Hara K, et al.
Contrast-enhanced sonography of small pancreatic mass lesions. J
Ultrasound Med 2002;21:983-91.
30. Sahani DV, Kadavigere R, Saokar A, Fernandez-del Castillo C,
Brugge WR, Hahn PF. Cystic pancreatic lesions: A simple imaging
based classification system for guiding management. RadioGraphics
2005;25:1471-84.
31. Gunaratnam NT, Sarma AV, Norton ID, Wiersema MJ. A prospective
study of EUS-guided celiac plexus neurolysis for pancreatic cancer
pain. Gastrointest Endosc 2001;54:316-24.
32. Janssen J. (E) US elastrography: Current status and perspectives. Z
Gastroenterol 2008;46:572-9.
33. Delbeke D, Rose DM, Chapman WC, Pinson CW, Wright JK,
Beauchamp RD, et al. Optimal interpretation of FDG-PET in the
diagnosis, staging and management of pancreatic carcinoma. J Nucl
Med 1999;40:1784-91.
34. Bares R, Klever P, Hauptmann S, Hellwiq D, Fass J, Cremerius U,
et al. F-18 Florodeoxyglucose PET in vivo evaluation of pancreatic
glucose metabolism for detection of pancreatic cancer. Radiology
1994;192:79-86.
35. Zimny M, Fass J, Bares R, Crèmerius U, Sabri O, Buechin P, et al.
Florodeoxyglucose positron emission tomography and the prognosis
of pancreatic carcinoma. Scand J Gastroenterol 2000;35:883-8.
36. Franke C, Klapdor R, Meyerhoff K, Schauman M. 18-FDG positron
emission tomography of the pancreas: diagnostic benefit in the
follow-up of pancreatic carcinoma. Anticancer Res 1999;19:2437-42.
37. Higashi T, Sakahara H, Torizuka T, Nakomoto Y, Kanamori S,
Hiraoka M, et al. Evaluation of intraoperative radiation therapy
for unresectable pancreatic cancer with FDG-PET. J Nucl Med
1999;40:1424-33.
38. Ahlstrom H, Eriksson B, Bergstrom M, Bjurling P, Langstrom B,
Oberg K, et al. Pancreatic neuroendocrine tumors: Diagnosis with
PET. Radiology 1995;195:333-7.
39. Sperti C, Pasquali C, Chierichetti F, Liessi G, Ferlin G, Pedrazzzoli
S. Value of 18-fluorodeoxyglucose positron emission tomography
in the management of patients with cystic tumors of the pancreas.
Ann Surg 2001;234:675-80.
40. Pfannenberg AC, Eschmann SM, Horger M, Lamberts R, Vonthein
R, Claussen CD, et al. Benefit of anatomical-functional image fusion
in the diagnostic work-up of neuroendocrine neoplasms. Eur J Nucl
Med Mol Imaging 2003;30:835-43.
41. Testoni PA, Mangiavillano B. Optical coherence tomography for
bile and pancreatic duct imaging. Gastrointest Endosc Clin N Am
2009;19:637-53.
Cite this article as: Chaudhary V, Bano S. Imaging of the pancreas: Recent
advances. Indian J Endocr Metab 2011;15:S25-32.
Source of Support: Nil, Conict of Interest: Nil.
Chaudhary and Bano: Pancreatic imaging
[Downloaded free from http://www.ijem.in on Wednesday, September 28, 2016, IP: 83.84.166.247]
... The size and location of the pancreas is the variety from person to person [1]. Therefore, the detection of the pancreas in computed tomography (CT) and magnetic resonance imaging (MRI) modality requires time and effort [2]. Pancreatic cancer is the riskiest type of cancer after lung cancer and bronchial cancer. ...
... TP+TN TP+FP+TN+FN (1) Precision= TP TP+FP (2) Sensitivity= TP TP+FN (3) Specificity= TN TN+FP (4) Dice= 2*TP 2*TP+FP+FN (5) IoU= TP TP+FP+FN (6) True Positive (TP), True Negative (TN), False Positive (FP), and False Negative (FN) refer to the number of correctly classified positive classes, the number of correctly classified negative classes, the number of incorrectly classified positive classes, and the number of the incorrectly classified negative class, respectively. In this context, classes indicate pixel-based classes. ...
Article
Full-text available
The pancreas is one of the vital organs in the human body. Early diagnosis of a disease in the pancreas is critical. In this way, the effects of pancreas diseases, especially pancreatic cancer on the person are decreased. With this purpose, artificial intelligence-assisted pancreatic cancer segmentation was performed for early diagnosis in this paper. For this aim, several state-of-the-art segmentation networks, UNet, LinkNet, SegNet, SQ-Net, DABNet, EDANet, and ESNet were used in this study. In the comparative analysis, the best segmentation performance has been achieved by SQ-Net. SQ-Net has achieved a 0.917 dice score, 0.847 IoU score, 0.920 sensitivity, 1.000 specificity, 0.914 precision, and 0.999 accuracy. Considering these results, an artificial intelligence-based decision support system was created in the study.
... Most individuals with pancreatitis (chronic), pancreatic cancer, or other pancreatic disorders have poor prognoses. Pancreatic imaging advancements may make it simpler to detect these diseases when they 2 are still treatable at an earlier stage. Imaging the pancreas is crucial for detecting lesions comprising malignancy or pre-malignancy and identifying the symptom origins. ...
Article
Introduction: MDCT is found to be superior among all the imaging modalities for detecting pathological lesions but the correlation studies with surgical or histopathological ndings regarding its effectiveness in the characterization of pancreatic lesions are limited. Aims and objectives: The aim of the present prospective investigation is to determine the effectiveness of MDCT in the evaluation and characterization of cystic lesions in the pancreas in patients undergoing CT abdomen who was established to possess cystic lesions in the pancreas by comparing the MDCT results with transabdominal and FNAC. The prospective research was carried out in the department of radiolo Materials and methods: gy, and the patients (n=50) who had been referred to the radiology department with abdominal pain for MDCT abdomen, aged 25 years and above, from March 2021 to July 2022 (1 year and 6-month period duration) and who were established to have pancreatic cysts under the conditions of below-stated inclusion and exclusion criteria. Transabdominal ultrasound ndings, MDCT results showing the location of the cystic lesions, vascular invasions, distant metastasis, and FNAC ndings, along with the surgical management procedure, were collected and entered into an excel spreadsheet and analysed using SPSS version 25. Results: The total study population's mean age was determined to be 45.1±12.0 years (ranging from 26 to 71 years) and was found to be male dominant (62%). The overall true positives, true negatives, and false positives observed with transabdominal ultrasound were estimated to be 36%, 28%, and 36% individually and in the case of MDCT, there were nil results with true negative and false negative when compared with FNAC results. MDCT revealed true positive cases were 70% and the false positive case were 30% while 36% and 64% individually. The existing study established that MDCT depicted the ndings of lesions involved Conclusion: with pancreatic cysts were almost matched with the end outcome results of FNAC in 70% of the cases than transabdominal ultrasound which indicated the fact that MDCT is identied to be superior to transabdominal ultrasound tools in detecting the lesions of pancreatic cysts.
... In MRI and computed tomography, less than 100 µm of spatial resolution could be used to delineate the pancreas [167]. This method, despite the fact that the pancreas is an extremely vascularized soft organ and the islets of Langerhans only constitute 2 to 3% of the pancreas' tissues, makes it virtually impossible to distinguish the islets from other pancreatic tissues due to their high vascularization level. ...
Article
Diabetes poses major economic, social, and public health challenges in all countries worldwide. Besides cardiovascular disease and microangiopathy, diabetes is a leading cause of foot ulcers and lower limb amputations. With the continued rise of diabetes prevalence, it is expected that the future burden of diabetes complications, early mortality, and disabilities will increase. The diabetes epidemic is partly caused by the current lack of clinical imaging diagnostic tools, the timely monitoring of insulin secretion and insulin-expressing cell mass (beta (β)-cells), and the lack of patients' adherence to treatment, because some drugs are not tolerated or invasively administrated. In addition to this, there is a lack of efficient topical treatment capable of stopping the progression of disabilities, in particular for treating foot ulcers. In this context, polymer-based nanostructures garnered significant interest due to their tunable physicochemical characteristics, rich diversity, and biocompatibility. This review article emphasizes the last advances and discusses the prospects in the use of polymeric materials as nanocarriers for β-cell imaging and non-invasive drug delivery of insulin and antidiabetic drugs in the management of blood glucose and foot ulcers.
... However, potential risk indicator of DM is associated with obesity and severe pancreas alteration that can be detected on the patient through imaging and diagnostic history [28,29]. The volumetric evaluation of the pancreatic changes that occur in DM is assessed with MRI CT scan radiological modalities [30]. It was reported that pancreas volume decreases with DM duration in the patient with CT scan modality evaluation [31]. ...
Article
Full-text available
Diabetes has become a health care problem of increasing concern in the world as number of individuals with disease are on the rise over the past 2 decades. Imaging technologies are noninvasive tools which are currently receiving an increase significance to guiding physician in finding and therapeutic treatment of patients. Advanced imaging has upgraded our ability to assess the prospect of diabetics in patients. However, the capacity of imaging to detect the existence of osteomyelitis and myotitis does not automatically mean that it should be employ on a regular basis for tentative diagnosis of the diabetics. Diabetes is a multisystemic disease that exerts its effects through decrease insulin sensitivity causing various stages of metablilic conditions. The Pathogenesis of the complication related to diabetes is solely depended to duration of the disease with other health challenges which are refer to co-existing factors. The effects of long-standing effect of diabetes are usually associated with microvascular disease resulting to diabetic retinopathy nephropathy and cardiomyopathy. However, imaging performs an important part in diagnosis and follow-up of associated complications. This paper reviews the pathophysiology of diabetics, discuss the imaging appearance and provide detail review of the diagnostic image profile of patient with diabetics
... The size and location of the pancreas is the variety from person to person [1]. Therefore, the detection of the pancreas in computed tomography (CT) and magnetic resonance imaging (MRI) modality requires time and effort [2]. Pancreatic cancer is the riskiest type of cancer after lung cancer and bronchial cancer. ...
Preprint
Full-text available
The pancreas is one of the vital organs in the human body. Early diagnosis of a disease in the pancreas is critical. In this way, the effects of pancreas diseases, especially pancreatic cancer on the person are decreased. With this purpose, artificial intelligence-assisted pancreatic cancer segmentation was performed for early diagnosis in this paper. For this aim, several state-of-the-art segmentation networks, UNet, LinkNet, SegNet, SQ-Net, DABNet, EDANet, and ESNet were used in this study. In the comparative analysis, the best segmentation performance has been achieved by SQ-Net. SQ-Net has achieved a 0.917 dice score, 0.847 IoU score, 0.920 sensitivity, 1.000 specificity, 0.914 precision, and 0.999 accuracy. Considering these results, an artificial intelligence-based decision support system was created in the study.
... Computed Tomography (CT) and Magnetic Resonance Imaging (MRI) modalities are generally used to diagnose pancreatic diseases (Chaudhary & Bano, 2011). The CT modality is frequently used for determination of pancreatic diseases. ...
Article
Full-text available
The pancreas is one of the small size organs in the abdomen. Moreover, anatomical differences make it difficult to detect the pancreas. This project aims to automatically segmentation of pancreas. For this purpose, NIH-CT82 data set, which includes CT images from 82 patients was used. U-Net which is state-of-the-art model and its different versions, namely Attention U-Net, Residual U-Net, Attention Residual U-Net, and Residual U-Net++ were tested. Best predict performance was achieved by Residual U-Net with the dice of 0.903, IoU of 0.823, sensitivity of 0.898, specificity of 1.000, precision of 0.908, and accuracy of 0.999. Consequently, an artificial intelligence (AI) supported decision support system was created for pancreas segmentation.
... Pancreatic diseases can be summarized as malignant and benign neoplasms of the pancreas, acute or chronic inflammatory processes, involvement of systemic diseases, diabetes mellitus characterized by deterioration of pancreatic endocrine functions, and cystic fibrosis characterized by deterioration of exocrine functions [1]. Although transabdominal ultrasound is used for imaging the pancreas because it is a cheap and easily accessible method, computed tomography, magnetic resonance imaging, and endoscopic ultrasound are more preferred for pancreatic imaging due to their high resolution [2]. ...
Article
Full-text available
Purpose The use of ultrasound (US) elastography to assess the pancreas is subject to serious limitations, and it is not easy to perform US elastography for the pancreas with satisfactory and reproducible accuracy. The aim of our study was to show the normal pancreatic stiffness values with shear wave elastography (SWE) and to evaluate the pancreatic tissue stiffness at two different respiratory phases. Methods Sixty-three subjects were included in the study. Median values of pancreatic stiffness were recorded. Values during deep inspiration and free breathing were compared. Results The stiffness values of the pancreatic tissue measured during deep inspiration breath holds and during free breathing were 5.70 ± 1.74 kPa and 4.15 ± 1.70 kPa, respectively. It was found that the values of pancreatic tissue stiffness measured during deep inspiration were statistically significantly higher than those measured during physiological breathing (p < 0.001). Conclusions Because of the reference value differences between different ultrasound elastography devices, it is important to know the reference ranges of normal pancreatic stiffness according to the device, in order to distinguish possible pathologies. In addition, the variability of pancreatic stiffness measurements with respiratory phases should be considered in clinical applications. Graphical abstract
... [7,8] The diagnosis of pancreatic diseases generally requires the combined use of different imaging modalities, allowing a more complex evaluation of the gland and adjacent soft tissues. [9] Congenital anomalies and normal variants of the pancreas can present a diagnostic challenge when encountered during diagnostic procedures. [10,11] Moreover, they are often incidentally detected due to the increasing accessibility of diagnostic imaging and some may present with symptoms. ...
Article
Full-text available
Introduction: Anomalies and variants of development of the pancreas are relatively frequent. These variations can often lead to misdiagnosis and unnecessary medical procedures. Although, pancreatic diseases are a constantly researched field, fundamental research is relatively understudied and re-evaluation of the pancreatic morphology is performed rarely. Aim: The goal of the current study was to analyse pancreatic surface morphology by means of macroscopic anatomical dissection of 78 organ complex. Material and methods: The anatomy of the pancreas was studied by means of macroscopic anatomical dissection. The study was performed on 78 organ complexes (the pancreas, spleen, and duodenum with surrounding abdominal fat) donated to the department of human anatomy from patients who died of causes not related to pancreatic diseases. The organs were fixed in a 10% formalin solution separately to accurately preserve their forms. The organs were measured by a caliper. Results: The lie of the pancreas was classified according to Kreel and co-workers into 6 types: oblique shape in 11.54% of cases, sigmoid – 30.77%, transverse – 25.64%, horseshoe – 11.54%, L-shaped – 14.1%, and inverted V shape in 5.13%. Moreover, there were two possible L-shapes, the classical (8.97%) and inverted L-shape (5.13%). In one case, the pancreas had an unusual M-shape (1.28%). One specimen was a case of short pancreas (1.28%). Surface clefts were encountered in 17.95% of cases and in 5.13% of these cases, there was a branch of the splenic artery. Conclusions: The current study demonstrates variations in the lie of the pancreas as well as several new possible variants. Moreover, we propose a modified classification based on these findings. Therefore, the pancreas can have an oblique, sigmoid, transverse, horseshoe, L shape, inverted L, inverted V, and M shaped lie. Pancreatic clefts are another frequently encountered variation that should be kept in mind. The size of the pancreas alone is variable and should be analyzed together with its structure to avoid possible misinterpretation.
... Since the introduction of computerized tomography (CT) scan in late 1970s, there has been a drastic improvement in the pancreatic imaging. 1 The conventional CT technology has been reformed to unleash new technologies such as spiral CT and multi-detector CT (MDCT). The spiral CT has been designed to overcome the limitations of the conventional CT requiring the connection between the power cables and CT tube. 2 Spiral CT is quicker and more reliable than conventional CT. ...
Article
Full-text available
The pancreas is an important exocrine and endocrine gland in the human body located in the upper abdomen. A great deal of information about the pancreas can be obtained on multi-detector computerized tomography (MDCT), including the exact location of the lesion, characterization and relation to the surrounding structures. The present study was done to evaluate the spectrum of pathologies of pancreas visualized on MDCT. A cross-sectional single center study was conducted from November 2018 to January 2020. Patients who were diagnosed with pancreatic pathology of all etiologies and satisfying the inclusion and exclusion criteria were invited to participate in the study. CT examination of the abdomen was typically performed using neutral oral contrast and non-ionic low osmolar iodinated intravenous contrast agent. Abdominal CT images were evaluated as per the standard reporting pattern and the images of pancreas were analyzed. In our study out of 33 patients, 25 patients were male and eight were female patients. Most of the patients belonged to the age group of 40-50 years. Among the various lesions diagnosed on MDCT inflammatory lesions were most common accounting for 60.6% of the cases, followed by tumors (33.3%), and congenital lesions (6.1%). MDCT is a very useful investigation to diagnose various pancreatic pathologies. Predominant pathologies diagnosed were inflammatory lesions (pancreatitis) followed by neoplasms.
Chapter
Pancreatic cancer remains one of the most lethal malignancies in the world. Most patients diagnosed with pancreatic cancer present at an advanced stage of the disease. Therefore, early diagnosis and accurate staging are essential in the management of these patients. Cross-sectional imaging techniques and interventional endoscopies such as endoscopic ultrasound (EUS) and endoscopic retrograde cholangiopancreatography (ERCP) play an important role in the initial evaluation, staging, and treatment of pancreatic lesions. Progressive biliary obstruction is often expected in pancreatic cancer, which could result in cholangitis, cholestasis, pruritus, liver failure, and premature mortality. Endoscopy-guided biliary decompression (ERCP and EUS) in resectable and unresectable tumors has gained more ground by replacing percutaneous drainage and surgery as the first approach. The main advantage of EUS over ERCP sampling is the possibility of obtaining a tissue diagnosis by performing fine needle aspiration biopsy (EUS-FNA), however, ERCP-guided biliary sampling could be performed first if a concurrent biliary decompression is required. Contrast-enhanced EUS and elastography are ancillary techniques that could be useful in clinical practice. Moreover, there are emerging EUS-guided therapeutic techniques that play an essential role in the management of pancreatic cancer.KeywordsPancreatic cancerEndoscopic ultrasoundEndoscopic ultrasound-guide fine needle aspirationEndoscopic retrograde cholangiopancreatographyBiliary drainage
Article
Full-text available
Background Accurate staging of pancreatic cancer is essential for surgical planning and for identification of locally advanced and metastatic disease that is incurable by surgery. Advances in endoscopic sonography (EUS), computed tomography (CT), and positron emission tomography have improved the accuracy of staging and reduced the number of incomplete surgical resections. Tissue acquisition is necessary in nonsurgical cases when chemoradiotherapy is considered. The complex regional anatomy of the pancreas makes cytologic diagnosis of malignancy at this region difficult without exploratory surgery. Although CT-guided fine-needle aspiration (FNA) is used for this purpose, reports of an increased risk of peritoneal dissemination of cancer cells and a false-negative rate of nearly 20% make this a poor choice. The ability to position the EUS-transducer in direct proximity to the pancreas by means of the stomach and duodenum, combined with the use of FNA, increases the specificity of EUS in detecting pancreatic malignancies. Methods The current literature regarding the accuracy of EUS with FNA in the evaluation of pancreatic cancer is reviewed. Results EUS accuracy ranges from 78% to 94% for tumor staging and from 64% to 82% for nodal staging. EUS also enables FNA of lesions that are too small to be identified by CT or MRI or too well encased by surrounding vascular structures to safely allow percutaneous biopsy. The accuracy for detecting invasion into the superior mesenteric artery and vein is lower than that for detecting portal or splenic vein invasion, especially for large tumors. EUS permits delivery of localized therapy such as celiac plexus neurolysis for pain control and direct intra-lesional injection of antitumor therapy. Conclusions EUS in combination with FNA is a highly accurate method of preoperative staging of pancreatic cancer, especially those too small to be characterized by CT or MRI, and it has the ability to obtain cytological confirmation of pancreatic cancer.
Article
Full-text available
Total fat replacement of the pancreas is rare. Focal fatty replacement is the most common degenerative lesion of pancreas. Focal fatty deposits have no major clinical significance; however, extreme fat replacement is of pathologic significance, as it is associated with marked reduction in exocrine function of pancreas, resulting in malabsorption due to pancreatic enzyme insufficiency.
Article
Full-text available
We performed perfusion computed tomography (P-CT) and angiography of the pancreas in patients with severe acute pancreatitis (SAP) and compared the usefulness of these two methods in predicting the development of pancreatic necrosis. We compared P-CT and angiography results taken within 3 days after symptom onset in 21 SAP patients. We divided the pancreas into three areas, the head, body, and tail, and examined each area for perfusion defects (via P-CT) and arterial vasospasms (by angiography). Three weeks later, all patients underwent contrast-enhanced CT to determine whether pancreatic necrosis had developed. Of the 21 SAP patients, 16 exhibited perfusion defects, while 17 proved positive for vasospasms in at least one area. Fourteen patients developed pancreatic necrosis. Of the 63 pancreatic areas from the 21 SAP patients, perfusion defects appeared in 25 areas (39.7%), 24 of which showed vasospasms (96.0%). Angiography showed 33 areas with vasospasms (52.4%), of which 24 showed perfusion defects (72.7%). Of the 25 areas with perfusion defects, 21 developed pancreatic necrosis (84.0%). Of the 33 areas with vasospasms, 21 developed necrosis (63.6%). Pancreatic necrosis developed only in the areas positive both for perfusion defects and for vasospasms. No areas without perfusion defect or vasospasms developed pancreatic necrosis. P-CT predicted the development of pancreatic necrosis with significantly higher accuracy than angiography. While both P-CT and angiography are useful in predicting the development of pancreatic necrosis in patients with SAP, P-CT appears to be more accurate for this purpose.
Article
Focal fatty replacement of the pancreas is a rare benign disease that has been associated with obesity, diabetes mellitus, chronic pancreatitis, hereditary pancreatitis, and obstruction of the pancreatic duct. This report of a 48-year-old woman with focal fatty replacement of the pancreas illustrates the typical imaging findings on chemical shift magnetic resonance imaging. The differential diagnosis and a review of literature are discussed.
Article
To prospectively determine whether perfusion computed tomography (CT) parameters, such as volume transfer constant (K(trans)) between blood plasma and extracellular extravascular space (EES) and blood volume calculated from dynamic CT data, can be used to predict response of pancreatic cancer to concurrent chemotherapy and radiation therapy (CCRT). This prospective study was institutional review board approved, and written informed consent was obtained. Thirty patients with pancreatic cancer underwent perfusion CT with 64-detector row CT before gemcitabine-based CCRT. Two perfusion parameters (K(trans) and blood volume) measured before treatment were compared between patients who responded to treatment and those who did not, as determined with World Health Organization criteria from first and second posttherapeutic follow-up CT examinations, which were performed at 3- and 6-month follow-up. Statistical analysis was performed with the two-sample t test. A receiver operating characteristic curve was used to determine the best cutoff value of perfusion parameters for differentiation of responders from nonresponders. Twenty of 30 patients examined at 3-month follow-up responded to therapy. Their pretreatment K(trans) value was significantly higher than that of nonresponders (50.8 mL/100 mL/min +/- 30.5 [standard deviation] vs 19.0 mL/100 mL/min +/- 10.8, P = .001). The best cutoff value for differentiating between responders and nonresponders was 31.8 mL/100 mL/min, which yielded 75.0% sensitivity and 90.0% specificity. Ten of 18 patients examined at 6-month follow-up responded to therapy. Their pretreatment K(trans) value was significantly higher than that of nonresponders (58.6 mL/100 mL/min +/- 43.2 vs 19.8 mL/100 mL/min +/- 10.9, P = .002). Responders also had higher blood volume values, but this difference was not significant. Tumors with a high pretreatment K(trans) value tended to respond better to CCRT than did tumors with a low pretreatment K(trans) value. Perfusion CT may be used to predict tumor response to CCRT in patients with pancreatic cancer. This might aid in development of a tailored approach to therapy in these patients.
Article
To retrospectively assess apparent diffusion coefficients (ADCs) of different subtypes of pancreatic endocrine tumors based on the World Health Organization (WHO) classification system and analyze the potentially responsible histopathologic characteristics. Following Institutional Review Board (IRB) approval, 18 patients with surgical pathology-proven pancreatic endocrine tumors were evaluated. Tumors were subcategorized based on the WHO grading classification into well-differentiated tumors with benign and uncertain behavior and endocrine carcinomas with well and poor differentiation. ADCs were measured on diffusion-weighted (DW) images and compared using Student's t-test and one-way analysis of variance. The correlation between ADCs, tumor cellularity, Ki-67 labeling index (an index of cell growth), and extracellular fibrosis were analyzed. A difference was demonstrated in mean ADCs between well-differentiated endocrine tumors (1.75 ± 0.53) and endocrine carcinomas (1.00 ± 0.19 × 10(-3) mm(2) /sec) (P < 0.01). After excluding the three well-differentiated endocrine tumors with benign behavior and marked fibrosis, a significant inverse correlation between ADC values and cellularity of endocrine tumors was observed. An inverse correlation was seen between Ki-67 labeling index and ADC values (r = -0.70; P < 0.01). Tumor cellularity and/or extracellular fibrosis may account for various ADCs in pancreatic endocrine tumors. ADC correlates well with the Ki-67 labeling index and may help predict growth of endocrine tumors.
Article
To prospectively compare 320-detector volumetric and 64-detector helical computed tomographic (CT) images of the pancreas for depiction of anatomic structures, image noise, and radiation exposure. This study was approved by the institutional review board, and written informed consent was obtained. A total of 154 patients (85 men, 69 women; age range, 26-85 years; mean age, 67 years) who underwent biphasic (arterial and pancreatic phase) contrast material-enhanced CT performed with a 320-detector scanner were randomized into two groups: the 320-detector group and the 64-detector group. Biphasic transaxial multiplanar reformatted images and volume-rendered CT angiograms were obtained. CT numbers in the abdominal aorta, pancreas, and abdominal wall fat tissue; signal-to-noise ratio (SNR); and dose-length product (DLP) were compared. In addition, image quality and focal lesion depiction (n = 35) were qualitatively determined in the two groups. Unpaired t and Mann-Whitney tests were used for quantitative and qualitative assessment, respectively. No significant difference in CT numbers of the abdominal aorta and pancreas was noted between the two groups. Mean DLP was 43% lower in the 320-detector group (675.4 mGy·cm) than in the 64-detector group (1187.8 mGy·cm) (P < .001). SNR of the abdominal aorta, pancreas, and abdominal wall fat on biphasic images was significantly lower in the 320-detector group than in the 64-detector group (P < .001). Image quality was acceptable in both groups and was slightly better in the 64-detector group for pancreatic phase axial images (P = .02) and arterial phase multiplanar reformatted images (P < .01). No significant difference was found in the depiction of pancreatic parenchyma, main pancreatic duct, focal pancreatic lesions, splanchnic arteries, or most of the small splanchnic arterial branches. A 320-detector CT scan facilitates fast volumetric contrast-enhanced CT of the entire pancreas with acceptable image quality, even though SNR was significantly lower at 320-detector volumetric scanning.
Article
To investigate 64 computed tomography (CT) perfusion imaging features of patients with pancreatic cancer and mass-forming chronic pancreatitis. Between January 2003 and April 2010, 234 patients with pancreatic mass underwent 64-CT perfusion imaging. Among them, the histopathological results of 64 patients were proven to be pancreatic adenocarcinoma and 15 patients were proven to be mass-forming chronic pancreatitis. Additionally, CT perfusion imaging was performed in 33 healthy volunteers served as controls. The slice data were processed using CT perfusion software. Perfusion parameters including time density curve, blood flow, blood volume, permeability, peak enhancement, and time to peak were recorded. Blood flow was 77% lower in patients with pancreatic adenocarcinoma than in controls, 48% lower in patients with mass-forming chronic pancreatitis than in controls, and 56% lower in patients with pancreatic adenocarcinoma than with mass-forming chronic pancreatitis (P < .016). Blood volume was 65% lower in pancreatic adenocarcinoma than in controls, 27% lower in mass-forming chronic pancreatitis than in controls, and 53% lower in cancer than mass-forming chronic pancreatitis (P < .016). Permeability was 559% higher in pancreatic adenocarcinoma than in controls, 821% higher in mass-forming chronic pancreatitis than in controls, and 28% lower in cancer than mass-forming chronic pancreatitis (P < .016). Peak enhancement was 27% lower and time to peak 23% longer in pancreatic adenocarcinoma than mass-forming chronic pancreatitis (P < .016). Time-density curve showed the peak of mass-forming chronic pancreatitis is earlier and higher than that of pancreatic adenocarcinoma, and the peak of mass-forming chronic pancreatitis is later and lower than that of controls. CT perfusion imaging can provide additional quantitative hemodynamic information of pancreatic adenocarcinoma and mass-forming chronic pancreatitis.
Article
Optical coherence tomography (OCT) is an optical imaging modality introduced in 1991 that performs high-resolution, cross-sectional, subsurface tomographic imaging of the microstructure in materials and biologic systems by measuring backscattered or backreflected infrared light. OCT has been used for biomedical applications where many factors affect the feasibility and effectiveness of any imaging technique. The highly scattering and absorbing living tissues greatly limit the application of optical imaging modalities. In the last decade, OCT technology has evolved from an experimental laboratory tool to a new diagnostic imaging modality with a wide spectrum of clinical applications in medical practice, including the gastrointestinal (GI) tract and pancreaticobiliary ductal system.
Article
Magnetic resonance imaging (MRI) plays an important role in the evaluation of pancreas transplantation. Standard MRI, magnetic resonance angiography, and MR cholangiopancreatography can demonstrate the changes of the anatomy after transplantation. Vascular complications are assessed by MR angiography. Magnetic resonance cholangiopancreatography reveals ductal changes resulting from acute and/or chronic rejection and determines leaks with the use of a secretin-stimulated MR cholangiopancreatography. Serial contrast-enhanced MRI may detect the diminished perfusion that is related to the graft rejection or vascular complications. In this paper, we reviewed types of pancreas transplantation procedures, complications that arise in a short and/or a long term after the transplantation, and their assessment by MRI.