ArticlePDF AvailableLiterature Review

Peritoneal carcinomatosis: Limits of diagnosis and the case for liquid biopsy

Authors:

Abstract and Figures

Peritoneal Carcinomatosis (PC) is a late stage manifestation of several gastrointestinal malignancies including appendiceal, colorectal, and gastric cancer. In PC, tumors metastasize to and deposit on the peritoneal surface and often leave patients with only palliative treatment options. For colorectal PC, median survival is approximately five months, and palliative systemic therapy is able to extend this to approximately 12 months. However, cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (CRS/HIPEC) with a curative intent is possible in some patients with limited tumor burden. In well-selected patients undergoing complete cytoreduction, median survival has been reported as high as 63 month. Identifying patients earlier who are either at risk for, or who have recently developed PC may provide them with additional treatment options such as CRS/HIPEC. PC is diagnosed late by imaging findings or often times during an invasive procedures such as laparoscopy or laparotomy. In order to improve the outcomes of PC patients, a minimally invasive, accurate, and specific PC screening method needs to be developed. By utilizing circulating PC biomarkers in the serum of patients, a " liquid biopsy, " may be able to be generated to allow a tailored treatment plan and early intervention. Exosomes, stable patient-derived nanovesicles present in blood, urine, and many other bodily fluids, show promise as a tool for the evaluation of labile biomarkers. If liquid biopsies can be perfected in PC, manifestations of this cancer may be more effectively treated, thus offering improved survival.
Content may be subject to copyright.
Oncotarget1
www.impactjournals.com/oncotarget
www.impactjournals.com/oncotarget/ Oncotarget, Advance Publications 2017
Peritoneal carcinomatosis: limits of diagnosis and the case for
liquid biopsy
James R.W. McMullen1, Matthew Selleck2, Nathan R. Wall1 and Maheswari Senthil2
1 Department of Basic Sciences, Center for Health Disparities & Molecular Medicine, Division of Biochemistry, Loma Linda
University Medical Center, Loma Linda, CA, USA
2 Department of Surgery, Division of Surgical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
Correspondence to: Nathan R. Wall, email: nwall@llu.edu
Correspondence to: Maheswari Senthil, email: msenthil@llu.edu
Keywords: peritoneal carcinomatosis, liquid biopsy, biomarker, exosomes
Received: January 21, 2017 Accepted: March 15, 2017 Published: March 22, 2017
ABSTRACT
Peritoneal Carcinomatosis (PC) is a late stage manifestation of several
gastrointestinal malignancies including appendiceal, colorectal, and gastric cancer.
In PC, tumors metastasize to and deposit on the peritoneal surface and often leave
patients with only palliative treatment options. For colorectal PC, median survival
is approximately ve months, and palliative systemic therapy is able to extend this
to approximately 12 months. However, cytoreductive surgery with hyperthermic
intraperitoneal chemotherapy (CRS/HIPEC) with a curative intent is possible in
some patients with limited tumor burden. In well-selected patients undergoing
complete cytoreduction, median survival has been reported as high as 63 month.
Identifying patients earlier who are either at risk for, or who have recently developed
PC may provide them with additional treatment options such as CRS/HIPEC. PC is
diagnosed late by imaging ndings or often times during an invasive procedures such
as laparoscopy or laparotomy. In order to improve the outcomes of PC patients, a
minimally invasive, accurate, and specic PC screening method needs to be developed.
By utilizing circulating PC biomarkers in the serum of patients, a “liquid biopsy,” may
be able to be generated to allow a tailored treatment plan and early intervention.
Exosomes, stable patient-derived nanovesicles present in blood, urine, and many
other bodily uids, show promise as a tool for the evaluation of labile biomarkers.
If liquid biopsies can be perfected in PC, manifestations of this cancer may be more
eectively treated, thus oering improved survival.
INTRODUCTION
Peritoneal Carcinomatosis (PC) is a late stage
manifestation of several gastrointestinal malignancies
characterized by tumor deposition across the peritoneal
surface [1]. This can be entirely asymptomatic in its early
stages, or as the disease progresses, symptoms such as
nausea, diarrhea, abdominal pain, bloating, and weight
loss may develop [1]. The disease is often discovered
when ascites or intestinal obstruction develop, occurring
generally with greater tumor burden which is more
dicult to treat [2]. Early PC detection when there is
limited tumor burden may increase the eectiveness of
current treatment options [3].
Colorectal cancer (CRC), the third most common
cancer in the world, provides a good case study of PC. In
2016 there will be an estimated 95,270 cases of colorectal
cancer in the United States and nearly 1.4 million cases
worldwide [4]. Synchronous PC is diagnosed around the
time of diagnosis of primary tumor while metachronous
PC is diagnosed at a later time, typically months to
years after the original diagnosis [5]. The incidence
of synchronous isolated peritoneal carcinomatosis in
patients with CRC varies somewhat in the literature from
4%-18% [6-9]. This may even be a low estimate given
the lack of sensitivity of imaging for PC and that it may
not be discovered until surgical exploration. Meanwhile,
metachronous PC has been reported in 5-19% of patients
following denitive treatment.
Oncotarget2
www.impactjournals.com/oncotarget
CYTOREDUCTIVE SURGERY (CRS)
PC used to be considered a lethal disease with
no curative surgical options. However, the growing
acceptance of CRS with hyperthermic intraperitoneal
chemotherapy (HIPEC), has oered the possibility of
improved survival for carefully selected patients [3].
This method makes use of “cytoreduction” to surgically
remove gross visible tumor deposits followed by direct
contact of heated cytotoxic chemotherapy agents to
aect any residual disease. Given during surgery, this
protocol maximizes potential contact with the peritoneal
surface while minimizing systemic toxicity. Specically,
hyperthermia between 41 and 43 degrees centigrade is
combined with large molecular weight drugs that penetrate
between a few cells deep to 3mm causing cytotoxic eects
[10]. Dr. Paul Sugarbaker is credited with developing this
treatment option by combing these elements into a curative
approach to peritoneal dissemination of gastrointestinal
malignancies [11, 12]. Median survival of CRC PC
without any treatment is approximately 4-7 months, while
palliative systemic therapy may extend this to 12-23
months based on several series [13-15]. Median overall
survival with CRS/HIPEC has been reported to range
from 22 to 63 months with a 5-year survival of 40-51%
in selected patients [13, 15, 16]. The outcomes of CRS/
HIPEC are strongly inuenced by careful patient selection
and complete cytoreduction (CC-0) (see Table 1) [17].
Survival of patients with colorectal cancer who receive
less than complete cytoreduction (CC-1 or CC-2) or have
a higher burden of disease as indicated by the peritoneal
carcinomatosis index (PCI) (see Figure 1) is signicantly
diminished compared to that of a CC-0 resection [17-19].
Extensive disease burden at identication often leaves
patients with only palliative treatment options [20].
Despite the benet of CRS/HIPEC, only about 25% of
patients with PC will be eligible for this approach given
the late presentation and burden of disease. In order to
expand patient eligibility and oer treatment with a
curative intent, early detection of PC, before signicant
tumor burden develops, is essential.
IMAGING
Traditional imaging, such as CT and MRI lack
sensitivity to both detect and estimate disease burden
in PC. Classic computed tomography (CT) signs of PC
such as “omental caking,” thickening of the omentum,
and peritoneal nodules are not common radiographic
ndings in “early” disease states. Several studies designed
to determine the specicity and accuracy of CT scans in
assessing tumor burden for PC found that CT signicantly
underestimated the amount of disease present in the
peritoneal cavity [21-24]. Sensitivity for CT detection
of tumor nodules less than 0.5 cm and 1cm had been
reported to be 11% and 25-50% respectively [23]. This
is particularly important in colorectal PC, where PCI is
a critical determinant of complete cytoreduction and
Figure 1: Peritoneal Cancer Index (PCI) scoring system. PCI is a diagnostic and prognostic tool that is a sum of scores in thirteen
abdominal regions. Each receives a score of 0-3 based on the largest tumor size in each region. Scores range from 0 to 39. Higher scores
indicate more widespread and/or larger tumors in the peritoneal cavity.
Oncotarget3
www.impactjournals.com/oncotarget
long-term outcomes [25]. In a study by Koh et al looking
specically at CRC PC they determined CT signicantly
underestimated clinical PCI [23]. In fact sensitivity of
small bowel involvement in each region ranged from
8-17%. Despite this, the Fifth International Workshop on
Peritoneal Surface Malignancy in Milan, identied CT as
the principal imaging modality to assess suitability for
CRS [26]. Ultrasound also has very limited sensitivity
for detecting PC nodules [27-29]. Magnetic resonance
imaging (MRI), and particularly diusion weighted
images, has been demonstrated in prospective studies to
have increased accuracy in detection of carcinomatosis
within certain areas of the abdomen [30]. This however
carries its own limitations due to the motion artifacts
of peristalsis, cost, and the need for radiologists trained
in their interpretation and inter-observer variation.
Additionally, positron emission technology (PET) may
have increased sensitivity, but similar limitations and
absence of added clinical value often precludes its use for
determining resectability [24, 31-33]. These diculties,
especially limited sensitivity, lack of meaningful clinical
correlation and high cost, reduce the utility of non-
invasive imaging in the early detection of PC. With the
current technology, laparoscopy or exploratory surgery is
often necessary to conrm the diagnosis and extent of PC
(see Table 2) [34].
LIQUID BIOPSY
Given the narrow subset of patients who are oered
CRS/HIPEC, a potentially life-saving treatment, we are
in need of a paradigm change. Patients who are either at
risk for developing PC or who are in the earliest stage
of this disease process may signicantly benet from
expanded treatment options. The term “liquid biopsy” has
reached prolic use as large-scale investigations seek to
identify tumor markers in the serum. This usually refers to
molecular diagnostic studies that are performed on blood
or body uid as opposed to cancerous tissue itself [35].
Multiple serum tumor markers: carcinoembryonic antigen
(CEA), carbohydrate antigen CA 19-9, and CA 125, are
commonly elevated in patients with PC and the degree of
elevation tends to correlate with the extent of PC [36].
However, these serum tumor markers are inadequate for
early detection of PC. Moreover, they lack specicity to
predict the presence or risk of PC in patients with CRC.
There is a critical clinical need to identify circulating
tumor biomarkers of aggressiveness, likelihood of
recurrence, risk of metastasis such as PC, or even the
presence of a malignancy to better tailor therapy for
patients. For example, if a patient with a newly diagnosed
stage III colorectal cancer is known to be at signicant risk
for peritoneal recurrence due to the presence of a specic
set of biomarkers in their serum, they may benet from
prophylactic HIPEC. This is just one example of how this
Table 1: CC is the completeness of cytoreduction score. It indicates the size of the largest tumor that remains after
cytoreductive surgery.
Completeness of Cytoreduction scores
Score Size of largest post-surgery residual tumor
CC-0 No visible tumor
CC-1 Less than 0.25 cm
CC-2 Between 0.25 cm and 2.5 cm
CC-3 > 2.5 cm or conuent
Table 2: This table summarizes the pros and cons of each non-invasive imaging modality in assessing PC
Non-Invasive Imaging Utility in PC Detection
Image
Modality Pros Cons
Sensitivity/Specicity as
compared to surgical analysis References
Ultrasound
Inexpensive,
Eective for
ascites detection
Limited PC nodule
sensitivity, highly operator
dependent
Non-specic [26] [24, 25]
CT
Standard staging
workup
Limited small PC nodule
sensitivity, Inter-observer
variability
25-100%/78-100% with only 11-
48% sensitivity for tumors less
than 5 mm [26] [18-21]
MRI
High PC
sensitivity
Relatively Expensive, slight
peristalsis motion artifact,
inter-observer variability
90%/95.5% (diusion weighted)
[26] [28]
PET/ PET-CT
High PC
sensitivity
Relatively Expensive,
Peristalsis motion artifact 78-97%/55-90% [26] [27, 28]
Oncotarget4
www.impactjournals.com/oncotarget
technology may be applied.
If blood-borne biomarkers for PC with high
sensitivity and specicity are discovered, patients
developing PC may be quickly identied with a blood test,
a liquid biopsy. One such type of biomarker, microRNAs
(miRs), short, non-coding RNAs that regulate mRNAs, has
demonstrated diagnostic utility by correctly identifying
several cancers of unknown primary with reasonable
accuracy [37]. The diagnostic miR prole that was used
in this study was generated from miR analysis in well
dierentiated primary tumors [37]. Several miRs, such as
miR-21, have been linked to gastrointestinal cancers as
potential diagnostic targets and prognostic indicators [38].
However these miRs and other types of RNAs are rapidly
degraded in the plasma [39-41].
EXOSOMES
Exosomes, small cell-derived vesicles (Figure 2),
can protect RNAs and miRNAs, from being degraded [42-
46]. When researchers exposed miRs to RNase, the miRs
that were in exosomes and cells were protected while the
free RNAs were degraded [42]. When exosomes were
exposed to RNase the contained RNAs were protected
from degradation while cellular RNA was degraded by the
same RNase [45]. Exosomes hold great potential for both
diagnosis and prognosis of diseases and are exceptionally
useful as cancer biomarkers [47]. When a panel of lung
cancer associated miRs was examined in solid tumors and
tumor exosomes from patient plasma, most of the miRs
were found to have highly comparable expression levels
(see Table 3 for the miRs) [48, 49]. Cervical cancer cell
line tumor derived exosomes (TEXs) contain survivin,
which contributes to cancer aggressiveness and metastatic
potential [50, 51]. In a study of ovarian cancer, greater
numbers of cancer exosomes were found in the serum
as more advanced cancer stages were examined [52].
Furthermore, approximately four times more serum
exosomes were discovered in lung cancer patients as
compared to cancer free controls and the exosomes
contained more than twice the miRs [48]. In our research,
we found increased serum exosome levels in patients with
prostate and breast cancers as compared to disease free
controls [53-55]. TEXs are prevalent in patient serum from
Table 3: These lung cancer associated miRs were discovered in both solid tumor and in tumor exosomes.
miR-17-3p miR-21 miR-106a miR-146 miR-155 miR-191
miR-192 miR-203 miR-205 miR-210 miR-212 miR-214
Table 4: Gastric Cancer PC associated exosomal miRs and their prevalence in various cancers.
Gastric Cancer Lung
Cancer
Liver
Cancer
Breast
Cancer
Prostate
Cancer
Colorectal
Cancer Adrenal Cancer
miR 1202
↔malignant ascites,
peritoneal lavage
uid, cell culture
[52]
X X
serum
exosomes
[54] XX↑Ø tumor tissue [62]
miR 1207-
5p
↔malignant ascites,
peritoneal lavage
uid, cell culture
[52], ↓tNm tumor
tissue [63],
↓ tumor tissue [64]
X
solid
tumor [56]
serum
exosomes
[54] ↑ serum [65]
solid
tumor
[66] X
miR 1225-
5p
↑tnM peritoneal
lavage uid [52],
↓ cancer tissue [53] X↓ blood
[67]
serum
exosomes
[54]
↑ blood [68],
↓ solid tumor
[69]
solid
tumor [70]
multiple types of
solid tumors [71]
miR 320c
↔malignant ascites,
peritoneal lavage
uid, cell culture
[52]
solid
tumor
[72] X X X X X
miR 4270
↔malignant ascites,
peritoneal lavage
uid, cell culture
[52]
X X
serum
exosomes
[54] X X X
All data is from human studies
↑, ↓-increased/decreased expression associated with cancer
↑tnM, ↓tnM -increased/decreased expression associated with metastatic cancer
↑tNm, ↓tNm -increased/decreased expression associated with lymph node metastasis
↑ Ø, ↓Ø -increased/decreased expression associated with shorter patient survival
↔-present but no signicant dierential regulation
X -no research discovered in literature
Oncotarget5
www.impactjournals.com/oncotarget
multiple cancer types and protect labile biomarkers from
degradation [42, 45, 48, 53-55].
Exosomes have not been extensively studied in
PC diagnostics. Andre and colleagues examined ascites
exosomes from patients with PC and found that the tumor
specic markers Her2/Neu, TRP1, and Mart1 were present
in ascites tumor exosomes [56]. Tokuhisa and colleagues
have identied several RNAs present in exosomes within
peritoneal ascites, peritoneal lavage, and PC metastatic
cell lines [57]. After exosome miR screening, 5 miRNAs
were selected as a panel of signicantly dierentially
regulated RNAs: miR 1202, 1207-5p, 1225-5p, 320c, and
4270 [57]. miR 21 had the strongest signal intensity in
malignant ascites [57]. Exosomes from peritoneal lavage
were probed for miR 21, 1225-5p, and 320c; miR 21 and
1225-5p were found to be upregulated in later stages of
gastric cancer and correlated with serosal invasion [57]. In
a study on primary gastric cancer tissue, miR 1255-5p was
generally downregulated and was found to inhibit cancer
cell growth, motility, as well as cancer invasion [58]. This
apparently contradictory result suggests that miR 1225-
5p in peritoneal lavage either is being released by non-
cancerous tissue in an attempt to stop abnormal growths
or is being used by cancers in order to facilitate better
attachment to the peritoneum. In apparently contradictory
results to the above study, in gastric cancer cell lines and
in vivo, miR 1255-5p was generally downregulated and
was found to inhibit cell growth and motility as well as
cancer invasion [58].
The prospective gastric PC miR biomarkers
described in the study by Tokuhisa and colleagues [57]
have been found to be associated with other cancers as
well (see Table 4). Notably, miR 1202, 1207-5p, 1225-5p,
and 4270, were found circulating in the blood of breast
cancer patients [59]. In human bladder cancer tissue
samples, miR 320c was signicantly downregulated [60].
In hepatocellular carcinoma tissue samples, miR 1207-
5p was found to be signicantly downregulated [61].
In adrenocortical carcinoma tissue samples, increased
miR 1202 expression was found to be associated with
signicantly reduced patient lifespan [62]. The gaps in our
knowledge of RNA signaling in cancer are immediately
apparent from Table 4. The observation that these miRs,
either individually or in a group, are associated with
multiple types of cancer and are found in exosomes from
the peritoneal cavity suggest the potential of exosomal
diagnosis of PC.
RNAS AND GASTRIC CANCER
RNAs are found to be globally downregulated in
cancer [37]. Upregulated RNAs and miRs are likely to
be related to cancer growth and function or the body’s
response to cancer. miR 320c inhibited cell growth and
motility in bladder cancer [60]. miR 1207-5p functioned
to inhibit cell growth and invasion in liver cancer but
functions to increase stemness in colorectal cancer [61,
63]. Since these exosomal-associated miRs have known
functions in tumors, the possibility emerges of tailoring
PC treatment based on what biomarkers are discovered
in a patient’s exosomes. miR 320c inhibited cell growth
and motility and miR 1207-5p functioned to inhibit cell
Figure 2: Tumor Cells release nanovesicles called exosomes which carry RNAs, including microRNAs and messenger
RNAs, and proteins.
Oncotarget6
www.impactjournals.com/oncotarget
growth and invasion. The observation that these exosomal-
associated miRs have known functions in tumors opens
up the possibility of utilizing these biomarkers to tailor an
individual patient’s treatment. [60, 61]
FUTURE DIRECTIONS
Additional research is needed to identify biomarkers
in peritoneal carcinomatosis. Distinguishing metastatic
disease of peritoneal origin from solid organ metastases
should be both biologically feasible and clinically
useful. As we look to exosomes to provide insight into
the eld, shared challenges in nanovesicle isolation and
validation will need to be addressed. Reliable and ecient
methods as well as recognized standards will need to be
established for clinical use. Future clinical work in this
eld should include the prospective collection of samples
for retrospective investigation. This will be instrumental
in establishing clinical validity and utility. We are hopeful
that the shared work of many will continue to yield
advances in reducing the burden of life lost from this
aggressive malady.
CONCLUSIONS
Recently, TEXs have been implicated in facilitating
metastasis. TEXs can be taken up by multiple cell types,
including endothelial cells, bone marrow progenitor
cells, and other cancer cells [64-67]. These exosomes
demonstrated the ability to deliver functional RNAs
and proteins to recipient cells, modifying their growth
patterns to be pro-oncogenic [65, 66]. Further, TEXs have
demonstrated the ability to greatly increase metastatic
tumor burden in a mouse model [64]. Studying TEXs
in the context of metastasis is a promising eld. PC is
currently dicult to detect at its onset. Late PC detection
usually leaves the disease incurable. PC must be detected
sooner for better patient outcomes. Non-invasive imaging
is impractical for early PC detection. Detection of PC by
means of markers within a patient’s biouids, such as a
serum “liquid biopsy” would be ideal. Both serum and
ascites contain biomarkers released by PC. Exosomes
are released into serum by multiple types of cancers and
protect their contents from degradation in the blood. These
provide us with a likely source of a sensitive and specic
diagnostic modality to detect PC in its earliest most
treatable from.
ACKNOWLEDGMENTS
The authors would like to thank the Center for
Health Disparities & Molecular Medicine for its support.
They would also like to thank members of the Wall
Laboratory for careful review of this manuscript.
FUNDING
Research reported in this publication was supported
by NIH award P20MD006988 (NRW).
CONFLICTS OF INTEREST
The Authors declare no conicts of interest.
REFERENCES
1. Levy AD, Shaw JC, Sobin LH. Secondary Tumors and
Tumorlike Lesions of the Peritoneal Cavity: Imaging
Features with Pathologic Correlation. RadioGraphics. 2009;
29: 347-73. doi: 10.1148/rg.292085189.
2. Sadeghi B, Arvieux C, Glehen O, Beaujard AC, Rivoire
M, Baulieux J, Fontaumard E, Brachet A, Caillot JL, Faure
JL, Porcheron J, Peix JL, François Y, et al. Peritoneal
carcinomatosis from non-gynecologic malignancies.
Cancer. 2000; 88: 358-63. doi: 10.1002/(SICI)1097-
0142(20000115)88:2 < 358::AID-CNCR16 > 3.0.CO;2-O.
3. Chia CS, You B, Decullier E, Vaudoyer D, Lorimier G,
Abboud K, Bereder JM, Arvieux C, Boschetti G, Glehen
O, Group tBIGR. Patients with Peritoneal Carcinomatosis
from Gastric Cancer Treated with Cytoreductive Surgery
and Hyperthermic Intraperitoneal Chemotherapy: Is Cure
a Possibility? Annals of Surgical Oncology. 2016: 1-9. doi:
10.1245/s10434-015-5081-3.
4. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016.
CA: A Cancer Journal for Clinicians. 2016; 66: 7-30. doi:
10.3322/caac.21332.
5. Mekenkamp LJM, Koopman M, Teerenstra S, van Krieken
JHJM, Mol L, Nagtegaal ID, Punt CJA. Clinicopathological
features and outcome in advanced colorectal cancer
patients with synchronous vs metachronous metastases.
British Journal of Cancer. 2010; 103: 159-64. doi: 10.1038/
sj.bjc.6605737.
6. Klaver YLB, Lemmens VEPP, Nienhuijs SW, Luyer MDP,
de Hingh IHJT. Peritoneal carcinomatosis of colorectal
origin: Incidence, prognosis and treatment options. World
Journal of Gastroenterology : WJG. 2012; 18: 5489-94. doi:
10.3748/wjg.v18.i39.5489.
7. Klaver CEL, Musters GD, Bemelman WA, Punt CJA,
Verwaal VJ, Dijkgraaf MGW, Aalbers AGJ, van der Bilt
JDW, Boerma D, Bremers AJA, Burger JWA, Buskens
CJ, Evers P, et al. Adjuvant hyperthermic intraperitoneal
chemotherapy (HIPEC) in patients with colon cancer at
high risk of peritoneal carcinomatosis; the COLOPEC
randomized multicentre trial. BMC Cancer. 2015; 15. doi:
10.1186/s12885-015-1430-7.
8. Mitry E, Guiu B, Cosconea S, Jooste V, Faivre J,
Bouvier A-M. Epidemiology, management and prognosis
of colorectal cancer with lung metastases: a 30-year
population-based study. Gut. 2010; 59: 1383-8. doi:
Oncotarget7
www.impactjournals.com/oncotarget
10.1136/gut.2010.211557.
9. Quere P, Facy O, Manfredi S, Jooste V, Faivre J, Lepage
C, Bouvier AM. Epidemiology, Management, and Survival
of Peritoneal Carcinomatosis from Colorectal Cancer: A
Population-Based Study. Diseases of the Colon & Rectum.
2015; 58: 743-52. doi: 10.1097/DCR.0000000000000412.
10. Yan TD, Cao CQ, Munkholm-Larsen S. A pharmacological
review on intraperitoneal chemotherapy for peritoneal
malignancy. World J Gastrointest Oncol. 2010; 2: 109-16.
doi: 10.4251/wjgo.v2.i2.109.
11. Goodman MD, McPartland S, Detelich D, Saif MW.
Chemotherapy for intraperitoneal use: a review of
hyperthermic intraperitoneal chemotherapy and early post-
operative intraperitoneal chemotherapy. J Gastrointest
Oncol. 2016; 7: 45-57. doi: 10.3978/j.issn.2078-
6891.2015.111.
12. Sugarbaker PH, Kern K, Lack E. Malignant pseudomyxoma
peritonei of colonic origin. Natural history and presentation
of a curative approach to treatment. Dis Colon Rectum.
1987; 30: 772-9. doi:
13. Elias D, Lefevre JH, Chevalier J, Brouquet A, Marchal
F, Classe J-M, Ferron G, Guilloit J-M, Meeus P, Goéré
D, Bonastre J. Complete Cytoreductive Surgery Plus
Intraperitoneal Chemohyperthermia With Oxaliplatin for
Peritoneal Carcinomatosis of Colorectal Origin. Journal
of Clinical Oncology. 2009; 27: 681-5. doi: 10.1200/
JCO.2008.19.7160.
14. Franko J, Shi Q, Goldman CD, Pockaj BA, Nelson GD,
Goldberg RM, Pitot HC, Grothey A, Alberts SR, Sargent
DJ. Treatment of Colorectal Peritoneal Carcinomatosis
With Systemic Chemotherapy: A Pooled Analysis of North
Central Cancer Treatment Group Phase III Trials N9741
and N9841. Journal of Clinical Oncology. 2012; 30: 263-7.
doi: 10.1200/JCO.2011.37.1039.
15. Verwaal VJ, Ruth Sv, Bree Ed, Slooten GWv, Tinteren
Hv, Boot H, Zoetmulder FAN. Randomized Trial
of Cytoreduction and Hyperthermic Intraperitoneal
Chemotherapy Versus Systemic Chemotherapy
and Palliative Surgery in Patients With Peritoneal
Carcinomatosis of Colorectal Cancer. Journal of
Clinical Oncology. 2003; 21: 3737-43. doi: 10.1200/
JCO.2003.04.187.
16. Chua TC. Progress in the combined modality management
of peritoneal carcinomatosis. Journal of Surgical Oncology.
2010; 102: 728-9. doi: 10.1002/jso.21748.
17. Sugarbaker PH. Successful management of microscopic
residual disease in large bowel cancer. Cancer
Chemotherapy and Pharmacology. 1999; 43: S15-S25. doi:
10.1007/s002800051093.
18. Harmon RL, Sugarbaker PH. Prognostic indicators in
peritoneal carcinomatosis from gastrointestinal cancer.
International Seminars in Surgical Oncology. 2005; 2: 3.
doi: 10.1186/1477-7800-2-3.
19. Verwaal VJ, Ruth Sv, Witkamp A, Boot H, Slooten Gv,
Zoetmulder FAN. Long-Term Survival of Peritoneal
Carcinomatosis of Colorectal Origin. Annals of Surgical
Oncology. 2004; 12: 65-71. doi: 10.1007/s10434-004-
1167-z.
20. Verwaal VJ, Kusamura S, Baratti D, Deraco M. The
eligibility for local-regional treatment of peritoneal surface
malignancy. Journal of Surgical Oncology. 2008; 98: 220-3.
doi: 10.1002/jso.21060.
21. Dromain C, Leboulleux S, Auperin A, Goere D, Malka D,
Lumbroso J, Schumberger M, Sigal R, Elias D. Staging
of peritoneal carcinomatosis: enhanced CT vs. PET/CT.
Abdominal Imaging. 2007; 33: 87-93. doi: 10.1007/s00261-
007-9211-7.
22. Esquivel J, Chua Tc, Stojadinovic A, Melero JT, Levine Ea,
Gutman M, Howard R, Piso P, Nissan A, Gomez-Portilla
A, Gonzalez-Bayon L, Gonzalez-Moreno S, Shen P, et al.
Accuracy and clinical relevance of computed tomography
scan interpretation of peritoneal cancer index in colorectal
cancer peritoneal carcinomatosis: A multi-institutional
study. Journal of Surgical Oncology. 2010; 102: 565-70.
doi: 10.1002/jso.21601.
23. Koh J-L, Yan TD, Glenn D, Morris DL. Evaluation
of Preoperative Computed Tomography in Estimating
Peritoneal Cancer Index in Colorectal Peritoneal
Carcinomatosis. Annals of Surgical Oncology. 2008; 16:
327-33. doi: 10.1245/s10434-008-0234-2.
24. Low RN, Barone RM, Lucero J. Comparison of MRI
and CT for predicting the Peritoneal Cancer Index
(PCI) preoperatively in patients being considered for
cytoreductive surgical procedures. Annals of Surgical
Oncology. 2015; 22: 1708-15. doi: 10.1245/s10434-014-
4041-7.
25. da Silva RG, Sugarbaker PH. Analysis of prognostic
factors in seventy patients having a complete cytoreduction
plus perioperative intraperitoneal chemotherapy for
carcinomatosis from colorectal cancer. J Am Coll Surg.
2006; 203: 878-86. doi: 10.1016/j.jamcollsurg.2006.08.024.
26. Baratti D, Kusamura S, Deraco M. The Fifth International
Workshop on Peritoneal Surface Malignancy (Milan, Italy,
December 4-6, 2006): methodology of disease-specic
consensus. Journal of Surgical Oncology. 2008; 98: 258-
62. doi: 10.1002/jso.21056.
27. Repiso A, Gómez-Rodríguez R, López-Pardo R, Lombera
MM, Romero M, Aranzana A, Abad S, Rodríguez-Merlo
R, López L, Carboles JM. Usefulness of endoscopic
ultrasonography in preoperative gastric cancer staging:
diagnostic yield and therapeutic impact. Revista Española
De Enfermedades Digestivas: Organo Ocial De La
Sociedad Española De Patología Digestiva. 2010; 102: 413-
20. doi:
28. Savelli L, De Iaco P, Ceccaroni M, Ghi T, Ceccarini M,
Seracchioli R, Cacciatore B. Transvaginal sonographic
features of peritoneal carcinomatosis. Ultrasound in
Obstetrics and Gynecology. 2005; 26: 552-7. doi: 10.1002/
uog.2587.
Oncotarget8
www.impactjournals.com/oncotarget
29. Patel CM, Sahdev A, Reznek RH. CT, MRI and PET
imaging in peritoneal malignancy. Cancer Imaging. 2011;
11: 123-39. doi: 10.1102/1470-7330.2011.0016.
30. Fehniger J, Thomas S, Lengyel E, Liao C, Tenney M, Oto
A, Yamada SD. A prospective study evaluating diusion
weighted magnetic resonance imaging (DW-MRI) in
the detection of peritoneal carcinomatosis in suspected
gynecologic malignancies. Gynecol Oncol. 2016; 142: 169-
75. doi: 10.1016/j.ygyno.2016.04.018.
31. De Vos N, Goethals I, Ceelen W. Clinical Value of (18)
F-FDG- PET-CT in the Preoperative Staging of Peritoneal
Carcinomatosis from Colorectal Origin. Acta Chirurgica
Belgica. 2014; 114: 370-5. doi:
32. Klumpp BD, Schwenzer N, Ascho P, Miller S, Kramer U,
Claussen CD, Bruecher B, Koenigsrainer A, Pfannenberg
C. Preoperative assessment of peritoneal carcinomatosis:
intraindividual comparison of 18F-FDG PET/CT and MRI.
Abdominal Imaging. 2012; 38: 64-71. doi: 10.1007/s00261-
012-9881-7.
33. Low RN, Barone RM, Lacey C, Sigeti JS, Alzate GD,
Sebrechts CP. Peritoneal tumor: MR imaging with dilute
oral barium and intravenous gadolinium-containing
contrast agents compared with unenhanced MR imaging
and CT. Radiology. 1997; 204: 513-20. doi: 10.1148/
radiology.204.2.9240546.
34. Yonemura Y, Canbay E, Endou Y, Ishibashi H, Mizumoto
A, Miura M, Li Y, Liu Y, Takeshita K, Ichinose M, Takao
N, Hirano M, Sako S, et al. Peritoneal cancer treatment.
Expert Opinion on Pharmacotherapy. 2014; 15: 623-36. doi:
10.1517/14656566.2014.879571.
35. Kin C, Kidess E, Poultsides GA, Visser BC, Jerey SS.
Colorectal cancer diagnostics: biomarkers, cell-free
DNA, circulating tumor cells and dening heterogeneous
populations by single-cell analysis. Expert Review
of Molecular Diagnostics. 2013; 13: 581-99. doi:
10.1586/14737159.2013.811896.
36. Wagner PL, Austin F, Sathaiah M, Magge D, Maduekwe
U, Ramalingam L, Jones HL, Holtzman MP, Ahrendt SA,
Zureikat AH, Pingpank JF, Zeh HJ, Bartlett DL, et al.
Signicance of Serum Tumor Marker Levels in Peritoneal
Carcinomatosis of Appendiceal Origin. Annals of surgical
oncology. 2013; 20. doi: 10.1245/s10434-012-2627-5.
37. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J,
Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando
AA, Downing JR, Jacks T, Horvitz HR, et al. MicroRNA
expression proles classify human cancers. Nature. 2005;
435: 834-8. doi: 10.1038/nature03702.
38. Yin C, Zhou X, Dang Y, Yan J, Zhang G. Potential Role
of Circulating MiR-21 in the Diagnosis and Prognosis of
Digestive System Cancer: A Systematic Review and Meta-
Analysis. Medicine (Baltimore). 2015; 94: e2123. doi:
10.1097/MD.0000000000002123.
39. Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J,
Zhang Y, Chen J, Guo X, Li Q, Li X, Wang W, et al.
Characterization of microRNAs in serum: a novel class of
biomarkers for diagnosis of cancer and other diseases. Cell
Research. 2008; 18: 997-1006. doi: 10.1038/cr.2008.282.
40. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman
SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom
J, O’Briant KC, Allen A, Lin DW, Urban N, Drescher
CW, et al. Circulating microRNAs as stable blood-based
markers for cancer detection. Proceedings of the National
Academy of Sciences. 2008; 105: 10513-8. doi: 10.1073/
pnas.0804549105.
41. Tsui NBY, Ng EKO, Lo YMD. Stability of Endogenous
and Added RNA in Blood Specimens, Serum, and Plasma.
Clinical Chemistry. 2002; 48: 1647-53. doi:
42. Koga A, Aoyagi K, Imaizumi T, Miyagi M, Shirouzu K.
Comparison between the Gastric Cancer Cell Line MKN-
45 and the High-Potential Peritoneal Dissemination Gastric
Cancer Cell Line MKN-45P. The Kurume Medical Journal.
2011; 58: 73-9. doi: 10.2739/kurumemedj.58.73.
43. Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van
Eijndhoven MAJ, Hopmans ES, Lindenberg JL, de Gruijl
TD, Würdinger T, Middeldorp JM. Functional delivery of
viral miRNAs via exosomes. Proceedings of the National
Academy of Sciences of the United States of America.
2010; 107: 6328-33. doi: 10.1073/pnas.0914843107.
44. Skog J, Wurdinger T, van Rijn S, Meijer D, Gainche L,
Sena-Esteves M, Curry WT, Carter RS, Krichevsky AM,
Breakeeld XO. Glioblastoma microvesicles transport
RNA and protein that promote tumor growth and provide
diagnostic biomarkers. Nature cell biology. 2008; 10: 1470-
6. doi: 10.1038/ncb1800.
45. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ,
Lötvall JO. Exosome-mediated transfer of mRNAs and
microRNAs is a novel mechanism of genetic exchange
between cells. Nature Cell Biology. 2007; 9: 654-9. doi:
10.1038/ncb1596.
46. Cheng L, Sharples RA, Scicluna BJ, Hill AF. Exosomes
provide a protective and enriched source of miRNA for
biomarker proling compared to intracellular and cell-
free blood. J Extracell Vesicles. 2014; 3. doi: 10.3402/jev.
v3.23743.
47. Khan S, Bennit HF, Wall NR. The emerging role of
exosomes in survivin secretion. Histol Histopathol. 2015;
30: 43-50. doi: 10.14670/hh-30.43.
48. Rabinowits G, Gerçel-Taylor C, Day JM, Taylor DD,
Kloecker GH. Exosomal MicroRNA: A Diagnostic Marker
for Lung Cancer. Clinical Lung Cancer. 2009; 10: 42-6. doi:
10.3816/CLC.2009.n.006.
49. Yanaihara N, Caplen N, Bowman E, Seike M, Kumamoto
K, Yi M, Stephens RM, Okamoto A, Yokota J, Tanaka T,
Calin GA, Liu CG, Croce CM, et al. Unique microRNA
molecular proles in lung cancer diagnosis and
prognosis. Cancer Cell. 2006; 9: 189-98. doi: 10.1016/j.
ccr.2006.01.025.
50. Khan S, Aspe JR, Asumen MG, Almaguel F, Odumosu
O, Acevedo-Martinez S, De Leon M, Langridge WH,
Oncotarget9
www.impactjournals.com/oncotarget
Wall NR. Extracellular, cell-permeable survivin inhibits
apoptosis while promoting proliferative and metastatic
potential. Br J Cancer. 2009; 100: 1073-86. doi: 6604978
[pii] 10.1038/sj.bjc.6604978.
51. Khan S, Jutzy JM, Aspe JR, McGregor DW, Neidigh
JW, Wall NR. Survivin is released from cancer cells via
exosomes. Apoptosis. 2011; 16: 1-12. doi: 10.1007/s10495-
010-0534-4.
52. Taylor DD, Gercel-Taylor C. MicroRNA signatures of
tumor-derived exosomes as diagnostic biomarkers of
ovarian cancer. Gynecologic Oncology. 2008; 110: 13-21.
doi: 10.1016/j.ygyno.2008.04.033.
53. Khan S, Bennit HF, Turay D, Perez M, Mirshahidi S, Yuan
Y, Wall NR. Early diagnostic value of survivin and its
alternative splice variants in breast cancer. BMC Cancer.
2014; 14: 176. doi: 10.1186/1471-2407-14-176.
54. Khan S, Jutzy JM, Valenzuela MM, Turay D, Aspe JR,
Ashok A, Mirshahidi S, Mercola D, Lilly MB, Wall NR.
Plasma-derived exosomal survivin, a plausible biomarker
for early detection of prostate cancer. PLoS One. 2012; 7:
e46737. doi: 10.1371/journal.pone.0046737.
55. Turay D, Khan S, Diaz Osterman CJ, Curtis MP, Khaira
B, Neidigh JW, Mirshahidi S, Casiano CA, Wall NR.
Proteomic Proling of Serum-Derived Exosomes from
Ethnically Diverse Prostate Cancer Patients. Cancer Invest.
2016; 34: 1-11. doi: 10.3109/07357907.2015.1081921.
56. Andre F, Schartz NE, Movassagh M, Flament C, Pautier P,
Morice P, Pomel C, Lhomme C, Escudier B, Le Chevalier
T, Tursz T, Amigorena S, Raposo G, et al. Malignant
eusions and immunogenic tumour-derived exosomes.
Lancet. 2002; 360: 295-305. doi: S0140-6736(02)09552-1
[pii] 10.1016/S0140-6736(02)09552-1.
57. Tokuhisa M, Ichikawa Y, Kosaka N, Ochiya T, Yashiro M,
Hirakawa K, Kosaka T, Makino H, Akiyama H, Kunisaki C,
Endo I. Exosomal miRNAs from Peritoneum Lavage Fluid
as Potential Prognostic Biomarkers of Peritoneal Metastasis
in Gastric Cancer. PLoS One. 2015; 10: e0130472. doi:
10.1371/journal.pone.0130472.
58. Zheng H, Zhang F, Lin X, Huang C, Zhang Y, Li Y, Lin J,
Chen W, Lin X. MicroRNA-1225-5p inhibits proliferation
and metastasis of gastric carcinoma through repressing
insulin receptor substrate-1 and activation of β-catenin
signaling. Oncotarget. 2015; 7: 4647-63. doi: 10.18632/
oncotarget.6615.
59. Hamam R, Ali AM, Alsaleh KA, Kassem M, Alfayez M,
Aldahmash A, Alajez NM. microRNA expression proling
on individual breast cancer patients identies novel panel
of circulating microRNA for early detection. Scientic
Reports. 2016; 6: 25997. doi: 10.1038/srep25997.
60. Wang X, Wu J, Lin Y, Zhu Y, Xu X, Xu X, Liang Z, Li S,
Hu Z, Zheng X, Xie L. MicroRNA-320c inhibits tumorous
behaviors of bladder cancer by targeting Cyclin-dependent
kinase 6. Journal of Experimental & Clinical Cancer
Research : CR. 2014; 33: 69. doi: 10.1186/s13046-014-
0069-6.
61. Zhao G, Dong L, Shi H, Li H, Lu X, Guo X, Wang J.
MicroRNA-1207-5p inhibits hepatocellular carcinoma
cell growth and invasion through the fatty acid synthase-
mediated Akt/mTOR signalling pathway. Oncology
Reports. 2016. doi: 10.3892/or.2016.4952.
62. Özata DM, Caramuta S, Velázquez-Fernández D, Akçakaya
P, Xie H, Höög A, Zedenius J, Bäckdahl M, Larsson C,
Lui W-O. The role of microRNA deregulation in the
pathogenesis of adrenocortical carcinoma. Endocrine-
Related Cancer. 2011; 18: 643-55. doi: 10.1530/ERC-11-
0082.
63. Farhana L, Antaki F, Anees MR, Nangia‐Makker P,
Judd S, Hadden T, Levi E, Murshed F, Yu Y, Van Buren
E, Ahmed K, Dyson G, Majumdar APN. Role of cancer
stem cells in racial disparity in colorectal cancer. Cancer
Medicine. 2016; 5: 1268-78. doi: 10.1002/cam4.690.
64. Hoshino A, Costa-Silva B, Shen TL, Rodrigues G,
Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di
Giannatale A, Ceder S, Singh S, Williams C, Soplop N,
et al. Tumour exosome integrins determine organotropic
metastasis. Nature. 2015; 527: 329-35. doi: 10.1038/
nature15756.
65. Paggetti J, Haderk F, Seiert M, Janji B, Distler U,
Ammerlaan W, Kim YJ, Adam J, Lichter P, Solary E,
Berchem G, Moussay E. Exosomes released by chronic
lymphocytic leukemia cells induce the transition of stromal
cells into cancer-associated broblasts. Blood. 2015; 126:
1106-17. doi: 10.1182/blood-2014-12-618025.
66. Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-
Silva B, Moreno-Bueno G, Hergueta-Redondo M, Williams
C, Garcia-Santos G, Ghajar C, Nitadori-Hoshino A,
Homan C, Badal K, et al. Melanoma exosomes educate
bone marrow progenitor cells toward a pro-metastatic
phenotype through MET. Nat Med. 2012; 18: 883-91. doi:
10.1038/nm.2753.
67. Franzen CA, Simms PE, Van Huis AF, Foreman KE,
Kuo PC, Gupta GN. Characterization of uptake and
internalization of exosomes by bladder cancer cells. Biomed
Res Int. 2014; 2014: 619829. doi: 10.1155/2014/619829.
68. Huang K-H, Lan Y-T, Fang W-L, Chen J-H, Lo S-S, Li
AF-Y, Chiou S-H, Wu C-W, Shyr Y-M. The Correlation
between miRNA and Lymph Node Metastasis in Gastric
Cancer. BioMed Research International. 2015; 2015. doi:
10.1155/2015/543163.
69. Chen L, Lü MH, Zhang D, Hao NB, Fan YH, Wu YY,
Wang SM, Xie R, Fang DC, Zhang H, Hu CJ, Yang SM.
miR-1207-5p and miR-1266 suppress gastric cancer growth
and invasion by targeting telomerase reverse transcriptase.
Cell Death & Disease. 2014; 5: e1034. doi: 10.1038/
cddis.2013.553.
70. Moltzahn F, Olshen AB, Baehner L, Peek A, Fong L,
Stöppler H, Simko J, Hilton JF, Carroll P, Blelloch R.
Microuidic based multiplex qRT-PCR identies diagnostic
and prognostic microRNA signatures in sera of prostate
cancer patients. Cancer research. 2011; 71: 550-60. doi:
Oncotarget10
www.impactjournals.com/oncotarget
10.1158/0008-5472.CAN-10-1229.
71. Zhu J, Chen L, Zou L, Yang P, Wu R, Mao Y, Zhou H,
Li R, Wang K, Wang W, Hua D, Zhang X. MiR-20b,
-21, and -130b inhibit PTEN expression resulting in
B7-H1 over-expression in advanced colorectal cancer.
Human Immunology. 2014; 75: 348-53. doi: 10.1016/j.
humimm.2014.01.006.
72. Fricke A, Ullrich PV, Heinz J, Pfeifer D, Scholber J, Herget
GW, Hauschild O, Bronsert P, Stark GB, Bannasch H,
Eisenhardt SU, Braig D. Identication of a blood-borne
miRNA signature of synovial sarcoma. Molecular Cancer.
2015; 14. doi: 10.1186/s12943-015-0424-z.
73. Leidinger P, Hart M, Backes C, Rheinheimer S, Keck B,
Wullich B, Keller A, Meese E. Dierential blood-based
diagnosis between benign prostatic hyperplasia and prostate
cancer: miRNA as source for biomarkers independent of
PSA level, Gleason score, or TNM status. Tumor Biology.
2016; 37: 10177-85. doi: 10.1007/s13277-016-4883-7.
74. He H-c, Han Z-d, Dai Q-s, Ling X-h, Fu X, Lin Z-y,
Deng Y-h, Qin G-q, Cai C, Chen J-h, Jiang F-n, Liu X,
Zhong W-d. Global analysis of the dierentially expressed
miRNAs of prostate cancer in Chinese patients. BMC
Genomics. 2013; 14: 757. doi: 10.1186/1471-2164-14-757.
75. Bartley AN, Yao H, Barkoh BA, Ivan C, Mishra BM,
Rashid A, Calin GA, Luthra R, Hamilton SR. Complex
Patterns of Altered MicroRNA Expression during the
Adenoma-Adenocarcinoma Sequence for Microsatellite-
Stable Colorectal Cancer. Clinical cancer research : an
ocial journal of the American Association for Cancer
Research. 2011; 17: 7283-93. doi: 10.1158/1078-0432.
CCR-11-1452.
76. Velázquez-Fernández D, Caramuta S, Özata DM, Lu M,
Höög A, Bäckdahl M, Larsson C, Lui W-O, Zedenius
J. MicroRNA expression patterns associated with
hyperfunctioning and non-hyperfunctioning phenotypes
in adrenocortical adenomas. European Journal of
Endocrinology. 2014; 170: 583-91. doi: 10.1530/EJE-13-
0817.
77. Lei T, Zhu Y, Jiang C, Wang Y, Fu J, Fan Z, Qin H.
MicroRNA-320 was downregulated in non-small cell
lung cancer and inhibited cell proliferation, migration
and invasion by targeting fatty acid synthase. Molecular
Medicine Reports. 2016; 14: 1255-62. doi: 10.3892/
mmr.2016.5370.
... The term peritoneal carcinomatosis (PC) was first coined in 1931 to describe an invasion of the peritoneal wall by ovarian cancer cells [1,2]. Up until today, peritoneal carcinomatosis, peritoneal carcinosis, or peritoneal dissemination usually referred to the state when solid cancer spreads to the peritoneal lining, or peritoneal wall [3]. The emphasis the presence of PC signifies a very poor prognosis. ...
... The emphasis the presence of PC signifies a very poor prognosis. Novel therapeutic options such as cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy (HIPEC) as well as pressurized intraperitoneal aerosol chemotherapy (PIPAC) [4,6] enable longer overall survival of some PC patients [3,18,19]. However, the patients for this type of treatment need to be carefully selected [20,21]; thus, for most of the affected, a PC verdict means a fatal late-stage condition oftentimes managed using only palliative care approaches [5,22]. ...
... The undeniable value of this work comes from the fact that, as one of very few at the time, it provided an analysis of miRNA levels in accumulated peritoneal fluid, ascites, which accompanies intraperitoneal tumors, most commonly ovarian cancer [38,39]. The authors in this proof-of-concept study showed that the measurement of miRNAs in ascites is feasible and should be considered a viable option for future semi-invasive diagnostic purposes [27] since current diagnostic strategies such as CT and ultrasound lack the sensitivity to detect PC, causing the disease to be discovered much too late [3]. ...
Article
Full-text available
Simple Summary Peritoneal carcinomatosis is a term for cancer cells spreading from tumors of internal organs and massively invading a large part of the membrane lining the abdomen and pelvis. For most patients, peritoneal carcinomatosis suggests only several months of life left. Current medicine can offer only alleviation of symptoms from this incurable disease. Researchers are intensely exploring some new therapeutic targets. Among promising candidates are non-coding RNAs, short molecules serving as important regulators in cells. When a disease such as cancer develops in the body, it is accompanied by typical changes in levels of non-coding RNAs. In this review, we provide an overview of current state of knowledge regarding the changes of non-coding RNA levels in peritoneal carcinomatosis. Deeper understanding of this topic could lead to the identification of non-coding RNAs as feasible specific biomarkers or novel therapeutic targets in the treatment of peritoneal carcinomatosis. Abstract Peritoneal carcinomatosis represents an advanced stage of tumors within the peritoneal cavity. Once considered an incurable terminal cancer metastasis, contemporary medicine is on the hunt for certain potentially curative options alongside the present day’s palliative disease management. However, for most patients, peritoneal carcinomatosis continues to pose a fatal late-stage prognosis with a grim future outlook. Over the past two decades, non-coding RNAs have garnered significant attention due to their undeniable significance in regulating cellular processes across all levels. Disruption of the intricate regulation led by non-coding RNAs has been demonstrated to have a substantial impact on various human diseases, particularly in cancer, including solid tumors originating from the organs of the peritoneal cavity. This review aims to offer a comprehensive overview of the current state of knowledge in the under-researched field of peritoneal carcinomatosis, focusing specifically on the role of non-coding RNAs in the development of this condition and delineating potential avenues for future research.
... More than 50% of patients with CRC develop liver metastases, 66 and peritoneal carcinomatosis 67 is common in patients with BRAF mutation. 68 Cremolini explained that, historically, patients with BRAF-mutated mCRC were rarely offered surgery with radical intent because of the systemic nature of the disease; however, surgery may improve disease control and long-term outcome, 31,69-71 and should be an option for these patients. ...
... Taieb concurred that patients with BRAF mutation and resectable metastatic lesions should not be denied surgical intervention; however, clinicians must ensure that all metastatic lesions are identified and resected, which is particularly challenging in the case of peritoneal carcinomatosis. 67 58 was likely because the patients in the real-world setting were more heavily pretreated than those enrolled in the clinical trial. Cremolini stated that this is an indication to use encorafenib plus cetuximab earlier in the treatment pathway for these patients. ...
Article
Colorectal cancer (CRC) is the third most common cancer worldwide, and the second leading cause of cancer death. Approximately one in five patients with CRC present with metastatic disease at diagnosis. The BRAF V600E mutation occurs in 8–12% of patients with metastatic colorectal cancer (mCRC), and is characterised by an aggressive clinical course and poor prognosis. This article is based on a webinar discussion in March 2024, between two experts in gastrointestinal cancers, Chiara Cremolini, University of Pisa, Italy; and Julien Taieb, Georges Pompidou European Hospital, Université Paris-Cité, France, both of whom have a wealth of experience and expertise in the clinical management of CRC. The experts described the most important recent advances in the treatment of BRAF V600E-mutated mCRC, including data presented at the European Society for Medical Oncology (ESMO) Congress in October 2023, and the American Society of Clinical Oncology (ASCO) Gastrointestinal (GI) Cancers Symposium in January 2024. Cremolini and Taieb gave valuable insights into topics such as the aggressive nature of BRAF V600E-mutated mCRC, and how this impacts choice of treatment, patient outcomes, and quality of life, as well as the importance of early testing and monitoring. The experts also discussed how the BRAF V600E mutation impacts treatment response and outcomes in patients with microsatellite unstable (microsatellite instability [MSI]) versus microsatellite stable (MSS) tumours, and recent key clinical trials in BRAF V600E-mutated mCRC. The importance of surgery in the multidisciplinary management of patients with BRAF V600E-mutated mCRC, BRAF as a prognostic marker in resected CRC, and real-world studies in this field were also explored. Finally, Cremolini and Taieb described what the future of the management of patients with BRAF V600E-mutated mCRC might look like, and which advancements in research they would like to see.
... It is a late-stage complication seen in gastrointestinal, genitourinary, and gynecological malignancies wherein tumors metastasize to and deposit on the peritoneal surface, often leaving patients with palliative care options. 8 However, a panel of ascitic tumor markers (CEA, CA15-3, CA19-9, and CA125) can be used to differentiate between the 2 conditions with symptoms ranging from abdominal pain and distension to abnormal bowel motility and obstruction. Radiologically, ascites, omental caking, thickening of the peritoneal reflection, soft nodules, and occasionally calcifications can be seen. ...
... Corticosteroids and nonsteroidal anti-inflammatory drugs such as indomethacin have been used to treat this disorder; however, it is unclear whether these strategies could prevent the formation of adhesions and fistulas. [2][3][4][5][6][7][8][9][10][11] Conclusion Postoperative granulomatous peritonitis is rare. However, it should be considered as a differential diagnosis in patients with features of omental stranding and nodularity after abdominal surgery. ...
Article
Full-text available
Background. Granulomatous peritonitis is a rare postoperative complication caused by a delayed hypersensitivity reaction to foreign substances. It can be challenging to diagnose owing to its vague presentations, and its possibility is often overlooked. Tubercular peritonitis and peritoneal carcinomatosis are the 2 crucial differential diagnoses that need to be taken into account. However, making a clinical differentiation between these 2 entities is challenging and necessitates a careful histopathological and microbiological analysis. Case Presentation. In this report, we present the case of a 28-year-old female who developed granulomatous peritonitis following a right ovarian cystectomy. The diagnosis was confirmed by histopathological examination. Conclusion. We must be aware of this rare entity, which, if left untreated, could have serious consequences, and consider its possibility in cases where the patient complains of abdominal pain after any abdominal procedure. We hope to provide insights into the importance of histopathological examination in aiding a confirmatory diagnosis of this entity.
... Case Report (ISSN: 2832-5788) Int Clinc Med Case Rep Jour (ICMCRJ) 2024 | Volume 3 | Issue 11 regions, engage in an aggressive, infiltrative process [1,2]. Distinguishing between primary and secondary peritoneal cancers is paramount for tailoring effective therapeutic strategies. ...
Article
Full-text available
Peritoneal carcinomatosis poses a diagnostic challenge due to its diverse range of differentials and late-stage presentation. This case report highlights the diagnostic journey of a 71-year-old white male with Peritoneal carcinomatosis secondary to metastatic adenocarcinoma with signet ring cell features originating from the upper gastrointestinal tract. A 71-year-old white male presented with worsening abdominal pain and discomfort over three months. Imaging revealed omental thickening, malignant ascites, and subsequent investigations led to the diagnosis of metastatic adenocarcinoma with signet ring cell features. Management complexities included pulmonary embolism, FOLFOX chemotherapy, recurrent paracentesis, and small bowel obstruction from an inguinal hernia.This case underscores the importance of early detection in optimizing treatment outcomes for Peritoneal carcinomatosis. Collaboration among specialties is crucial for managing the complexities associated with this condition. Further research is needed to refine diagnostic and therapeutic strategies for advanced gastrointestinal malignancies presenting with Peritoneal carcinomatosis
... Another potential imaging modality for assessing PC is MRI, with high sensitivity. Nevertheless, MRI is relatively costly, more susceptible to peristalsis or respiratory motion, cannot be used in patients with non-MRI compatible fiducial markers, and has substantial inter-observer variability [25]. So, MRI may not be suitable for all patients. ...
Article
Full-text available
Objective To assess the value of material density (MD) images generated from a rapid kilovoltage-switching dual-energy CT (rsDECT) in early detection of peritoneal carcinomatosis (PC). Materials and methods Thirty patients (60 ± 13 years; 24 women) with PC detected on multiple abdominal DECT scans were included. Four separate DECTs with varying findings of PC from each patient were used for qualitative/quantitative analysis, resulting in a total of 120 DECT scans (n = 30 × 4). Three radiologists independently reviewed DECT images (65 keV alone and 65 keV + MD) for diagnosis of PC (diagnostic confidence, lesion conspicuity, sharpness/delineation and image quality) using a 5-point Likert scale. Quantitative estimation of contrast-to-noise ratio (CNR) was done. Wilcoxon signed-rank test and Odds ratio calculation were used to compare between the two protocols. Inter-observer agreement was evaluated using Kappa coefficient analysis. P values < 0.05 were considered statistically significant. Results 65 keV + MD images showed a slightly higher sensitivity (89%[95%CI:84,92]) for PC detection compared with 65 keV images alone without statistical significance (84%[95%CI:78,88], p = 0.11) with the experienced reader showing significant improvement (98%[95%CI:93,100] vs. 90%[95%CI:83,94], p = 0.02). On a per-patient basis, use of MD images allowed earlier diagnosis for PC in an additional 13–23% of patients. On sub-group analysis, earlier diagnosis of PC was particularly beneficial in patients with BMI ≤ 29.9 kg/m². 65 keV + MD images showed higher diagnostic confidence, lesion conspicuity, and lesion sharpness for the experienced reader (p < 0.001). CNR was higher in MD images (1.7 ± 0.5) than 65 keV images (0.1 ± 0.02, p < 0.001). All readers showed moderate interobserver agreement for determining PC by both protocols (κ = 0.58 and κ = 0.47). Conclusion MD images allow earlier and improved detection of PC with the degree of benefit varying based on reader experience and patient body habitus. Graphical abstract
... They are contrast enhancing nodular lesions generally distributed over the peritoneum, mesentery and the omentum. Associated mild to moderate ascites is a common occurrence in peritoneal carcinomatosis (not present in our patient) [5]. ...
Article
The Peritoneal Cancer Index (PCI), calculated intraoperatively, has previously yielded mixed results when correlated with computed tomography. This study aimed to quantify variation in this scoring method comparing radiologists’ and surgeons’ radiologic PCI (rPCI) assessment. The rPCI of 104 patients treated at a single institution for peritoneal carcinomatosis was calculated by an abdominal radiologist and a surgeon. An additional 36-patient cohort was studied to compare preoperative rPCI with intraoperative gold standard PCI. Agreement was compared using kappa statistics. The rPCI of the 104 patients studied ranged from 2 to 39 (median, 12; interquartile range [IQR], 6–23) by the radiologist’s analysis and 2 to 37 (median, 9; IQR, 6–15) by the surgeon’s analysis. There was good agreement for PCI cutoffs of 15 (77.48%; kappa, 0.40) and 20 (78.63%; kappa, 0.24). The 36-patient cohort undergoing surgical exploration showed a median rPCI of 4 (IQR, 2–5.75) and a median intraoperative PCI of 11 (IQR, 6–12), with a significant difference in score by method (p < 0.001, Wilcoxon signed-rank test). For rPCI cutoffs greater than 15 and 20, the surgeon’s and radiologist’s rPCI showed strong concordance, denoting the interobserver reproducibility of rPCI. Moreover, concordance with intraoperative PCI translated to radiographic assessment. The rPCI consistently underestimated intraoperative PCI, suggesting that rPCI may be a useful conservative tool for assessing peritoneal burden. Although surgical exploration is needed to “rule in” patients as candidates for CRS, the authors suggest that rPCI can be used to “rule out” patients as CRS candidates based on institutional PCI cutoffs.
Chapter
Peritoneal carcinomatosis is a malignant cancer that spreads to the surface lining of a person's abdominal cavity and is usually caused by infection from other organs. AI developments, one of which is YOLO, can be used to help detect peritoneal carcinomatosis lesions. This research detects peritoneal carcinomatosis lesions by comparing several versions of YOLO with different scales, namely YOLOv5n, YOLOv5s, YOLOv5m, YOLOv5l, YOLOv6sn, YOLOv6s, YOLOv6m, YOLOv6l, YOLOv8n, YOLOv8s, YOLOv8m, and YOLOv8l. Recall, precision and mean Average Precision (mAP) metrics are all used in this study as well as inference time. The results show that the recommended models are YOLOv8l and YOLOv5l where both get the same high results with mAP of 0.799, followed by YOLOv8s, with mAP results of 0.796. The study's findings are intended to direct future clinical applications and determine the most appropriate model for the identification of peritoneal carcinomatosis. This study provides in-depth information that forms the basis for informed decision-making, highlighting the accuracy required to address issues related to peritoneal carcinomatosis
Article
Full-text available
Exosomes are vesicles of endocytic origin released by many cells. These vesicles can mediate communication between cells, facilitating processes such as antigen presentation. Here, we show that exosomes from a mouse and a human mast cell line (MC/9 and HMC-1, respectively), as well as primary bone marrow-derived mouse mast cells, contain RNA. Microarray assessments revealed the presence of mRNA from approximately 1300 genes, many of which are not present in the cytoplasm of the donor cell. In vitro translation proved that the exosome mRNAs were functional. Quality control RNA analysis of total RNA derived from exosomes also revealed presence of small RNAs, including microRNAs. The RNA from mast cell exosomes is transferable to other mouse and human mast cells. After transfer of mouse exosomal RNA to human mast cells, new mouse proteins were found in the recipient cells, indicating that transferred exosomal mRNA can be translated after entering another cell. In summary, we show that exosomes contain both mRNA and microRNA, which can be delivered to another cell, and can be functional in this new location. We propose that this RNA is called " exosomal shuttle RNA " (esRNA). Exosomes are small (50–90 nm) membrane vesicles of endocytic origin that are released into the extracellular environment on fusion of multivesicular bodies (MVB) with the plasma membrane 1. Many cells have the capacity to release exosomes, including reticulo-cytes 2 , dendritic cells 3 , B cells 4 , T cells 5 , mast cells 6 , epithelial cells 7 and tumour cells 8. The functions of exosomes are not completely understood, although it has been shown that exosomes can participate in the signalling events contributing to antigen presentation to T cells 4 and the development of tolerance 9. Several mechanisms have been hypothesized describing the interactions of exosomes and recipient cells. Exosomes can bind to cells through recep-tor–ligand interactions, similar to cell–cell communication mediating , for example, antigen presentation 4. Alternatively, exosomes could putatively attach or fuse with the target-cell membrane, delivering exosomal surface proteins and perhaps cytoplasm to the recipient cell 10,11. Finally, exosomes may also be internalized by the recipient cells by mechanisms such as endocytosis 12. Exosomes were isolated from a mast-cell line (MC/9), primary bone marrow-derived mast cells (BMMC) and a human mast-cell line (HMC-1) through a series of microfiltration and ultracentrifugation steps modified from what has been previously described 4. To confirm that the structures studied indeed are exosomes, they were examined by electron microscopy (Fig. 1a), flow cytometric analysis (FACS; Fig. 1b), and proteomic analysis (see Supplementary Information, Table S1). The electron micrographs of the exosomes revealed rounded structures with a size of approximately 50–80 nm, similar to previously described exo-somes 4,13–15. The identity of the studied vesicles was further confirmed as exosomes by FACS analysis (Fig. 1b), which show the presence of the surface protein CD63 — a commonly used marker of exosomes. Finally, extensive protein analysis of the MC/9 exosomes was performed on multiple samples using LC-MS/MS technology. A total of 271 proteins were identified (see Supplementary Information, Table S1) from three preparations of the isolated vesicles, of which 47 proteins were present in all three samples. More importantly, a large number of the proteins found in the preparations were the same as proteins previously identified in exosomes produced by other cells (that is, exosomes derived from intestinal epithelial cells, urine, dendritic cells, microglia, melanoma, T-cells and B-cells). In particular, 60% of the 47 proteins found in all samples of mast-cell exosomes have been previously found in other types of exosomes. Moreover, 39% of the 271 total proteins found in the analysed exosome samples have also been previously found in other types of exosomes. Thus, the electron microscopy, the FACS, and the detailed protein analyses each provided significant evidence in favour of the identification of the isolated vesicles as exosomes. The presence of nucleic acids was examined in exosomes derived from MC/9, BMMC and HMC-1 cells to define a potential mechanism by which exosomes may mediate cell–cell communication. These assessments showed that isolated exosomes contain no DNA (see Supplementary Information, Fig. S1). However, substantial amounts of RNA were detected by agarose gel electrophoresis, spectrophotometry
Article
Full-text available
Breast cancer (BC) is the most common cancer type and the second cause of cancer-related death among women. Therefore, better understanding of breast cancer tumor biology and the identification of novel biomarkers is essential for the early diagnosis and for better disease stratification and management choices. Herein we developed a novel approach which relies on the isolation of circulating microRNAs through an enrichment step using speed-vacuum concentration which resulted in 5-fold increase in microRNA abundance. Global miRNA microarray expression profiling performed on individual samples from 23 BC and 9 normals identified 18 up-regulated miRNAs in BC patients (p(corr) < 0.05). Nine miRNAs (hsa-miR-4270, hsa-miR-1225-5p, hsa-miR-188-5p, hsa-miR-1202, hsa-miR-4281, hsa-miR-1207-5p, hsa-miR-642b-3p, hsa-miR-1290, and hsa-miR-3141) were subsequently validated using qRT-PCR in a cohort of 46 BC and 14 controls. The expression of those microRNAs was overall higher in patients with stage I, II, and III, compared to stage IV, with potential utilization for early detection. The expression of this microRNA panel was slightly higher in the HER2 and TN compared to patients with luminal subtype. Therefore, we developed a novel approach which led to the identification of a novel microRNA panel which was upregulated in BC patients with potential utilization in disease diagnosis and stratification.
Article
Full-text available
Although African-Americans (AAs) have a higher incidence of colorectal cancer (CRC) than White people, the underlying biochemical mechanisms for this increase are poorly understood. The current investigation was undertaken to examine whether differences in self-renewing cancer stem/stem-like cells (CSCs) in the colonic mucosa, whose stemness is regulated by certain microRNAs (miRs), could partly be responsible for the racial disparity in CRC. The study contains 53 AAs and 47 White people. We found the number of adenomas and the proportion of CD44(+) CD166(- ) CSC phenotype in the colon to be significantly higher in AAs than White people. MicroRNAs profile in CSC-enriched colonic mucosal cells, expressed as ratio of high-risk (≥3 adenomas) to low-risk (no adenoma) CRC patients revealed an 8-fold increase in miR-1207-5p in AAs, compared to a 1.2-fold increase of the same in White people. This increase in AA was associated with a marked rise in lncRNA PVT1 (plasmacytoma variant translocation 1), a host gene of miR-1207-5p. Forced expression of miR-1207-5p in normal human colonic epithelial cells HCoEpiC and CCD841 produced an increase in stemness, as evidenced by morphologically elongated epithelial mesenchymal transition( EMT) phenotype and significant increases in CSC markers (CD44, CD166, and CD133) as well as TGF-β, CTNNB1, MMP2, Slug, Snail, and Vimentin, and reduction in Twist and N-Cadherin. Our findings suggest that an increase in CSCs, specifically the CD44(+) CD166(-) phenotype in the colon could be a predisposing factor for the increased incidence of CRC among AAs. MicroRNA 1207-5p appears to play a crucial role in regulating stemness in colonic epithelial cells in AAs.
Article
Full-text available
Recent evidences indicate that circulating microRNAs (miRNAs) exhibit aberrant expression in the plasma of patients suffering from cancer compared to normal individuals, suggesting that it may be a useful noninvasion diagnostic method. MiR-21 plays crucial roles in carcinogenesis and can be served as a biomarker for the detection of various cancers. Therefore, the aim of this meta-analysis is to assess the potential role of miR-21 for digestive system cancer. By searching the PubMed, Embase, and Web of Science for publications concerning the diagnostic value of miR-21 for digestive system cancer, total of 23 publications were included in this meta-analysis. Receiver operating characteristic curves (ROC) were used to check the overall test performance. For prognostic meta-analysis, pooled hazard ratios (HRs) of circulating miR-21 for survival were calculated. Totally 23 eligible publications were included in this meta-analysis (15 articles for diagnosis and 8 articles for prognosis). For diagnostic meta-analysis, the summary estimates revealed that the pooled sensitivity and specificity were 0.76(95% CI = 0.70–0.82) and 0.84 (95% CI = 0.78–0.89). Besides, the area under the summary ROC curve (AUC) is 0.87. For prognostic meta-analysis, the pooled HR of higher miR-21 expression in circulation was 1.94 (95% CI = 0.99–3.82, P = 0.055), which indicated higher miR-21 expression could be likely to predict poorer survival in digestive system cancer. The subgroup analysis implied the higher expression of miR-21 was correlated with worse overall survival in the Asian population in digestive system cancer (HR = 2.41, 95% CI = 1.21–4.77, P = 0.012). The current evidence suggests circulating miR-21 may be suitable to be a diagnostic and prognostic biomarker for digestive system cancer in the Asians.
Article
Background: Circulating RNA in plasma/serum is an emerging field for noninvasive molecular diagnosis. Because RNA is widely thought to be labile in the circulation, we investigated the stability and various preanalytical factors that may affect RNA concentrations in blood specimens. Methods: Blood samples were collected from 65 healthy volunteers. The effects of two preanalytical variables were studied: (a) time delay in processing of EDTA blood and clotted blood after venesection, and (b) freezing and thawing of plasma and serum. The lability of free added RNA in plasma was also investigated. Plasma/serum RNA was measured by a real-time quantitative reverse transcription-PCR assay for glyceraldehyde 3-phosphate dehydrogenase mRNA, whereas DNA was measured by a real-time quantitative PCR assay for the β-globin gene. Results: No significant difference was found for plasma RNA concentrations obtained from uncentrifuged EDTA blood that had been left at 4 °C for 0, 6, and 24 h (P =0.182). On the other hand, the serum RNA concentrations increased significantly over 24 h when uncentrifuged clotted blood was stored at 4 °C (P <0.05). In comparison, >99% of the free added RNA could no longer be amplified after incubation in plasma for 15 s. Never-frozen plasma, freeze-thawed plasma, and thawed plasma left at room temperature for 1 h showed no significant differences in RNA concentration (P =0.465). No significant difference was observed for freeze-thawed serum (P = 0.430). Conclusions: Plasma RNA is stable in uncentrifuged EDTA blood stored at 4 °C, but to obtain a stable serum RNA concentration, uncentrifuged clotted blood should be stored at 4 °C and processed within 6 h. A single freeze/thaw cycle produces no significant effect on the RNA concentration of plasma or serum.
Article
Fatty acid synthase (FASN) has emerged as a unique oncologic target for the treatment of cancers, including hepatocellular carcinoma (HCC). However, effective inhibitors of FASN for cancer treatment are lacking. MicroRNAs (miRNAs) have emerged as novel and endogenic inhibitors of gene expression. In the present study, we aimed to investigate the role of miR‑1207‑5p in HCC and the regulation of FASN through miR‑1207‑5p. The expression of miR-1207-5p was markedly reduced in HCC tissues and cell lines as detected with real‑time quantitative polymerase chain reaction (qPCR). Overexpression of miR-1207-5p significantly suppressed the cell growth and invasion of HCC cells. By contrast, inhibition of miR‑1207‑5p exhibited an opposite effect. Bioinformatics analysis showed that FASN is a predicted target of miR‑1207‑5p which was validated by dual‑luciferase reporter assay, qPCR and western blot analysis. Overexpression of miR‑1207‑5p inhibited the Akt/mTOR signalling pathway, and promotion of this pathway was noted following inhibition of miR‑1207‑5p. Rescue experiments showed that the restoration of FASN expression partially reversed the inhibitory effect of miR‑1207‑5p on cell growth, invasion and Akt phosphorylation. In conclusion, our study suggests that miR‑1207‑5p/FASN plays an important role in HCC, and provides novel insight into developing new inhibitors for FASN for therapeutic interventions for HCC.
Article
The expression and functions of microRNA (miR)-320 have been previously investigated in various types of cancer. However, to the best of our knowledge, no previous studies have investigated miR-320 in human lung cancer. The current study determined the expression, biological functions and molecular mechanisms of miR‑320 in human lung cancer. The expression level of miR‑320 in human non‑small cell lung cancer (NSCLC) and normal adjacent tissue samples (NATs), NSCLC cell lines and non‑tumorigenic bronchial epithelial cells was measured by reverse transcription‑quantitative polymerase chain reaction. Following transfection with miR‑320 mimics, 3‑(4,5‑dimethylthiazol‑2‑yl)‑2,5‑diphenyltetrazolium bromide, cell migration and cell invasion assays, western blot analysis and luciferase assay were performed in human NSCLC cell lines. The results demonstrated that miR‑320 was significantly downregulated in NSCLC tissue samples and cell lines compared with NATs and a control cell line, respectively. Statistical analysis demonstrated that expression of miR‑320 was significantly associated with the TNM classification and metastasis. It was also observed that miR‑320 inhibited cell growth, migration and invasion in NSCLC cells. Additionally, the present study provided evidence that miR‑320 may directly target fatty acid synthase. These results suggest that miR‑320 may serve as a therapeutic biomarker of NSCLC in the future.
Article
Objectives To evaluate and compare the ability of DW-MRI and CT to detect sites of peritoneal dissemination in gynecologic malignancies. The reproducibility of DW-MRI and CT interpretation between radiologists was also assessed. Methods Single institution prospective cohort study of women with suspected advanced gynecologic cancer who underwent surgical staging from 2010 to 2013. Participants underwent both DW-MRI and contrast-enhanced CT prior to surgery. Radiologists and surgeons were blinded, respectively, to surgical and DW-MRI results. The area under the receiver operator characteristic curve (AUC) was calculated for each disease site for CT and DW-MRI and compared to surgical findings. Kappa statistics quantified interobserver agreement between both radiologists. Results Twenty seven patients were enrolled. Mean age at surgery was 59 years. Ninety percent of participants had stage IIIC/IV disease. For right diaphragm disease, the AUC for DW-MRI was 0.95 compared to 0.81 for CT. For left diaphragm disease, the AUC was 0.89 for DW-MRI compared to 0.74 for CT. The AUC was similar for DW-MRI and CT for omental disease (0.79 versus 0.64); the liver surface (0.61 versus 0.67); bowel mesentery (0.73 versus 0.64); and cul de sac (0.75 versus 0.64). Interobserver agreement for DW-MRI was greater than CT for omental, Morrison's pouch, liver surface, and right diaphragm disease. Conclusions DW-MRI detects right diaphragmatic disease found at surgery with greater accuracy than CT. For other disease sites key to surgical planning, DW-MRI is equivalent to CT. Interobserver agreement was superior for a majority of disease sites evaluated by DW-MRI compared to CT.
Article
Peritoneal spread of tumors is a major problem in cancer management. Patients develop a marked deterioration in quality of life and shortened survival. This is in part due to bowel obstructions, marked ascites, and overall increase debilitation. Standard medical management has shown to be inadequate for the treatment of these problems. Surgery can palliate symptoms, however, it is unable to be complete at the microscopic level by a significant spillage of tumor cells throughout the abdomen. Chemotherapy can have some improvement in symptoms however it is short lived due to poor penetration into the peritoneal cavity. The role of intraperitoneal chemotherapy is to maximize tumor penetration and optimize cell death while minimizing systemic toxicity. Hyperthermic intraperitoneal chemotherapy (HIPEC) and early post-operative intraperitoneal chemotherapy (EPIC) are two treatment methods that serve this role and have been shown to improve survival. This review will discuss different chemotherapies used for both of these treatment options.
Article
Since the benefit of prostate-specific antigen (PSA) screening remains controversial, new non-invasive biomarkers for prostate carcinoma (PCa) are still required. There is evidence that microRNAs (miRNAs) in whole peripheral blood can separate patients with localized prostate cancer from healthy individuals. However, the potential of blood-based miRNAs for the differential diagnosis of PCa and benign prostatic hyperplasia (BPH) has not been tested. We compared the miRNome from blood of PCa and BPH patients and further investigated the influence of the tumor volume, tumor-node-metastasis (TNM) classification, Gleason score, pretreatment risk status, and the pretreatment PSA value on the miRNA pattern. By microarray approach, we identified seven miRNAs that were significantly deregulated in PCa patients compared to BPH patients. Using quantitative real time PCR (qRT-PCR), we confirmed downregulation of hsa-miR-221* (now hsa-miR-221-5p) and hsa-miR-708* (now hsa-miR-708-3p) in PCa compared to BPH. Clinical parameters like PSA level, Gleason score, or TNM status seem to have only limited impact on the overall abundance of miRNAs in patients' blood, suggesting a no influence of these factors on the expression of deregulated miRNAs.