ArticlePDF Available

Integrated In Silico Analysis of Proteogenomic and Drug Targets for Pancreatic Cancer Survival

Authors:

Abstract and Figures

Pancreatic cancer remains a major health concern, being among the deadliest forms of cancer with over 80% of the patients presenting with metastatic disease. According to the American Cancer Society, for all stages of pancreatic cancer combined, the 5-year survival rate is less than 10%. Genetic research on pancreatic cancer has generally been focused on familial pancreatic cancer, which is only 10% of all pancreatic cancer patients. This study focuses on finding genes that impact the survival of pancreatic cancer patients which can be used as biomarkers and potential targets to develop personalized treatment options. We used cBioPortal platform using NCI-initiated The Cancer Genome Atlas (TCGA) dataset to find genes that were altered differently in different ethnic groups which can serve as potential biomarkers and analyzed the genes' impact on patient survival. MD Anderson Cell Lines Project (MCLP) and genecards.org were also utilized to identify potential drug candidates that can target the proteins encoded by the genes. The results showed that there are unique genes that are associated with each race category which may influence the survival outcomes of patients, and their potential drug candidates were identified.
Content may be subject to copyright.
Chapter 19
Integrated In Silico Analysis of Proteogenomic and Drug
Targets for Pancreatic Cancer Survival
Alakesh Bera, Digonto Chatterjee, Jack Hester, and Meera Srivastava
Abstract
Pancreatic cancer remains a major health concern, being among the deadliest forms of cancer with over 80%
of the patients presenting with metastatic disease. According to the American Cancer Society, for all stages
of pancreatic cancer combined, the 5-year survival rate is less than 10%. Genetic research on pancreatic
cancer has generally been focused on familial pancreatic cancer, which is only 10% of all pancreatic cancer
patients. This study focuses on finding genes that impact the survival of pancreatic cancer patients which can
be used as biomarkers and potential targets to develop personalized treatment options. We used cBioPortal
platform using NCI-initiated The Cancer Genome Atlas (TCGA) dataset to find genes that were altered
differently in different ethnic groups which can serve as potential biomarkers and analyzed the genes’
impact on patient survival. MD Anderson Cell Lines Project (MCLP) and genecards.org were also utilized
to identify potential drug candidates that can target the proteins encoded by the genes. The results showed
that there are unique genes that are associated with each race category which may influence the survival
outcomes of patients, and their potential drug candidates were identified.
Key words Proteogenomics, Proteomics, Genomics, Survival outcomes, Drug targets, cBioPortal,
The Cancer Genome Atlas (TCGA), MD Anderson Cell Lines Project (MCLP), Copy number
alterations (CNAs), Socioeconomic status (SES), Disease-free survival (DFS), Overall survival (OS),
Pancreatic cancer
1 Introduction
Pancreatic cancer is among the deadliest forms of cancer. It is the
seventh most common cancer, yet it is the second leading cause of
cancer deaths in the United States [1]. It is estimated that in 2021,
48,220 patients will die from pancreatic cancer. Risk factors for
pancreatic cancer include smoking, diabetes, obesity, chronic pan-
creatitis, and family history [2]. Over 80% of the patients present
with metastatic disease. Despite advances in chemotherapy, the
average survival remains less than 5 years even after surgery [3].
Genetic testing has been primarily focused on familial pancreatic
cancer, which only accounts for ten percent (10%) of all pancreatic
Usha N. Kasid and Robert Clarke (eds.), Cancer Systems and Integrative Biology, Methods in Molecular Biology, vol. 2660,
https://doi.org/10.1007/978-1-0716-3163-8_19,
© The Author(s), under exclusive license to Springer Science+Busi ness Media, LLC, part of Springer Nature 2023
273
cancers. A study on non-familial pancreatic cancer patients revealed
six genes, namely, CDKN2A, TP53, MLH1, BRCA2, ATM, and
BRCA1, with significant associations between pancreatic cancer
and mutations in these genes [4]. A previous study has shown
that patient survival was associated with mutations in KRAS,
CDKN2A, SMAD4, and TP53 [5].
274 Alakesh Bera et al.
Similar to other common malignancies, pancreatic cancer is
associated with disparities by socioeconomic status (SES), ethnic
minority status, and insurance [6, 7]. In contrast to other types of
cancer (breast, colon) where screening can detect early-stage dis-
ease, no screening modality exists for pancreatic cancer. Thus,
disparities in outcomes for pancreatic cancer do not result from
lack of screening [8]. There are currently limited data on the
genetic susceptibility of pancreatic cancer survival based on race
or ancestral origin. The association of driver gene alterations based
on racial category and their association with patient outcomes has
not been clearly established. Therefore, we per formed a quantita-
tive genomic analysis to find genes associated with patient survival
for the pancreatic cancer patients based on their racial categories. In
this current study, we tried to establish a method to identify the
genetic basis of racial disparities in cancer survival. We tried to
determine whether White, African American/Black, and Asian
patients have different gene alterations and whether or not they
can be diagnosed and managed based on their specific signature
profile. Identification, prevention, and management of factors
based on race may help find effective strategies for clinical manage-
ment of pancreatic cancer.
2 Clinical Data from cBioPortal
cBioPortal was utilized to access and analyze the public database on
cancer tissues generated by the TCGA project (https://www.
cancer.gov/tcga). The Cancer Genome Atlas (TCGA) is an
NCI-initiated and established cancer genomic database which is
publicly available. There are over thirty thousand tumor samples
that have already been sequenced from over twenty different types
of cancer. The cBioPortal allows users to question datasets across
data types including genes and clinical samples, providing an oppor-
tunity to investigate several different biologically and/or clinically
relevant hypotheses. For this study, all datasets for pancreatic cancer
were selected particularly for patient’s survival with genetic signa-
ture. Data also included the specific patient’s number and their
contributions from different studies. Customized datasets were
created from Pancreatic Cancer Studies in cBioPortal along with
age, race, and sex distributions of patients (Fig. 1). Datasets that
did not provide race information were not included in this analysis.
Proteogenomic Analysis of Health Disparities in Pancreatic Cancer 275
Fig. 1 Different studies included in this analysis to study the pancreatic cancer patient’s survival with genetic
signature in particular. (a, b) Data also included the specific patient’s number and their contributions from
different studies. (c, d) Customized dataset created from Pancreatic Cancer Studies in cBioPortal. (c) Data
indicated the age distributions of patients. (d) Male vs female ratio
3 Methods
3.1 Copy Number
Alterations
Copy number alteration data was analyzed in each of three racial
categories, (1) White, (2) African American and Black, and
(3) Asian with customized datasets. Within those separate virtual
studies, the genes that were amplified in the most patients in terms
of CNA (copy number alterations) were recorded and compared
among the different virtual datasets.
3.2 Survival Curves Genes that were altered differently in the racial groups were
selected to analyze the genes’ impact on patient survival. Both
overall survival (OS) and disease-free survival (DFS) curves were
computed. This was done by utilizing the inbuilt statistical analysis
tools available within the cBioPortal platform. The data was tested
for significance using a log-rank test to compare the survival dis-
tributions between two samples. This was done automatically using
cBioPortal [9, 10].
276 Alakesh Bera et al.
3.3 Protein Drug
Analysis
MD Anderson Cell Lines Project (MCLP) and genecards.org were
utilized to find protein drug interactions using the genes found in
this study as drug targets [11, 12].
3.4 Pancreatic
Cancer Categories
More than 1200 patient data spanning ten [10] different pancreatic
cancer clinical studies were analyzed. The disease types were (1) aci-
nar cell carcinoma of the pancreas, (2) cystic tumor of the pancreas,
(3) five pancreatic adenocarcinoma studies, and (4) three pancreatic
neuroendocrine tumor studies (Fig. 1). The data were stratified
according to three race categories self-identified by patients:
(1) White, (2) African American or Black, and (3) Asian. There
were only two samples that were identified as Hispanic; therefore
the Hispanic category was not included in our study. A total of
436 patient samples were customized based on three (3) race cate-
gories. The samples were from three pancreatic adenocarcinoma
studies. For our analysis, Black or African American and Black were
taken together. The number of samples for White patients was
394, 22 for Asian, and 19 for Black and African American. There
are several studies pursued to determine the racial disparities in
cancer survival. However, it is not clear whether the disparity is
related to biological differences or consequences of social, eco-
nomic, or cultural environments. Given the challenges associated
with race and ethnicity definition and disease-linked biological
observations in this context, results from our study need to be
independently validated in larger patient population with ancestry
information.
3.5 Data Analysis The analysis began with an investigation of copy number alterations
(CNA) at the genomic DNA level for pancreatic cancer samples.
Copy number alterations (CNAs) refer to changes in the number of
copies of a genetic region caused by deletion or duplication events
in the genome. Recent technological advances have enabled the
identification of CNAs across the entire genome, associating cancer
with the alteration rates of various genes in cancer [8].
3.5.1 Copy Number
Alterations Based on Race
Categories
CBioPortal was utilized, which contains comprehensive geno-
mic and transcriptomic data from various cancer studies
[13]. Focusing on the customized pancreatic cancer datasets cre-
ated based on race categories as mentioned in the above section,
genes with the highest frequency of copy number alterations
(CNA) in each race category were determined. A few unique
genes specific to each race category that had copy number amplifi-
cations were found (Table 1). The altered genes’ locations are in
different cytobands for each race category. As previously reported,
Race Cytoband
CDKN2A was deleted in 30% of patients in every race category [5],
confirming our data analysis approach. Most genes that had deep
deletions in copy number were common among the different race
categories and therefore were not included in further investigation
and analysis.
Proteogenomic Analysis of Health Disparities in Pancreatic Cancer 277
Table 1
Genes with copy number alterations unique to each race category
Genes with copy number alterations
(AMP)
Alteration frequency
(%)
White GATA6, RECQL4, MIB1 18q11.2 (GATA5, MIB1)
8q24.3 (RECQL4)
14.8 (GATA6)
14.0(RECQL4)
12.8%(MIB1)
Black PKDL1, GARS1, NEUROD3 7p12.3(PKDL1)
7p14.3(GA RS1,
NEUROD3)
28.6
Asian FGFR3, ABCA11P, UVSSA (etc.) 4p16.3 27.3
3.5.2 Association of CNA
Amplification with Survival
Outcome
An evaluation was carried out between the association of the gene
alterations unique to each category and most frequently amplified
in each race category with survival outcome. The most frequently
altered genes in each category were found to have a direct associa-
tion with patient survival. KaplanMeyer curves for overall survival
(OS) and disease-free survival (DFS) were generated using the
cBioPortal platform for each gene in the specific race categories.
P-values were generated by cBioPortal using the log-rank test for
survival between 2 samples. This was compared to a significance
level of 0.05. The survival was calculated in median months, and
the 95% confidence interval was given as well.
Figure 2 shows representative KaplanMeier curves for disease-
free survival (DFS) outcomes associated with alterations of genes in
patients from different race categories. The P-values are 0.0031 for
White patients, 0.0516 for Asian patients, and 0.8680 for African
American and Black patients.
The results showed that there are unique genes that are asso-
ciated with each race category which have an effect on the survival
outcomes of patients. For patients that had expression of these
genes, the median month of survival was less compared to those
who did not have amplified expression.
White Patients For the genes amplified in the White race category,
the effect on OS was minimal while the DFS was significantly worse
than in patients where there was no amplification with a p-value of
0.0031. The genes amplified in the White race category were from
the cytoband 18p11.2 (GATA6, MIB1) and 8q24.3 (RECQL4). It
is notable that the genes that were from the same cytoband showed
nearly identical data.
Genes altered
278 Alakesh Bera et al.
Fig. 2 KaplanMeyer curves for disease-free survival in each race category. Statistical values (P-value) are
included in the figure
Table 2
Survival outcomes (in months) associated with altered genes in each race category
Race
category
Overall
Survival
(OS) unaltered
Overall
Survival
(OS) altered
Disease Free
Survival (DFS)-
unaltered
Disease Free
Survival (DFS)-
altered
GATA6 20.17 20.35 20.4 12.43
White RECQL4 20.35 15.11 20.37 9.57
MIB1 20.19 20.34 23.52 12.42
African
American
& black
PKDIL1.
GARS1,
NEUROD6
17.03 2.01 NA NA
Asian FGFR3.
ABCA11P,
UVSSA. (etc.)
66.89 11.61 12.32 49.68
Asian Patients For genes amplified in the Asian race category, the
survival outcomes for patients with amplified genes was signifi-
cantly worse for both overall survival and disease-free survival com-
pared to patients who did not have amplified genes with a p-value of
0.0516. The genes in the Asian race category were all from the same
cytoband (4p16.3), and while only 3 were listed in Table 2, there
were 39 genes from the cytoband which were all amplified and
showed the same survival outcomes.
African American and Black Patients For genes amplified in the
African American and Black race category, the effect on overall
survival for patients that had amplified genes in the 7p cytoband
had significantly poor outcomes (2 months) with alterations in the
genes PKD1L1, GARS1, and NEUROD6 compared to patients
with no amplification (17 months). There was no data for disease-
Drug candidates
free survival due to the limited number of samples. However, it is
notable to mention that data for the African American and Black
categories were limited (African American/Black, n = 19) and were
not statistically significant.
Proteogenomic Analysis of Health Disparities in Pancreatic Cancer 279
Table 3
Role of the genes in cancers and as potential drug candidates
Genes/race
categories
Role in
pancreatic
cancer
Role in other cancers (prognostic
value)
GATA6
(White)
No Renal (unfavorable) (detected in
many)
Spautin.l; parbendazole
RECQL4
(White)
No Liver (unfavorable) (detected in all) Rec 15/2615 dihydrochloride
MIB1 (White) No None (detected in all) SIB 1893
PKD1L1
(Black)
No None (detected in many) JKC 363; HS 014
GARS1
(Black)
NA NA MRS 1220; RS 100329
hydrochloride
NEUROD6
(Black)
No None (not detected) SB 224289 hydrochloride;
Pancuronium dibromide
FGFR3
(Asian)
No Endometrial (unfavorable)
(detected in many)
PD 173074
ABCA11P
(Asian)
NA NA Bobcat339, TBCA
UVSSA
(Asian)
No Renal (unfavorable) urothelial
(favorable) detected in all
Iressa, ASB 14780
3.5.3 Protein Expression
and ProteinDrug
Interaction
Using the Protein Atlas, proteindrug interaction from MD Ander-
son Cancer-Cell Lines Project (MCLP) dataset [11, 12] and
genecards.org data, an analysis of the role of the genes in cancers
that are the focus of our study was done, and drug candidates that
can target the proteins encoded by the genes were determined.
Potential drug candidates that can be further tested in clinical trials
to provide gene and race specific therapeutic options for pancreatic
cancer patients were proposed. Table 3 shows the role of the genes
in cancers, their role as prognostic markers, and drugs that can be
used to target these genes. The top-ranked 2 drugs targeting each
gene were included.
280 Alakesh Bera et al.
4 Notes
1. This study demonstrates that there are unique sets of genes
commonly associated with self-identified race.
2. Alterations in these genes are associated with patient outcomes
in patients with pancreatic cancer.
3. These data if validated in large population with ancestry infor-
mation may serve as potential biomarkers for specific race
groups to predict patient outcomes.
4. Additionally, several potential drug candidates proposed in our
study targeting the specific proteins encoded by the genes
specific to the individual race categories can be further studied
in clinical settings to develop more personalized race-based
therapeutic options.
5. Besides, understanding the molecular events and mechanisms
that determine patient outcomes has the potential to develop
new and improved treatment approaches for patients with
pancreatic cancer.
6. These results are correlated by other studies, as GATA6 is now
a known oncogene [14], meaning it has the potential to cause
cancer. Similarly, studies have shown copy number alterations
in the cytoband 8q24.3 having a role in cancer [1517].
7. The role of genes and their respective cytobands from the Black
and African American as well as Asian race category was more
novel. Of the genes found to be amplified in these two race
categories, only FGFR3 is known to be an oncogene [18].
8. It is notable that the cytoband 4p16.3 which was amplified
frequently in the Asian race category, showing 39 genes all
with frequent alteration, causes WolfHirschhorn syndrome
when deleted [19]; however, there has been little investigation
on its amplification and no study on its unique amplification in
the Asian race category or pancreatic cancer.
9. The biggest shortcoming was that the sample size was low,
especially for the Black and African American and Asian race
categories. Despite this, the results can be used as a good
starting point for further, more robust investigation.
10. Future goals include developing new and improved treatment
for patients with pancreatic cancer, potentially by targeting the
genes that were noted in this study.
Proteogenomic Analysis of Health Disparities in Pancreatic Cancer 281
11. Another potential goal would be to do statistical modeling and
larger population study to determine if these genes can be used
for diagnosis of pancreatic cancer, which would be especially
noteworthy as pancreatic cancer is known to be hard to diag-
nose early.
12. Furthermore, the specific role of each cytoband that was largely
amplified and its association with pancreatic cancer can be
investigated.
Acknowledgements
Alakesh Bera and Digonto Chatterjee contributed as co-equal first
authors on this study.
Disclaimer The opinions or assertions contained herein are the
private ones of the authors and are not to be construed as official or
reflecting the views of the Depar tment of Defense, the Uniformed
Services University of the Health Sciences, or any other agency of
the U.S. Government.
References
1. Siegel RL, Miller KD (2019) Jemal A. Cancer
statistics. CA Cancer J Clin 69(1):734.
https://doi.org/10.3322/caac.21551
2. Ryan DP, Hong TS, Bardeesy N (2014) Pan-
creatic adenocarcinoma. N Engl J Med
371(22):21402141. https://doi.org/10.
1056/NEJMc1412266
3. Noel M, Fiscella K (2019) Disparities in pan-
creatic cancer treatment and outcomes. Health
Equity 3(1):532540. https://doi.org/10.
1089/heq.2019.0057
4. Hu C, Hart SN, Polley EC et al (2018) Associ-
ation between inherited germline mutations in
cancer predisposition genes and risk of pancre-
atic cancer. JAMA 319(23):24012409.
https://doi.org/10.1001/jama.2018.6228
5. Qian ZR, Rubinson DA, Nowak JA et al
(2018) Association of alterations in main driver
genes with outcomes of patients with resected
pancreatic ductal adenocarcinoma. JAMA
Oncol 4(3):e173420. https://doi.org/10.
1001/jamaoncol.2017.3420
6. Khawja SN, Mohammed S, Silberfein EJ et al
(2015) Pancreatic cancer disparities in African
Americans. Pancreas 44(4):522527. https://
doi.org/10.1097/MPA.0000000000000323
7. Shapiro M, Chen Q, Huang Q et al (2016)
Associations of socioeconomic variables with
resection, stage, and survival in patients with
early-stage pancreatic cancer. JAMA Surg
151(4):338345. https://doi.org/10.1001/
jamasurg.2015.4239.
8. Zhang Q, Zeng L, Chen Y et al (2016) Pancre-
atic cancer epidemiology, detection, and man-
agement. Gastroenterol Res Pract 8962321.
https://doi.org/10.1155/2016/8962321
9. Cerami E, Gao J, Dogrusoz U et al (2012) The
cBio cancer genomics portal: an open platform
for exploring multidimensional cancer geno-
mics data. Cancer Discov 2(5):401404.
https://doi.org/10.1158/2159-8290.CD-
12-0095
10. Gao J, Aksoy BA, Dogrusoz U et al (2013)
Integrative analysis of complex cancer geno-
mics and clinical profiles using the cBioPortal.
Sci Signal 6(269):11. https://doi.org/10.
1126/scisignal.2004088
11. Chen MM, Li J, Mills GB, Liang H (2020)
Predicting cancer cell line dependencies from
the protein expression data of reverse-phase
protein arrays. JCO Clin Cancer Inform 4:
357366. https://doi.org/10.1200/CCI.19.
00144
12. Li J, Zhao W, Akbani R et al (2017) Character-
ization of human cancer cell lines by reverse-
phase protein arrays. Cancer Cell 31(2):
282 Alakesh Bera et al.
225239. https://doi.org/10.1016/j.ccell.
2017.01.005
13. Bera A, Subramanian M, Karaian J et al (2020)
Functional role of vitronectin in breast cancer.
PLoS One 15(11):e0242141. https://doi.
org/10.1371/journal.pone.0242141
14. Deng X, Jiang P, Chen J et al (2020) GATA6
promotes epithelial-mesenchymal transition
and metastasis through MUC1/beta-catenin
pathway in cholangiocarcinoma. Cell Death
Dis 11(10):860. https://doi.org/10.1038/
s41419-020-03070-z
15. Bera A, Radhakrishnan S, Russ E et al (2022)
Functional role of long non-coding RNA
YTHDF3-AS1 in breast cancer. Cancer Res
82(12_Suppl):5828
16. Bera A, Karaian J, Subramanian M et al (2020)
EXOSC4, a novel gene at chromosome 8q24
loci is linked with breast cancer progression and
is a prognostic marker for breast cancer sur-
vival. Cancer Res 80(16_Supplement):4322
17. Brusselaers N, Ekwall K, Durand-Dubief M
(2019) Copy number of 8q24.3 drives HSF1
expression and patient outcome in cancer: an
individual patient data meta-analysis. Hum
Genomics 13(1):54. https://doi.org/10.
1186/s40246-019-0241-3
18. Zingg D, Bhin J, Yemelyanenko J et al (2022)
Truncated FGFR2 is a clinically actionable
oncogene in multiple cancers. Nature 608
(7923):609617
19. Zollino M, Orteschi D, Ruiter M et al (2014)
Unusual 4p16.3 deletions suggest an addi-
tional chromosome region for the Wolf-
Hirschhorn syndrome-associated seizures dis-
order. Epilepsia 55(6):849857
Article
The insights provided by the holistic approaches of systems and integrative biology offer a means to address the multiple levels of complexity evident in cancer biology. Often focused on the use of large-scale, high-dimensional omics data for discovery in silico, integration with lower-dimensional data and lower-throughput wet laboratory studies allows for the development of a more mechanistic understanding of the control, execution, and operation of complex biological systems. While no single volume can cover all of the advances across this broad and rapidly developing field, we here provide reviews, methods, and detailed protocols for several state-of-the-art approaches to probe cancer biology from an integrative systems perspective. The protocols presented are intended for easy implementation in the laboratory and often offer a clear rationale for their development and application. This introduction provides a very brief description of systems and integrative biology as context for the chapters that follow, with a short overview of each chapter to allow the reader to easily and quickly find those protocols of most interest.
Article
Full-text available
Somatic hotspot mutations and structural amplifications and fusions that affect fibroblast growth factor receptor 2 (encoded by FGFR2) occur in multiple types of cancer1. However, clinical responses to FGFR inhibitors have remained variable1–9, emphasizing the need to better understand which FGFR2 alterations are oncogenic and therapeutically targetable. Here we apply transposon-based screening10,11 and tumour modelling in mice12,13, and find that the truncation of exon 18 (E18) of Fgfr2 is a potent driver mutation. Human oncogenomic datasets revealed a diverse set of FGFR2 alterations, including rearrangements, E1–E17 partial amplifications, and E18 nonsense and frameshift mutations, each causing the transcription of E18-truncated FGFR2 (FGFR2ΔE18). Functional in vitro and in vivo examination of a compendium of FGFR2ΔE18 and full-length variants pinpointed FGFR2-E18 truncation as single-driver alteration in cancer. By contrast, the oncogenic competence of FGFR2 full-length amplifications depended on a distinct landscape of cooperating driver genes. This suggests that genomic alterations that generate stable FGFR2ΔE18 variants are actionable therapeutic targets, which we confirmed in preclinical mouse and human tumour models, and in a clinical trial. We propose that cancers containing any FGFR2 variant with a truncated E18 should be considered for FGFR-targeted therapies. Truncation of exon 18 of FGFR2 (FGFR2ΔE18) is a potent driver mutation in mice and humans, and FGFR-targeted therapy should be considered for patients with cancer expressing stable FGFR2ΔE18 variants.
Article
Full-text available
Breast Cancer is the most common form of cancer in women worldwide, impacting nearly 2.1 million women each year. Identification of new biomarkers could be key for early diagnosis and detection. Vitronectin, a glycoprotein that is abundantly found in serum, extracellular matrix, and bone, binds to integrin αvβ3, and promotes cell adhesion and migration. Current studies indicate that patients with amplified vitronectin levels have lower survival rates than patients without amplified vitronectin levels. In this study, we focused on the role of vitronectin in breast cancer survival and its functional role as a non-invasive biomarker for early stage and stage specific breast cancer detection. To confirm that the expression of vitronectin is amplified in breast cancer, a total of 240 serum samples (n = 240), 200 from breast cancer patients and 40 controls were analyzed using the Reverse Phase Protein Array (RPPA) technique. Of the 240 samples, 120 samples were of African American (AA) descent, while the other 120 were of White American (WA) descent. Data indicated that there were some possible racial disparities in vitronectin levels and, differences also seen in the recurrent patient samples. Next, we tried to uncover the underlying mechanism which plays a critical role in vitronectin expression. The cellular data from four different breast cancer cell lines- MCF7, MDA-MB-231, MDA-MB-468, and HCC1599 indicated that the PI3K/AKT axis is modulating the expression of vitronectin. We believe that vitronectin concentration levels are involved and connected to the metastasis of breast cancer in certain patients, specifically based on recurrence or ethnicity, which is detrimental for poor prognosis. Therefore, in this current study we showed that the serum vitronectin levels could be an early marker for the breast cancer survival and we also determine the cellular signaling factors which modulate the expression and concentration of vitronectin.
Article
Full-text available
GATA6 acts as an oncogene or tumour suppressor in different cancers. Previously, we found that aberrant expression of GATA6 promoted metastasis in cholangiocarcinoma (CCA). However, the mechanism by which GATA6 promotes metastasis in CCA is unclear. In the present study, we aimed to investigate the role of GATA6 in CCA cell epithelial–mesenchymal transition (EMT). Our results showed that GATA6 expression was positively associated with N-cadherin and vimentin expression but negatively associated with E-cadherin expression in 91 CCA samples. GATA6 promoted EMT and metastasis in CCA cells in vitro and in vivo based on knockdown and overexpression analyses. ChIP-sequencing data revealed that MUC1 is a novel downstream target of GATA6. GATA6 upregulated MUC1 expression through binding to both the 1584 and 1456 GATA-motifs in the promoter region and enhancing its transcription by luciferase reporter assays and point-mutant assays. MUC1 expression was positively associated with N-cadherin and vimentin expression but negatively associated with E-cadherin expression in 91 CCA samples. In addition, MUC1 promoted EMT in CCA cells based on knockdown and overexpression analyses. Moreover, MUC1 knockdown significantly abrogated the GATA6-induced EMT in CCA cells, indicating that MUC1 promoted EMT through upregulating MUC1 in CCA cells. β-Catenin is a putative transcriptional coactivator that regulates EMT in cancers. Our data showed that MUC1 expression was positively associated with nuclear β-catenin expression in 91 CCA samples. MUC1 upregulated nuclear β-catenin expression in CCA cells. Moreover, MUC1 bound to β-catenin in CCA cells based on protein immunoprecipitation analyses. MUC1 knockdown significantly decreased the binding of MUC1 to β-catenin, and thereby decreased nuclear β-catenin protein levels in CCA cells, indicating that MUC1 bound to β-catenin and increased its nuclear expression in CCA cells. Together, our results show that GATA6 promotes EMT through MUC1/β-catenin pathway in CCA, indicating potential implications for anti-metastatic therapy.
Article
Full-text available
PURPOSE Predicting cancer dependencies from molecular data can help stratify patients and identify novel therapeutic targets. Recently available data on large-scale cancer cell line dependency allow a systematic assessment of the predictive power of diverse molecular features; however, the protein expression data have not been rigorously evaluated. By using the protein expression data generated by reverse-phase protein arrays, we aimed to assess their predictive power in identifying cancer dependencies and to develop a related analytic tool for community use. MATERIALS AND METHODS By using a machine learning schema, we conducted an analysis of feature importance based on cancer dependency and multiomic data from the DepMap and Cancer Cell Line Encyclopedia projects. We assessed the consistency of cancer dependency data between CRISPR/Cas9 and short hairpin RNA–mediated perturbation platforms. For a fair comparison, we focused on a set of genes with robust dependency data and four available expression-related features (copy number alteration, DNA methylation, messenger RNA expression, and protein expression) and performed the same-gene predictions of the cancer dependency using different molecular features. RESULTS For the genes surveyed, we observed that the protein expression data contained substantial predictive power for cancer dependencies, and they were the best predictive feature for the CRISPR/Cas9-based dependency data. We also developed a user-friendly protein-dependency analytic module and integrated it with The Cancer Proteome Atlas; this module allows researchers to explore and analyze our results intuitively. CONCLUSION This study provides a systematic assessment for predicting cancer dependencies of cell lines from different expression-related features of a gene. Our results suggest that protein expression data are a highly valuable information resource for understanding tumor vulnerabilities and identifying therapeutic opportunities.
Article
Full-text available
Background: The heat-shock transcription factor 1 (HSF1) has been linked to cell proliferation and survival in cancer and has been proposed as a biomarker for poor prognosis. Here, we assessed the role of HSF1 expression in relation to copy number alteration (CNA) and cancer prognosis. Methods: Using 10,287 cancer genomes from The Cancer Genome Atlas and Cbioportal databases, we assessed the association of HSF1 expression with CNA and cancer prognosis. CNA of 8q24.3 was categorized as diploid (reference), deletion (fewer copies), gain (+ 1 copy) and amplification (≥ + 2 copies). Multivariate logistic regression modeling was used to assess 5-year survival among those with a first cancer diagnosis and complete follow-up data (N = 9568), categorized per anatomical location and histology, assessing interaction with tumor stage, and expressed as odds ratios and 95% confidence intervals. Results: We found that only 54.1% of all tumors have a normal predicted 8q24.3 copy number and that 8q24.3 located genes including HSF1 are mainly overexpressed due to increased copies number of 8q24.3 in different cancers. The tumor of patients having respectively gain (+ 1 copy) and amplification (≥ + 2 copies) of 8q24.3 display a global increase of 5-year mortality (odds ratio = 1.98, 95% CI 1.22-3.21) and (OR = 2.19, 1.13-4.26) after full adjustment. For separate cancer types, tumor patients with 8q24.3 deletion showed a marked increase of 5-year mortality in uterine (OR = 4.84, [2.75-8.51]), colorectal (OR = 4.12, [1.15-14.82]), and ovarian (OR = 1.83, [1.39-2.41]) cancers; and decreased mortality in kidney cancer (OR = 0.41, [0.21-0.82]). Gain of 8q24.3 resulted in significant mortality changes in 5-year mortality for cancer of the uterus (OR = 3.67, [2.03-6.66]), lung (OR = 1.76, [1.24-2.51]), colorectal (OR = 1.75, [1.32-2.31]) cancers; and amplification for uterine (OR = 4.58, [1.43-14.65]), prostate (OR = 4.41 [3.41-5.71]), head and neck (OR = 2.68, [2.17-3.30]), and stomach (OR = 0.56, [0.36-0.87]) cancers. Conclusions: Here, we show that CNAs of 8q24.3 genes, including HSF1, are tightly linked to 8q24.3 copy number in tumor patients and can affect patient outcome. Our results indicate that the integration of 8q24.3 CNA detection may be a useful predictor for cancer prognosis.
Article
Full-text available
Purpose: Pancreatic cancer remains a major health concern; in the next 2 years, it will become the second leading cause of cancer deaths in the United States. Health disparities in the treatment of pancreatic cancer exist across many disciplines, including race and ethnicity, socioeconomic status (SES), and insurance. This narrative review discusses what is known about these disparities, with the goal of highlighting targets for equity promoting interventions. Methods: We performed a narrative review of health disparities in pancreatic cancer spanning greater than ten areas, including epidemiology, treatment, and outcome, using the PubMed NIH database from 2000 to 2019 in the Unites States. Results: African Americans (AAs) tend to present at diagnosis with later stage disease. AAs and Hispanics have lower rates of surgical resection, are more likely to be treated at low volume hospitals, and often experience higher rates of morbidity and mortality compared to white patients, although control for confounders is often limited. Insurance and SES also factor into the delivery of treatment for pancreatic cancer. Conclusion: Disparities by race and SES exist in the diagnosis and treatment of pancreatic cancer that are largely driven by race and SES. Improved understanding of underlying causes could inform interventions.
Article
Full-text available
Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States and compiles the most recent data on cancer incidence, mortality, and survival. Incidence data, available through 2015, were collected by the Surveillance, Epidemiology, and End Results Program; the National Program of Cancer Registries; and the North American Association of Central Cancer Registries. Mortality data, available through 2016, were collected by the National Center for Health Statistics. In 2019, 1,762,450 new cancer cases and 606,880 cancer deaths are projected to occur in the United States. Over the past decade of data, the cancer incidence rate (2006‐2015) was stable in women and declined by approximately 2% per year in men, whereas the cancer death rate (2007‐2016) declined annually by 1.4% and 1.8%, respectively. The overall cancer death rate dropped continuously from 1991 to 2016 by a total of 27%, translating into approximately 2,629,200 fewer cancer deaths than would have been expected if death rates had remained at their peak. Although the racial gap in cancer mortality is slowly narrowing, socioeconomic inequalities are widening, with the most notable gaps for the most preventable cancers. For example, compared with the most affluent counties, mortality rates in the poorest counties were 2‐fold higher for cervical cancer and 40% higher for male lung and liver cancers during 2012‐2016. Some states are home to both the wealthiest and the poorest counties, suggesting the opportunity for more equitable dissemination of effective cancer prevention, early detection, and treatment strategies. A broader application of existing cancer control knowledge with an emphasis on disadvantaged groups would undoubtedly accelerate progress against cancer.
Article
Breast cancer (BrCa) is the most common form of cancer in women worldwide, impacting nearly 2.1 million women each year. Importantly, current treatment options for metastatic breast cancer fail to elicit an anti-tumor response, leading to a 5-year survival rate below 30%. While most classes of therapy focus on inhibiting or targeting specific proteins, long non-coding RNAs (lncRNAs) offer a unique, relatively untapped, source of potential therapeutic targets. Ongoing experiments are evaluating the regulatory role of the lncRNA YTHDF3-AS1 (ENSG00000270673), which has the potential to develop into a novel therapeutic intervention of BrCa. Through genome-wide analysis, we found that YTHDF3-AS1 expression correlates with essential developmental regulatory genes such as CA3, VPS28, EXOSC4, and TM2D2 and according to pathway analysis is likely linked with the Wnt pathway. Additionally, TCGA data reveals that over 5% of patients with BrCa have an increased copy number of YTHDF3-AS1. Although the role of YTHDF3-AS1 has yet to be explored, YTHDF3 expression was recently shown to be a critical driver of breast cancer metastasis. Of note, YTHDF3-AS1 is upstream of the protein-coding region, but within the promoter region of the YTHDF3 gene. Mechanistically, this allows the lncRNA YTHDF3-AS1 to potentially upregulate the expression of the protein-coding YTHDF3 gene, as two known functions of antisense lncRNAs describe the ability to 1) induce nearby gene expression through binding to the promoter region and 2) stabilize the RNA product of the nearby gene through binding to the 5’UTR. Interestingly, this manner of relationship between antisense (or "divergent") lncRNAs and their protein-coding partner was shown to be highly prevalent in pluripotent stem cells, with the downregulation of both the antisense and protein-coding transcripts following differentiation. Together, our preliminary analysis and previous literature suggest that the lncRNA YTHDF3-AS1 may contribute to the upregulation of YTHDF3, which is a known metastatic promoting protein. Our current study aims to determine the functional role of YTHDF3-AS1 in BrCa oncogenesis, with an emphasis on the mechanistic actions of this lncRNA for the development of future therapeutic approaches. Citation Format: Alakesh Bera, Surya Radhakrishnan, Eric Russ, Diya Biswas, Madhan Subramanian, Harvey B. Pollard, Hai Hu, Craig D. Shriver, Roopa Biswas, Meera Srivastava. Functional role of long non-coding RNA YTHDF3-AS1 in breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5828.
Conference Paper
p>The chromosome 8q24 gene contains functional risk loci for multiple cancers, including prostate, colon, and breast. Breast cancer (BrCa) is one of the deadliest diseases among women. Our previous studies using TCGA clinical survival data (Cell. 2018; 173(2):400-416.e11) indicated that several genes are contributed significantly towards the survival of patients with BrCa. Over 75 such most significant genes were screened for further studies in terms of their contribution towards BrCa patient9s survival. We found two genes (EXOSC4 and VPS28) with over 10% genetic copy number alterations located at chromosome 8q24 gene loci. Exosome Component 4 (EXOSC4) is located at 8q24.3 and associated with a non-catalytic component of the RNA exosome complex, which has 39 to 59 exoribonuclease activity. It also participates in cellular RNA processing and degradation events. However, the functional role of EXOSC4 towards BrCa progression or survival remains unknown. To understand the molecular basis of an elevated level of EXOSC4 in BrCa propagation, we screened the expression of this protein in different BrCa cell-lines. We also compared the expression of VPS28 within same cell lines. Data indicated that EXOSC4 was co-expressed with VPS28 and Myc, and was also secreted into extracellular space. Therefore, we next evaluated the level of EXOSC4 in patient9s serum samples by reverse phase protein array (RPPA). We have well defined and categorized BrCa patients serum samples (n=240) from the Clinical Breast Care Project. The data indicated that the elevated level of EXOSC4 is associated with worse overall survival. In addition, results also showed that the high level of EXOSC4 is linked with breast cancer recurrence.In summary, through multiple modality studies we have confirmed the copy number alteration of EXOSC4 is associated with BrCa patient survival. Once validated, serum level of EXOSC4 could be a predictive biomarker for BrCa progression and survival. Additional experiments are ongoing to evaluate the regulatory role of the EXOSC4 gene which has the potential to develop into a novel therapeutic intervention of BrCa. Funding Sources: This work was supported by the grant to Dr. Meera Srivastava (DoD, DAMD17-03-1-0107). The work was also supported by the Collaborative Health Initiative Research Program (CHIRP) funding (CHIRP ID# IAA-A-HL-14-007). Disclaimers: The contents of this publication are the sole responsibility of the author(s) and do not necessarily reflect the views, opinions or policies of The Uniformed Services University of the Health Sciences (USUHS), The Henry M. Jackson Foundation (HJF), the Department of Defense (DoD), the Departments of the Army, Navy, or Air Force. Correspondence: Email: meera.srivastava@usuhs.edu Citation Format: Alakesh Bera, John Karaian, Madhan Subramanian, Michael Eklund, Eric Russ, Harvey B. Pollard, Hai Hu, Craig D. Shriver, Meera Srivastava. EXOSC4, a novel gene at chromosome 8q24 loci is linked with breast cancer progression and is a prognostic marker for breast cancer survival [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 4322.</p
Article
Importance Individuals genetically predisposed to pancreatic cancer may benefit from early detection. Genes that predispose to pancreatic cancer and the risks of pancreatic cancer associated with mutations in these genes are not well defined. Objective To determine whether inherited germline mutations in cancer predisposition genes are associated with increased risks of pancreatic cancer. Design, Setting, and Participants Case-control analysis to identify pancreatic cancer predisposition genes; longitudinal analysis of patients with pancreatic cancer for prognosis. The study included 3030 adults diagnosed as having pancreatic cancer and enrolled in a Mayo Clinic registry between October 12, 2000, and March 31, 2016, with last follow-up on June 22, 2017. Reference controls were 123 136 individuals with exome sequence data in the public Genome Aggregation Database and 53 105 in the Exome Aggregation Consortium database. Exposures Individuals were classified based on carrying a deleterious mutation in cancer predisposition genes and having a personal or family history of cancer. Main Outcomes and Measures Germline mutations in coding regions of 21 cancer predisposition genes were identified by sequencing of products from a custom multiplex polymerase chain reaction–based panel; associations of genes with pancreatic cancer were assessed by comparing frequency of mutations in genes of pancreatic cancer patients with those of reference controls. Results Comparing 3030 case patients with pancreatic cancer (43.2% female; 95.6% non-Hispanic white; mean age at diagnosis, 65.3 [SD, 10.7] years) with reference controls, significant associations were observed between pancreatic cancer and mutations in CDKN2A (0.3% of cases and 0.02% of controls; odds ratio [OR], 12.33; 95% CI, 5.43-25.61); TP53 (0.2% of cases and 0.02% of controls; OR, 6.70; 95% CI, 2.52-14.95); MLH1 (0.13% of cases and 0.02% of controls; OR, 6.66; 95% CI, 1.94-17.53); BRCA2 (1.9% of cases and 0.3% of controls; OR, 6.20; 95% CI, 4.62-8.17); ATM (2.3% of cases and 0.37% of controls; OR, 5.71; 95% CI, 4.38-7.33); and BRCA1 (0.6% of cases and 0.2% of controls; OR, 2.58; 95% CI, 1.54-4.05). Conclusions and Relevance In this case-control study, mutations in 6 genes associated with pancreatic cancer were found in 5.5% of all pancreatic cancer patients, including 7.9% of patients with a family history of pancreatic cancer and 5.2% of patients without a family history of pancreatic cancer. Further research is needed for replication in other populations.