Skip to main content
Top
Published in: Cancer Cell International 1/2022

Open Access 01-12-2022 | Research

Multidrug resistance genes screening of pancreatic ductal adenocarcinoma based on sensitivity profile to chemotherapeutic drugs

Authors: Bangbo Zhao, Cheng Qin, Zeru Li, Yuanyang Wang, Tianhao Li, Hongtao Cao, Xiaoying Yang, Tianyu Li, Weibin Wang

Published in: Cancer Cell International | Issue 1/2022

Login to get access

Abstract

Backgrounds

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancer types and chemotherapeutic drug resistance is a stumbling block in improving the overall survival of PDAC patients. The nature of specific drug resistant subpopulation within pancreatic ductal adenocarcinoma is believed to be partly attributed to epithelial-mesenchymal transition (EMT) and cell stemness. Various PDAC cell lines show various degrees of resistance to chemotherapeutic agents including gemcitabine (GEM) and 5-fluorouracil (5-FU). In-depth understanding of drug resistance mechanisms and profile heterogeneities could lead to the development of novel and precise therapeutic strategies for addressing the chemo-resistant dilemma in PDAC patients.

Methods

Cytotoxicity assays were performed by CCK8 in ten common PDAC cell lines including AsPC-1, BxPC-3, CAPAN-1, CFPAC, HPAFII, MIA PaCa-2, PANC-1, Patu-8988, SW1990 and T3M4. RNA-seq data of the ten cell lines were downloaded from Cancer Cell Line Encyclopedia (CCLE) database and subsequently analyzed for differentially expressed genes (DEGs). Based on first-line chemotherapy regimens of PDAC, DEGs between resistant and sensitive cell lines were validated by qRT-PCR. Enriched pathways of differentially expressed genes between the resistant and sensitive cell lines were acquired by Metascape database.

Results

We found that the top two toxic drugs for PDAC cell lines were paclitaxel (PTX) and GEM. Among the ten PDAC cell lines, SW1990 was the most resistant PDAC cell line with the highest IC50 levels for three drugs, while MIA PaCa-2 and BxPC-3 were the most sensitive PDAC cell lines. Differential expression analysis revealed the highest number of DEGs associated with cisplatin (CIS) sensitivity up to 642 genes, of which 181 genes were upregulated and 461 genes were downregulated in CIS-resistant cell lines. The least number of DEGs are associated with GEM sensitivity, of which 37 genes were highly expressed in GEM-resistant PDAC cell lines and 25 genes were lowly expressed. Enrichment analysis of the DEGs revealed that pathways associated with drug resistance were mainly extracellular matrix and cell–cell junction related pathways.

Conclusions

PDAC cell lines showed diverse sensitivities to commonly used chemotherapeutic agents, which was caused by differential gene expression between the resistant and sensitive cell lines. The heterogeneity and its associated genes were enriched in extracellular matrix and cell–cell junction related pathways. Our study first portrayed the sensitivity profile to chemotherapeutic drugs of PDAC, which would benefit the chemoresistance mechanism study by reemphasizing the vital role of extracellular matrix and cell–cell junction related pathways and helping the selection of suitable PDAC cell lines.
Appendix
Available only for authorised users
Literature
1.
go back to reference Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71(1):7–33.CrossRefPubMed Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71(1):7–33.CrossRefPubMed
2.
go back to reference Yang Y, Bai X, Bian D, Cai S, Chen R, Cao F, Dai M, Fang C, Fu D, Ge C, et al. Guidelines for the diagnosis and treatment of pancreatic ductal adenocarcinoma in China (2021). J Pancreatol. 2021;4(2):49–66.CrossRef Yang Y, Bai X, Bian D, Cai S, Chen R, Cao F, Dai M, Fang C, Fu D, Ge C, et al. Guidelines for the diagnosis and treatment of pancreatic ductal adenocarcinoma in China (2021). J Pancreatol. 2021;4(2):49–66.CrossRef
3.
go back to reference Christenson ES, Jaffee E, Azad NS. Current and emerging therapies for patients with advanced pancreatic ductal adenocarcinoma: a bright future. Lancet Oncol. 2020;21(3):e135–45.CrossRefPubMedPubMedCentral Christenson ES, Jaffee E, Azad NS. Current and emerging therapies for patients with advanced pancreatic ductal adenocarcinoma: a bright future. Lancet Oncol. 2020;21(3):e135–45.CrossRefPubMedPubMedCentral
4.
go back to reference Sarvepalli D, Rashid MU, Rahman AU, Ullah W, Hussain I, Hasan B, Jehanzeb S, Khan AK, Jain AG, Khetpal N, et al. Gemcitabine: a review of chemoresistance in pancreatic ductal adenocarcinoma. Crit Rev Oncog. 2019;24(2):199–212.CrossRefPubMed Sarvepalli D, Rashid MU, Rahman AU, Ullah W, Hussain I, Hasan B, Jehanzeb S, Khan AK, Jain AG, Khetpal N, et al. Gemcitabine: a review of chemoresistance in pancreatic ductal adenocarcinoma. Crit Rev Oncog. 2019;24(2):199–212.CrossRefPubMed
5.
6.
go back to reference Yang G, Guan W, Cao Z, Guo W, Xiong G, Zhao F, Feng M, Qiu J, Liu Y, Zhang MQ, et al. Integrative genomic analysis of gemcitabine resistance in pancreatic ductal adenocarcinoma by patient-derived xenograft models. Clin Cancer Res. 2021;27(12):3383–96.CrossRefPubMed Yang G, Guan W, Cao Z, Guo W, Xiong G, Zhao F, Feng M, Qiu J, Liu Y, Zhang MQ, et al. Integrative genomic analysis of gemcitabine resistance in pancreatic ductal adenocarcinoma by patient-derived xenograft models. Clin Cancer Res. 2021;27(12):3383–96.CrossRefPubMed
7.
go back to reference Grant RC, Holter S, Borgida A, Dhani NC, Hedley DW, Knox JJ, Akbari MR, Zogopoulos G, Gallinger S. Comparison of practice guidelines, BRCAPRO, and genetic counselor estimates to identify germline BRCA1 and BRCA2 mutations in pancreatic ductal adenocarcinoma. J Genet Couns. 2018;27(4):988–95.CrossRefPubMed Grant RC, Holter S, Borgida A, Dhani NC, Hedley DW, Knox JJ, Akbari MR, Zogopoulos G, Gallinger S. Comparison of practice guidelines, BRCAPRO, and genetic counselor estimates to identify germline BRCA1 and BRCA2 mutations in pancreatic ductal adenocarcinoma. J Genet Couns. 2018;27(4):988–95.CrossRefPubMed
8.
go back to reference Tutt A, Tovey H, Cheang MCU, Kernaghan S, Kilburn L, Gazinska P, Owen J, Abraham J, Barrett S, Barrett-Lee P, et al. Carboplatin in BRCA1/2-mutated and triple-negative breast cancer BRCAness subgroups: the TNT Trial. Nat Med. 2018;24(5):628–37.CrossRefPubMedPubMedCentral Tutt A, Tovey H, Cheang MCU, Kernaghan S, Kilburn L, Gazinska P, Owen J, Abraham J, Barrett S, Barrett-Lee P, et al. Carboplatin in BRCA1/2-mutated and triple-negative breast cancer BRCAness subgroups: the TNT Trial. Nat Med. 2018;24(5):628–37.CrossRefPubMedPubMedCentral
9.
go back to reference Cass I, Baldwin RL, Varkey T, Moslehi R, Narod SA, Karlan BY. Improved survival in women with BRCA-associated ovarian carcinoma. Cancer. 2003;97(9):2187–95.CrossRefPubMed Cass I, Baldwin RL, Varkey T, Moslehi R, Narod SA, Karlan BY. Improved survival in women with BRCA-associated ovarian carcinoma. Cancer. 2003;97(9):2187–95.CrossRefPubMed
10.
go back to reference Golan T, Kanji ZS, Epelbaum R, Devaud N, Dagan E, Holter S, Aderka D, Paluch-Shimon S, Kaufman B, Gershoni-Baruch R, et al. Overall survival and clinical characteristics of pancreatic ductal adenocarcinoma in BRCA mutation carriers. Br J Cancer. 2014;111(6):1132–8.CrossRefPubMedPubMedCentral Golan T, Kanji ZS, Epelbaum R, Devaud N, Dagan E, Holter S, Aderka D, Paluch-Shimon S, Kaufman B, Gershoni-Baruch R, et al. Overall survival and clinical characteristics of pancreatic ductal adenocarcinoma in BRCA mutation carriers. Br J Cancer. 2014;111(6):1132–8.CrossRefPubMedPubMedCentral
11.
go back to reference Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, Johns AL, Miller D, Nones K, Quek K, et al. Whole genomes redefine the mutational landscape of pancreatic ductal adenocarcinoma. Nature. 2015;518(7540):495–501.CrossRefPubMedPubMedCentral Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, Johns AL, Miller D, Nones K, Quek K, et al. Whole genomes redefine the mutational landscape of pancreatic ductal adenocarcinoma. Nature. 2015;518(7540):495–501.CrossRefPubMedPubMedCentral
12.
go back to reference Tempero MA. NCCN Guidelines Updates: Pancreatic ductal adenocarcinoma. J Natl Compr Canc Netw. 2019;17(5.5):603–5.PubMed Tempero MA. NCCN Guidelines Updates: Pancreatic ductal adenocarcinoma. J Natl Compr Canc Netw. 2019;17(5.5):603–5.PubMed
13.
go back to reference Conroy T, Hammel P, Hebbar M, Ben Abdelghani M, Wei AC, Raoul JL, Choné L, Francois E, Artru P, Biagi JJ, et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic ductal adenocarcinoma. N Engl J Med. 2018;379(25):2395–406.CrossRefPubMed Conroy T, Hammel P, Hebbar M, Ben Abdelghani M, Wei AC, Raoul JL, Choné L, Francois E, Artru P, Biagi JJ, et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic ductal adenocarcinoma. N Engl J Med. 2018;379(25):2395–406.CrossRefPubMed
14.
go back to reference Wang T, Lu Y, Polk A, Chowdhury P, Murga-Zamalloa C, Fujiwara H, Suemori K, Beyersdorf N, Hristov AC, Lim MS, et al. T-cell receptor signaling activates an ITK/NF-κB/GATA-3 axis in T-cell lymphomas facilitating resistance to chemotherapy. Clin Cancer Res. 2017;23(10):2506–15.CrossRefPubMed Wang T, Lu Y, Polk A, Chowdhury P, Murga-Zamalloa C, Fujiwara H, Suemori K, Beyersdorf N, Hristov AC, Lim MS, et al. T-cell receptor signaling activates an ITK/NF-κB/GATA-3 axis in T-cell lymphomas facilitating resistance to chemotherapy. Clin Cancer Res. 2017;23(10):2506–15.CrossRefPubMed
15.
go back to reference Yu J, Shi L, Lin W, Lu B, Zhao Y. UCP2 promotes proliferation and chemoresistance through regulating the NF-κB/β-catenin axis and mitochondrial ROS in gallbladder cancer. Biochem Pharmacol. 2020;172: 113745.CrossRefPubMed Yu J, Shi L, Lin W, Lu B, Zhao Y. UCP2 promotes proliferation and chemoresistance through regulating the NF-κB/β-catenin axis and mitochondrial ROS in gallbladder cancer. Biochem Pharmacol. 2020;172: 113745.CrossRefPubMed
16.
go back to reference Yu S, Zhang C, Xie KP. Therapeutic resistance of pancreatic ductal adenocarcinoma: roadmap to its reversal. Biochim Biophys Acta Rev Cancer. 2021;1875(1): 188461.CrossRefPubMed Yu S, Zhang C, Xie KP. Therapeutic resistance of pancreatic ductal adenocarcinoma: roadmap to its reversal. Biochim Biophys Acta Rev Cancer. 2021;1875(1): 188461.CrossRefPubMed
17.
go back to reference Zhou P, Li B, Liu F, Zhang M, Wang Q, Liu Y, Yao Y, Li D. The epithelial to mesenchymal transition (EMT) and cancer stem cells: implication for treatment resistance in pancreatic ductal adenocarcinoma. Mol Cancer. 2017;16(1):52.CrossRefPubMedPubMedCentral Zhou P, Li B, Liu F, Zhang M, Wang Q, Liu Y, Yao Y, Li D. The epithelial to mesenchymal transition (EMT) and cancer stem cells: implication for treatment resistance in pancreatic ductal adenocarcinoma. Mol Cancer. 2017;16(1):52.CrossRefPubMedPubMedCentral
18.
19.
go back to reference Ligorio M, Sil S, Malagon-Lopez J, Nieman LT, Misale S, Di Pilato M, Ebright RY, Karabacak MN, Kulkarni AS, Liu A, et al. Stromal microenvironment shapes the intratumoral architecture of pancreatic ductal adenocarcinoma. Cell. 2019;178(1):160-175.e127.CrossRefPubMedPubMedCentral Ligorio M, Sil S, Malagon-Lopez J, Nieman LT, Misale S, Di Pilato M, Ebright RY, Karabacak MN, Kulkarni AS, Liu A, et al. Stromal microenvironment shapes the intratumoral architecture of pancreatic ductal adenocarcinoma. Cell. 2019;178(1):160-175.e127.CrossRefPubMedPubMedCentral
20.
go back to reference Sheng W, Tang J, Cao R, Shi X, Ma Y, Dong M. Numb-PRRL promotes TGF-β1- and EGF-induced epithelial-to-mesenchymal transition in pancreatic ductal adenocarcinoma. Cell Death Dis. 2022;13(2):173.CrossRefPubMedPubMedCentral Sheng W, Tang J, Cao R, Shi X, Ma Y, Dong M. Numb-PRRL promotes TGF-β1- and EGF-induced epithelial-to-mesenchymal transition in pancreatic ductal adenocarcinoma. Cell Death Dis. 2022;13(2):173.CrossRefPubMedPubMedCentral
21.
go back to reference Drubay V, Skrypek N, Cordiez L, Vasseur R, Schulz C, Boukrout N, Duchêne B, Coppin L, Van Seuningen I, Jonckheere N. TGF-βRII Knock-down in pancreatic ductal adenocarcinoma cells promotes tumor growth and gemcitabine resistance importance of STAT3 phosphorylation on S727. Cancers (Basel). 2018;10(8):254.CrossRefPubMed Drubay V, Skrypek N, Cordiez L, Vasseur R, Schulz C, Boukrout N, Duchêne B, Coppin L, Van Seuningen I, Jonckheere N. TGF-βRII Knock-down in pancreatic ductal adenocarcinoma cells promotes tumor growth and gemcitabine resistance importance of STAT3 phosphorylation on S727. Cancers (Basel). 2018;10(8):254.CrossRefPubMed
22.
go back to reference Krebs AM, Mitschke J, Lasierra Losada M, Schmalhofer O, Boerries M, Busch H, Boettcher M, Mougiakakos D, Reichardt W, Bronsert P, et al. The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic ductal adenocarcinoma. Nat Cell Biol. 2017;19(5):518–29.CrossRefPubMed Krebs AM, Mitschke J, Lasierra Losada M, Schmalhofer O, Boerries M, Busch H, Boettcher M, Mougiakakos D, Reichardt W, Bronsert P, et al. The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic ductal adenocarcinoma. Nat Cell Biol. 2017;19(5):518–29.CrossRefPubMed
23.
go back to reference Meng Q, Shi S, Liang C, Liang D, Hua J, Zhang B, Xu J, Yu X. Abrogation of glutathione peroxidase-1 drives EMT and chemoresistance in pancreatic ductal adenocarcinoma by activating ROS-mediated Akt/GSK3β/Snail signaling. Oncogene. 2018;37(44):5843–57.CrossRefPubMed Meng Q, Shi S, Liang C, Liang D, Hua J, Zhang B, Xu J, Yu X. Abrogation of glutathione peroxidase-1 drives EMT and chemoresistance in pancreatic ductal adenocarcinoma by activating ROS-mediated Akt/GSK3β/Snail signaling. Oncogene. 2018;37(44):5843–57.CrossRefPubMed
Metadata
Title
Multidrug resistance genes screening of pancreatic ductal adenocarcinoma based on sensitivity profile to chemotherapeutic drugs
Authors
Bangbo Zhao
Cheng Qin
Zeru Li
Yuanyang Wang
Tianhao Li
Hongtao Cao
Xiaoying Yang
Tianyu Li
Weibin Wang
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2022
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-022-02785-7

Other articles of this Issue 1/2022

Cancer Cell International 1/2022 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine