Collecting duct carcinoma of the kidney is associated with CDKN2A deletion and SLC family gene up-regulation

Jianmin Wang, Antonios Papanicolau-Sengos, Sreenivasulu Chintala, Lei Wei, Biao Liu, Qiang Hu, Kiersten Marie Miles, Jeffrey M Conroy, Sean T Glenn, Manuela Costantini, Cristina Magi-Galluzzi, Sabina Signoretti, Toni Choueiri, Michele Gallucci, Steno Sentinelli, Vito M Fazio, Maria Luana Poeta, Song Liu, Carl Morrison, Roberto Pili, Jianmin Wang, Antonios Papanicolau-Sengos, Sreenivasulu Chintala, Lei Wei, Biao Liu, Qiang Hu, Kiersten Marie Miles, Jeffrey M Conroy, Sean T Glenn, Manuela Costantini, Cristina Magi-Galluzzi, Sabina Signoretti, Toni Choueiri, Michele Gallucci, Steno Sentinelli, Vito M Fazio, Maria Luana Poeta, Song Liu, Carl Morrison, Roberto Pili

Abstract

The genetic landscape and molecular features of collecting duct carcinoma (CDC) of the kidney remain largely unknown. Herein, we performed whole exome sequencing (WES) and transcriptome sequencing (RNASeq) on 7 CDC samples (CDC1 -7). Among the 7 samples, 4 samples with matched non-tumor tissue were used for copy number analysis by SNP array data. No recurrent somatic SNVs were observed except for MLL, which was found to be mutated (p.V297I and p.F407C) in 2 samples. We identified somatic SNVs in 14 other cancer census genes including: ATM, CREBBP, PRDM1, CBFB, FBXW7, IKZF1, KDR, KRAS, NACA, NF2, NUP98, SS18, TP53, and ZNF521. SNP array data identified a CDKN2A homozygous deletion in 3 samples and SNV analysis showed a non-sense mutation of the CDKN2A gene with unknown somatic status. To estimate the recurrent rate of CDKN2A abnormalities, we performed FISH screening of additional samples and confirmed the frequent loss (62.5%) of CDKN2A expression. Since cisplatin based therapy is the common treatment option for CDC, we investigated the expression of solute carrier (SLC) family transporters and found 45% alteration. In addition, SLC7A11 (cystine transporter, xCT), a cisplatin resistance associated gene, was found to be overexpressed in 4 out of 5 (80%) cases of CDC tumors tested, as compared to matched non-tumor tissue. In summary, our study provides a comprehensive genomic analysis of CDC and identifies potential pathways suitable for targeted therapies.

Keywords: CDKN2A; collecting duct carcinoma; solute carrier family genes.

Conflict of interest statement

The authors do not have any conflicts of interest.

Figures

Figure 1. Somatic alterations in kidney CDC
Figure 1. Somatic alterations in kidney CDC
a. A representative Circos plot of CDC samples. The plot shows (from outer to inner circle) genes with somatic amino acid changes (red genes are in Cancer Census Genes), chromosomes, allele frequencies of mutations, copy number aberration (orange for gain and blue for loss), and LOH (red means LOH and grey means no LOH). b. Somatic CNV in 7 CDC samples and summary of CNVs in CDC, ccRCC, chRCC, and pRCC. Red color represents copy number gain and blue copy number loss. c. Somatic SNVs in 4 CDC samples and significantly mutated genes in ccRCC and chRCC. Green means missense mutation, red means non-sense, frameshift mutations, and orange means both. TCGA data of ccRCC and pRCC, including clinical information, somatic mutations, SNP array CNV calls, and normalized RNASeqV2, were downloaded from the TCGA data portal. Alternation status of CDKN2A was determined by somatic mutation calls and CNV segmentation results. If a segment overlapped with CDKN2A and had a logR ratio less than −0.4, CDKN2A was considered a loss in this sample. For gene expression data, the RSEM quantified and normalized data were first log2 transferred, followed by significant test. All statistical tests were performed using R statistical program followed by a significance test.
Figure 2. CDKN2A losses in CDC
Figure 2. CDKN2A losses in CDC
a. Copy number data show biallelic loss of CDKN2A in CDC1, CDC2, and CDC4 with negative log R ratios and normal like B allele frequencies. SNP array data log2 ratios were calculated by comparing the tumor sample signals with pooled non-tumor samples from Illumina. b. Representative tumor sample slides and fluorescence in situ hybridization (FISH) results of normal, single copy loss, and biallelic loss of CDKN2A loci. I, II, III: H&Es of cases CDC2, CDC9, CDC15, respectively, with infiltrating pleomorphic collecting duct carcinoma. IV, CDC2, p16 FISH with no copies each of p16 and with preserved reference probe. V, CDC9, p16 FISH with two copies each of p16 and reference probe. VI, CDC15, p16 FISH with one copy of p16 and two copies of reference probe. c. Tabulation of p16 loss according to NGS and FISH data. Cases CDC6-CDC16 were not sequenced by NGS, noted as NA.
Figure 3. RNASeq profiles of CDC gene…
Figure 3. RNASeq profiles of CDC gene expression
a. Kidney specific genes were down-regulated in all CDC tumor samples. b. Selected top up- and down-regulated genes that have been associated with cancer prognosis. c. Gene expression changes (log2 fold changes) of a five-gene signature defined by a study of genomic alterations in non-clear cell RCC (http://www.nature.com/ng/journal/v47/n1/full/ng.3146.html) to classify non-ccRCCs. CDC shows a distinct pattern for those five genes compared with three subtypes of non-ccRCC. d. Four down-steam genes (CDK4, E2F1, EZH2, and TP53) of CDKN2A were all found to be significantly up-regulated by RNASeq analysis.
Figure 4. SLC family genes upregulated in…
Figure 4. SLC family genes upregulated in CDC
a.SLC family genes overexpressed in CDC tumors showed as log2 fold change. All the genes overexpressed were significantly different in CDC tumors, compared to matched non-tumor kidney. b.SLC6A7 mRNA expression in 4 matched non-tumor kidney and 5 CDC (1 non-matched) tumors. c.SLC7A11 mRNA expression in 4 matched non-tumor kidney and 5 CDC (1 non-matched) tumors. d.SLC1A3 mRNA expression in 4 matched normal kidney and 5 CDC (1 non-matched) tumors. e. Significant overexpression (log2 fold change) of drug resistance genes SLC6A7, SLC7A11, and SLC1A3 in CDC tumors.
Figure 5. Overexpression of xCT in CDC…
Figure 5. Overexpression of xCT in CDC tumors and the association of xCT (SLC7A11) overexpression with overall poor survival in ccRCC, chRCC and pRCC patients
a.. Immunohistochemical detection of xCT was performed on CDC tumors using xCT antibody (5ug/ml, Abcam, MA). The numbers denoted in the figures are de-identified numbers of CDC tumors in the TMA. CDC1 and CDC2 tumors are included in the genomic profiling studies. Photmicrographs were captured using the Aperio Webscope Spectrum. b. Percent tumors (80%, 12 out of 15) express high levels of xCT. TCGA data analysis revealed the significant poor survival of RCC patients with xCT upregulation: c. ccRCC patients, Logrank Test P-value 0.00464; d. chRCC patients, Logrank Test P-value:0.00211, e. pRCC patients, Logrank Test P-value: 7.668e-5.
Figure 6. Significant upregulation of SLC7A11 in…
Figure 6. Significant upregulation of SLC7A11 in 3 subtypes of RCC (ccRCC, chRCC and pRCC) tumors compared to normal tissue
TCGA data analysis revealed the differential expression of SLC7A11, SLC6A7 and SLC1A3 among the 3 subtypes of RCC tumors. Left panel showing the SLC7A11 expression in ccRCC, chRCC and pRCC; middle panel with SLC6A7, right panel with SLC1A3. N = Normal tissue, T = Tumor tissue
Figure 7. Overall poor survival in RCC…
Figure 7. Overall poor survival in RCC patients with overexpression of HMGA2 and CTHRC1 which were found highly upregulated (top two genes) in CDC tumors
The available TCGA data were downloaded and utilized to determine the survival probability in ccRCC patients in order to determine the significance of overexpression of these genes in CDC tumors. a. HMGA2 expression in ccRCC and pRCC. b. Overall survival probability of ccRCC and pRCC patients with high expression of HMGA2. c. CTHRC1 expression in ccRCC and pRCC. d. Overall survival probability of ccRCC and pRCC patients with high expression of CTHRC1.
Figure 8. Alteration of CDKN2A and renal…
Figure 8. Alteration of CDKN2A and renal cancer patients survival
CDKN2A/p16INK4A alteration significantly decreased the survival in ccRCC a. and pRCC patients b.. TCGA data were used to evaluate the survival probability in renal cancer patients.
Figure 9. SLC family genes downregulated in…
Figure 9. SLC family genes downregulated in CDC tumors compared to non-tumor kidney
SLC family genes downregulated in CDC tumors showed as log2 fold changes from 8.369 to 3.580 a. and 3.569 to 0.827 b.. All the genes listed are downregulated significantly in CDC tumors compared to matched non-tumor kidney. RNASeq data was used to identify SLC family gene expression levels in CDC tumors. The difference in the expression levels as a log2 fold change in expression levels was observed.
Figure 10. CDKN2A interacting proteins
Figure 10. CDKN2A interacting proteins
To determine human CDKN2A interacting proteins, STRING10 (http://string-db.org/) database which provides known and predicted protein interactions was used. Interaction views of confidence a., evidence b. and actions c. are shown using parameters of highest confidence (0.900) and no more than 5 interactions. Protein interaction data show that CDKN2A interacts with CDK4 and TP53 (listed top 5), E2F1 and EZH2, (listed top 20, data not shown), which were found overexpressed by RNASeq analysis (Figure 3d), suggesting the functional significance of CDKN2A deletion in CDC tumors.
Figure 11. Alteration of SLC6A7 and SLC1A3…
Figure 11. Alteration of SLC6A7 and SLC1A3 association with RCC patients survival
Upper panel-TCGA data analysis of SLC6A7 alteration in 3 subtypes of RCC (ccRCC, chRCC and pRCC). Lower panel- SLC1A3 alteration and RCC patients’ survival.

References

    1. Ciszewski S, Jakimow A, Smolska-Ciszewska B. Collecting (Bellini) duct carcinoma: A clinical study of a rare tumour and review of the literature. Can Urol Assoc J. 2015;9:E589–593.
    1. Gupta R, Billis A, Shah RB, Moch H, Osunkoya AO, Jochum W, Hes O, Bacchi CE, de Castro MG, Hansel DE, Zhou M, Vankalakunti M, Salles PG, Cabrera RA, Gown AM, Amin MB. Carcinoma of the collecting ducts of Bellini and renal medullary carcinoma: clinicopathologic analysis of 52 cases of rare aggressive subtypes of renal cell carcinoma with a focus on their interrelationship. Am J Surg Pathol. 2012;36:1265–1278.
    1. Srigley JR, Delahunt B. Uncommon and recently described renal carcinomas. Mod Pathol. 2009;22(Suppl 2):S2–S23.
    1. Dason S, Allard C, Sheridan-Jonah A, Gill J, Jamshaid H, Aziz T, Kajal B, Kapoor A. Management of renal collecting duct carcinoma: a systematic review and the McMaster experience. Curr Oncol. 2013;20:e223–232.
    1. Tokuda N, Naito S, Matsuzaki O, Nagashima Y, Ozono S, Igarashi T, Japanese Society of Renal C. Collecting duct (Bellini duct) renal cell carcinoma: a nationwide survey in Japan. The Journal of urology. 2006;176:40–43. discussion 43.
    1. Wright JL, Risk MC, Hotaling J, Lin DW. Effect of collecting duct histology on renal cell cancer outcome. The Journal of urology. 2009;182:2595–2599.
    1. Kwon KA, Oh SY, Kim HY, Kim HS, Lee HY, Kim TM, Lim HY, Lee NR, Lee HJ, Hong SH, Rha SY. Clinical features and treatment of collecting duct carcinoma of the kidney from the korean cancer study group genitourinary and gynecology cancer committee. Cancer research and treatment. 2014;46:141–147.
    1. May M, Ficarra V, Shariat SF, Zigeuner R, Chromecki T, Cindolo L, Burger M, Gunia S, Feciche B, Wenzl V, Aziz A, Chun F, Becker A, Pahernik S, Simeone C, Longo N, et al. Impact of clinical and histopathological parameters on disease specific survival in patients with collecting duct renal cell carcinoma: development of a disease specific risk model. The Journal of urology. 2013;190:458–463.
    1. Procopio G, Testa I, Iacovelli R, Grassi P, Verzoni E, Garanzini E, Colecchia M, Torelli T, De Braud F. Treatment of collecting duct carcinoma: current status and future perspectives. Anticancer research. 2014;34:1027–1030.
    1. Tannir NM, Plimack E, Ng C, Tamboli P, Bekele NB, Xiao L, Smith L, Lim Z, Pagliaro L, Araujo J, Aparicio A, Matin S, Wood CG, Jonasch E. A phase 2 trial of sunitinib in patients with advanced non-clear cell renal cell carcinoma. European urology. 2012;62:1013–1019.
    1. Zhao RN, Nie LH, Gong R, Wang JZ, Wazir R, Liu LR, Song TR, Wei Q. Active targeted therapy for metastatic collecting duct carcinoma of the kidney: a case report and review of the literature. International urology and nephrology. 2013;45:1017–1021.
    1. Wu ZS, Lee JH, Kwon JA, Kim SH, Han SH, An JS, Lee JH, Lee ES, Park HR, Kim YS. Genetic alterations and chemosensitivity profile in newly established human renal collecting duct carcinoma cell lines. BJU international. 2009;103:1721–1728.
    1. Pal SK, Choueiri TK, Wang K, Khaira D, Karam JA, Van Allen E, Palma NA, Stein MN, Johnson A, Squillace R, Elvin JA, Chmielecki J, Yelensky R, Yakirevich E, Lipson D, Lin DI, et al. Characterization of Clinical Cases of Collecting Duct Carcinoma of the Kidney Assessed by Comprehensive Genomic Profiling. European urology. 2015
    1. Becker F, Junker K, Parr M, Hartmann A, Fussel S, Toma M, Grobholz R, Pflugmann T, Wullich B, Strauss A, Behnes CL, Otto W, Stockle M, Jung V. Collecting duct carcinomas represent a unique tumor entity based on genetic alterations. PLoS One. 2013;8:e78137.
    1. Edmonson MN, Zhang J, Yan C, Finney RP, Meerzaman DM, Buetow KH. Bambino: a variant detector and alignment viewer for next-generation sequencing data in the SAM/BAM format. Bioinformatics. 2011;27:865–866.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760.
    1. Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38:e164.
    1. Dalgliesh GL, Furge K, Greenman C, Chen L, Bignell G, Butler A, Davies H, Edkins S, Hardy C, Latimer C, Teague J, Andrews J, Barthorpe S, Beare D, Buck G, Campbell PJ, et al. Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature. 2010;463:360–363.
    1. Varela I, Tarpey P, Raine K, Huang D, Ong CK, Stephens P, Davies H, Jones D, Lin ML, Teague J, Bignell G, Butler A, Cho J, Dalgliesh GL, Galappaththige D, Greenman C, et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature. 2011;469:539–542.
    1. Farley MN, Schmidt LS, Mester JL, Pena-Llopis S, Pavia-Jimenez A, Christie A, Vocke CD, Ricketts CJ, Peterson J, Middelton L, Kinch L, Grishin N, Merino MJ, Metwalli AR, Xing C, Xie XJ, et al. A novel germline mutation in BAP1 predisposes to familial clear-cell renal cell carcinoma. Molecular cancer research. 2013;11:1061–1071.
    1. Sherr CJ, McCormick F. The RB and p53 pathways in cancer. Cancer cell. 2002;2:103–112.
    1. Vogelstein B, Lane D, Levine AJ. Surfing the p53 network. Nature. 2000;408:307–310.
    1. D'Angelo D, Esposito F, Fusco A. Epigenetic Mechanisms Leading to Overexpression of HMGA Proteins in Human Pituitary Adenomas. Front Med (Lausanne) 2015;2:39.
    1. Ke Z, He W, Lai Y, Guo X, Chen S, Li S, Wang Y, Wang L. Overexpression of collagen triple helix repeat containing 1 (CTHRC1) is associated with tumour aggressiveness and poor prognosis in human non-small cell lung cancer. Oncotarget. 2014;5:9410–9424. doi: 10.18632/oncotarget.2421.
    1. Tang SC, Chen YC. Novel therapeutic targets for pancreatic cancer. World J Gastroenterol. 2014;20:10825–10844.
    1. Wu J, Wei JJ. HMGA2 and high-grade serous ovarian carcinoma. J Mol Med (Berl) 2013;91:1155–1165.
    1. Fujiwara R, Takenaka S, Hashimoto M, Narawa T, Itoh T. Expression of human solute carrier family transporters in skin: possible contributor to drug-induced skin disorders. Scientific reports. 2014;4:5251.
    1. Lin L, Yee SW, Kim RB, Giacomini KM. SLC transporters as therapeutic targets: emerging opportunities. Nature reviews Drug discovery. 2015;14:543–560.
    1. Liu W, Phang JM. MiRNA and Proline Metabolism in Cancer, Oncogene and Cancer - From Bench to Clinic. Siregar Yahwardiah., editor. InTech. 2013 doi: 10.5772/55139. Available from.
    1. Saye M, Miranda MR, di Girolamo F, de los Milagros Camara M, Pereira CA. Proline modulates the Trypanosoma cruzi resistance to reactive oxygen species and drugs through a novel D, L-proline transporter. PLoS One. 2014;9:e92028.
    1. Kanai Y, Clemencon B, Simonin A, Leuenberger M, Lochner M, Weisstanner M, Hediger MA. The SLC1 high-affinity glutamate and neutral amino acid transporter family. Mol Aspects Med. 2013;34:108–120.
    1. Pedraz-Cuesta E, Christensen S, Jensen AA, Jensen NF, Bunch L, Romer MU, Brunner N, Stenvang J, Pedersen SF. The glutamate transport inhibitor DL-Threo-beta-Benzyloxyaspartic acid (DL-TBOA) differentially affects SN38- and oxaliplatin-induced death of drug-resistant colorectal cancer cells. BMC Cancer. 2015;15:411.
    1. Dai L, Noverr MC, Parsons C, Kaleeba JA, Qin Z. xCT, not just an amino-acid transporter: a multi-functional regulator of microbial infection and associated diseases. Front Microbiol. 2015;6:120.
    1. Drayton RM, Catto JW. Molecular mechanisms of cisplatin resistance in bladder cancer. Expert Rev Anticancer Ther. 2012;12:271–281.
    1. Drayton RM, Dudziec E, Peter S, Bertz S, Hartmann A, Bryant HE, Catto JW. Reduced expression of miRNA-27a modulates cisplatin resistance in bladder cancer by targeting the cystine/glutamate exchanger SLC7A11. Clinical cancer research. 2014;20:1990–2000.
    1. Lewerenz J, Hewett SJ, Huang Y, Lambros M, Gout PW, Kalivas PW, Massie A, Smolders I, Methner A, Pergande M, Smith SB, Ganapathy V, Maher P. The cystine/glutamate antiporter system x(c)(−) in health and disease: from molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal. 2013;18:522–555.
    1. Januchowski R, Zawierucha P, Andrzejewska M, Rucinski M, Zabel M. Microarray-based detection and expression analysis of ABC and SLC transporters in drug-resistant ovarian cancer cell lines. Biomedicine & pharmacotherapy. 2013;67:240–245.
    1. Chintala S, Toth K, Yin MB, Bhattacharya A, Smith SB, Ola MS, Cao S, Durrani FA, Zinia TR, Dean R, Slocum HK, Rustum YM. Downregulation of cystine transporter xc by irinotecan in human head and neck cancer FaDu xenografts. Chemotherapy. 2010;56:223–233.
    1. Guan J, Lo M, Dockery P, Mahon S, Karp CM, Buckley AR, Lam S, Gout PW, Wang YZ. The xc- cystine/glutamate antiporter as a potential therapeutic target for small-cell lung cancer: use of sulfasalazine. Cancer chemotherapy and pharmacology. 2009;64:463–472.
    1. Lu H, Samanta D, Xiang L, Zhang H, Hu H, Chen I, Bullen JW, Semenza GL. Chemotherapy triggers HIF-1-dependent glutathione synthesis and copper chelation that induces the breast cancer stem cell phenotype. Proceedings of the National Academy of Sciences of the United States of America; 2015; pp. E4600–4609.
    1. Timmerman LA, Holton T, Yuneva M, Louie RJ, Padro M, Daemen A, Hu M, Chan DA, Ethier SP, van ‘t Veer LJ, Polyak K, McCormick F, Gray JW. Glutamine sensitivity analysis identifies the xCT antiporter as a common triple-negative breast tumor therapeutic target. Cancer cell. 2013;24:450–465.
    1. Ye P, Mimura J, Okada T, Sato H, Liu T, Maruyama A, Ohyama C, Itoh K. Nrf2- and ATF4-dependent upregulation of xCT modulates the sensitivity of T24 bladder carcinoma cells to proteasome inhibition. Mol Cell Biol. 2014;34:3421–3434.
    1. Yoshikawa M, Tsuchihashi K, Ishimoto T, Yae T, Motohara T, Sugihara E, Onishi N, Masuko T, Yoshizawa K, Kawashiri S, Mukai M, Asoda S, Kawana H, Nakagawa T, Saya H, Nagano O. xCT inhibition depletes CD44v-expressing tumor cells that are resistant to EGFR-targeted therapy in head and neck squamous cell carcinoma. Cancer research. 2013;73:1855–1866.
    1. Sugiyama T, Sadzuka Y. Theanine and glutamate transporter inhibitors enhance the antitumor efficacy of chemotherapeutic agents. Biochimica et biophysica acta. 2003;1653:47–59.
    1. Fazzari J, Lin H, Murphy C, Ungard R, Singh G. Inhibitors of glutamate release from breast cancer cells; new targets for cancer-induced bone-pain. Scientific reports. 2015;5:8380.
    1. Christensen S, Jensen N, Stoeckel J, Belling K, Romer M, Gupta R, Brünner N, Pedersen S, Stenvang J. Colorectal cancer cell lines made resistant to SN38-and Oxaliplatin: Roles of altered ion transporter function in resistance? The FASEB Journal. 2013;27:lb452.
    1. Loayza-Puch F, Rooijers K, Buil LC, Zijlstra J, Oude Vrielink JF, Lopes R, Ugalde AP, van Breugel P, Hofland I, Wesseling J, van Tellingen O, Bex A, Agami R. Tumour-specific proline vulnerability uncovered by differential ribosome codon reading. Nature. 2016;530:490–494.
    1. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, Antipin Y, Reva B, Goldberg AP, Sander C, Schultz N. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–404.
    1. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, Cerami E, Sander C, Schultz N. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6:pl1.
    1. Pecuchet N, Bigot F, Gachet J, Massard C, Albiges L, Teghom C, Allory Y, Mejean A, Escudier B, Oudard S. Triple combination of bevacizumab, gemcitabine and platinum salt in metastatic collecting duct carcinoma. Annals of oncology. 2013;24:2963–2967.
    1. Bronchud MH, Castillo S, Escriva de Romani S, Mourelo S, Fernandez A, Baena C, Murillo J, Julia JC, Esquius J, Romero R, Andreu X. HER2 blockade in metastatic collecting duct carcinoma (CDC) of the kidney: a case report. Onkologie. 2012;35:776–779.
    1. Sallami S, Tanguour M, Ben Rhouma S, Cherif K, Kchir N, Horchani A. Bellini renal cell carcinoma : diagnosis and treatment. A report of 7 cases [Article in French] La Tunisie medicale. 2011;89:792–796.
    1. Tazi EM, Essadi I, Tazi MF, Ahellal Y, M'Rabti H, Errihani H. Metastatic collecting duct carcinoma of the kidney treated with sunitinib. World journal of surgical oncology. 2011;9:73.
    1. Ma MZ, Chen G, Wang P, Lu WH, Zhu CF, Song M, Yang J, Wen S, Xu RH, Hu Y, Huang P. Xc- inhibitor sulfasalazine sensitizes colorectal cancer to cisplatin by a GSH-dependent mechanism. Cancer letters. 2015;368:88–96.
    1. Yang Y, Zhang P, Zhao Y, Yang J, Jiang G, Fan J. Decreased MicroRNA-26a expression causes cisplatin resistance in human non-small cell lung cancer. Cancer biology & therapy. 2015:0.
    1. Dangi-Garimella S, Krantz SB, Barron MR, Shields MA, Heiferman MJ, Grippo PJ, Bentrem DJ, Munshi HG. Three-dimensional collagen I promotes gemcitabine resistance in pancreatic cancer through MT1-MMP-mediated expression of HMGA2. Cancer research. 2011;71:1019–1028.
    1. Marijon H, Dokmak S, Paradis V, Zappa M, Bieche I, Bouattour M, Raymond E, Faivre S. Epithelial-to-mesenchymal transition and acquired resistance to sunitinib in a patient with hepatocellular carcinoma. Journal of hepatology. 2011;54:1073–1078.
    1. Park EH, Kim S, Jo JY, Kim SJ, Hwang Y, Kim JM, Song SY, Lee DK, Koh SS. Collagen triple helix repeat containing-1 promotes pancreatic cancer progression by regulating migration and adhesion of tumor cells. Carcinogenesis. 2013;34:694–702.
    1. Tang L, Dai DL, Su M, Martinka M, Li G, Zhou Y. Aberrant expression of collagen triple helix repeat containing 1 in human solid cancers. Clinical cancer research. 2006;12:3716–3722.
    1. Yau C, Mouradov D, Jorissen RN, Colella S, Mirza G, Steers G, Harris A, Ragoussis J, Sieber O, Holmes CC. A statistical approach for detecting genomic aberrations in heterogeneous tumor samples from single nucleotide polymorphism genotyping data. Genome biology. 2010;11:R92.
    1. Anders S, Pyl PT, Huber W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31:166–169.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome biology. 2014;15:550.
    1. Xiong Q, Mukherjee S, Furey TS. GSAASeqSP: a toolset for gene set association analysis of RNA-Seq data. Scientific reports. 2014;4:6347.
    1. Ciamporcero E, Shen H, Ramakrishnan S, Yu Ku S, Chintala S, Shen L, Adelaiye R, Miles KM, Ullio C, Pizzimenti S, Daga M, Azabdaftari G, Attwood K, Johnson C, Zhang J, Barrera G, et al. YAP activation protects urothelial cell carcinoma from treatment-induced DNA damage. Oncogene. 2016;35:1541–53. doi: 10.1038/onc.2015.219.

Source: PubMed

3
S'abonner