Skip to main content
Erschienen in: Medical Oncology 4/2022

01.04.2022 | Original Paper

A linkage between effectual genes in progression of CRC through canonical and non-canonical TGF-β signaling pathways

verfasst von: Marzieh Shirin, Sajedeh Madadi, Noshad Peyravian, Zahra Pezeshkian, Leili Rejali, Masoumeh Hosseini, Afshin Moradi, Binazir Khanabadi, Ghazal Sherkat, Hamid Asadzadeh Aghdaei, Ehsan Nazemalhosseini-Mojarad

Erschienen in: Medical Oncology | Ausgabe 4/2022

Einloggen, um Zugang zu erhalten

Abstract

Different molecular signaling pathways have been involved in the incidence and progression of CRC. We aimed to examine the correlation between eight candidate genes, including TFGβ, SMAD2, SMAD4, RhoA, EGFR, MAP2K1, MTA1, and LEF1 in the progression of colorectal cancer (CRC) and their association with clinicopathological variables and CRC patients prognosis. Immunohistochemistry and quantitative real-time polymerase chain reaction (qRT-PCR) analysis 2−ΔΔct, were performed to assess the expression of eight genes in 64 and 122 patients with CRC, respectively and 20 normal samples were added for verification. We showed a positive correlation between SMAD2 and MAP2K1 (r = 0.337, P < 0.001), MAP2K1 and LEF1 (r = 0.187, P = 0.03), SMAD4 and RhoA (r = 0.214, P = 0.01) and as well, a negative correlation between SMAD2 and TGFβ (r =  − 0.197, P = 0.02), and RhoA and LEF1 (r =  − 0.180, P = 0.04) in tumor tissues. A decrease in RhoA mRNA expression was associated with the advanced TNM stage (P = 0.01), while the EGFR and SMAD2 mRNA expression upregulated in advanced stages (P = 0.03, P = 0.03), respectively. Also, an increase in EGFR and SMAD4 protein expression was significantly associated with the advanced TNM stage (P = 0.000) (P = .002), respectively. Perceiving the connections between canonical and non-canonical Transforming growth factor (TGF-β) signaling pathway along with the epidermal growth factor receptor (EGFR) and WNT cascades may trigger the development of novel approaches for CRC prediction.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
2.
Zurück zum Zitat Asadzadeh Aghdaei H, Pezeshkian Z, Abdollahpour-Alitappeh M, Nazemalhosseini Mojarad E, Zali MR. The role of angiogenesis in colorectal polyps and cancer, a review. Med Lab J. 2018;12(4):1–6. Asadzadeh Aghdaei H, Pezeshkian Z, Abdollahpour-Alitappeh M, Nazemalhosseini Mojarad E, Zali MR. The role of angiogenesis in colorectal polyps and cancer, a review. Med Lab J. 2018;12(4):1–6.
3.
Zurück zum Zitat Peyravian N, Larki P, Gharib E, Nazemalhosseini-Mojarad E, Anaraki F, Young C, et al. The application of gene expression profiling in predictions of occult lymph node metastasis in colorectal Cancer patients. Biomedicines. 2018;6(1):27.PubMedCentral Peyravian N, Larki P, Gharib E, Nazemalhosseini-Mojarad E, Anaraki F, Young C, et al. The application of gene expression profiling in predictions of occult lymph node metastasis in colorectal Cancer patients. Biomedicines. 2018;6(1):27.PubMedCentral
4.
Zurück zum Zitat Pranteda A, Piastra V, Stramucci L, Fratantonio D, Bossi G. The p38 MAPK signaling activation in colorectal cancer upon therapeutic treatments. Int J Mol Sci. 2020;21:2773.PubMedCentral Pranteda A, Piastra V, Stramucci L, Fratantonio D, Bossi G. The p38 MAPK signaling activation in colorectal cancer upon therapeutic treatments. Int J Mol Sci. 2020;21:2773.PubMedCentral
5.
Zurück zum Zitat Nie X, Liu H, Liu L, Wang Y-D, Chen W-D. Emerging roles of Wnt ligands in human colorectal cancer. Front Oncol. 2020;10:1341.PubMedPubMedCentral Nie X, Liu H, Liu L, Wang Y-D, Chen W-D. Emerging roles of Wnt ligands in human colorectal cancer. Front Oncol. 2020;10:1341.PubMedPubMedCentral
6.
Zurück zum Zitat Cheruku H, Mohamedali A, Cantor D, Tan SH, Nice E, Baker M. Transforming growth Factor-β, MAPK and Wnt signaling interactions in colorectal cancer. EuPA Open Proteom. 2015;8:104–15. Cheruku H, Mohamedali A, Cantor D, Tan SH, Nice E, Baker M. Transforming growth Factor-β, MAPK and Wnt signaling interactions in colorectal cancer. EuPA Open Proteom. 2015;8:104–15.
7.
Zurück zum Zitat Hua W, Dijke P, Kostidis S, Giera M, Hornsveld M. TGFβ-induced metabolic reprogramming during epithelial-to-mesenchymal transition in cancer. Cell Mol Life Sci. 2020;77:2103–23.PubMed Hua W, Dijke P, Kostidis S, Giera M, Hornsveld M. TGFβ-induced metabolic reprogramming during epithelial-to-mesenchymal transition in cancer. Cell Mol Life Sci. 2020;77:2103–23.PubMed
8.
Zurück zum Zitat Ishikawa S, Nishida N, Fujino S, Ogino T, Takahashi H, Miyoshi N, et al. Comprehensive profiling of novel epithelial–mesenchymal transition mediators and their clinical significance in colorectal cancer. Sci Rep. 2021;11(1):1–16. Ishikawa S, Nishida N, Fujino S, Ogino T, Takahashi H, Miyoshi N, et al. Comprehensive profiling of novel epithelial–mesenchymal transition mediators and their clinical significance in colorectal cancer. Sci Rep. 2021;11(1):1–16.
9.
Zurück zum Zitat Sinha A, Dijke P. E3 Ubiquitin ligases: key regulators of TGFβ signaling in cancer progression. Int J Mol Sci. 2021;22:476.PubMedCentral Sinha A, Dijke P. E3 Ubiquitin ligases: key regulators of TGFβ signaling in cancer progression. Int J Mol Sci. 2021;22:476.PubMedCentral
10.
Zurück zum Zitat Angioni R, Sánchez-Rodríguez R, Viola A, Molon B. TGF-β in Cancer: metabolic driver of the tolerogenic crosstalk in the tumor microenvironment. Cancers. 2021;13:401.PubMedPubMedCentral Angioni R, Sánchez-Rodríguez R, Viola A, Molon B. TGF-β in Cancer: metabolic driver of the tolerogenic crosstalk in the tumor microenvironment. Cancers. 2021;13:401.PubMedPubMedCentral
11.
Zurück zum Zitat Ciardiello D, Elez E, Tabernero J, Seoane J. Clinical development of therapies targeting TGFβ: current knowledge and future perspectives. Ann Oncol. 2020;31:1336–49. Ciardiello D, Elez E, Tabernero J, Seoane J. Clinical development of therapies targeting TGFβ: current knowledge and future perspectives. Ann Oncol. 2020;31:1336–49.
12.
Zurück zum Zitat Dardare J, Witz A, Merlin J-L, Gilson P, Harlé A. SMAD4 and the TGFβ pathway in patients with pancreatic ductal adenocarcinoma. Int J Mol Sci. 2020;21:3534. Dardare J, Witz A, Merlin J-L, Gilson P, Harlé A. SMAD4 and the TGFβ pathway in patients with pancreatic ductal adenocarcinoma. Int J Mol Sci. 2020;21:3534.
14.
Zurück zum Zitat Jung H, Yoon S, Lim J, Cho HJ, Lee H. Dysregulation of Rho GTPases in Human Cancers. Cancers. 2020;12:1179.PubMedCentral Jung H, Yoon S, Lim J, Cho HJ, Lee H. Dysregulation of Rho GTPases in Human Cancers. Cancers. 2020;12:1179.PubMedCentral
16.
Zurück zum Zitat Rodrigues P, Macaya I, Bazzocco S, Mazzolini R, Andretta E, Dopeso H, et al. Abstract 2058: RHOA inactivation enhances Wnt signaling and promotes colorectal cancer. Nat Commun. 2014;5:5458.PubMed Rodrigues P, Macaya I, Bazzocco S, Mazzolini R, Andretta E, Dopeso H, et al. Abstract 2058: RHOA inactivation enhances Wnt signaling and promotes colorectal cancer. Nat Commun. 2014;5:5458.PubMed
17.
Zurück zum Zitat Chen J, Ding Z-y, Li S, Liu S, Xiao C, Li Z, et al. Targeting transforming growth factor-β signaling for enhanced cancer chemotherapy. Theranostics. 2021;11:1345–63.PubMedPubMedCentral Chen J, Ding Z-y, Li S, Liu S, Xiao C, Li Z, et al. Targeting transforming growth factor-β signaling for enhanced cancer chemotherapy. Theranostics. 2021;11:1345–63.PubMedPubMedCentral
18.
Zurück zum Zitat Cammareri P, Vincent D, Hodder M, Ridgway R, Murgia C, Nobis M, et al. TGFβ pathway limits dedifferentiation following WNT and MAPK pathway activation to suppress intestinal tumourigenesis. Cell Death Differ. 2017;24:1681–93.PubMedPubMedCentral Cammareri P, Vincent D, Hodder M, Ridgway R, Murgia C, Nobis M, et al. TGFβ pathway limits dedifferentiation following WNT and MAPK pathway activation to suppress intestinal tumourigenesis. Cell Death Differ. 2017;24:1681–93.PubMedPubMedCentral
19.
Zurück zum Zitat Vu T, Datta PK. Regulation of EMT in colorectal cancer: a culprit in metastasis. Cancers. 2017;9(12):171.PubMedCentral Vu T, Datta PK. Regulation of EMT in colorectal cancer: a culprit in metastasis. Cancers. 2017;9(12):171.PubMedCentral
21.
Zurück zum Zitat Massagué J, Blain SW, Lo RS. TGFβ signaling in growth control, cancer, and heritable disorders. Cell. 2000;103(2):295–309.PubMed Massagué J, Blain SW, Lo RS. TGFβ signaling in growth control, cancer, and heritable disorders. Cell. 2000;103(2):295–309.PubMed
22.
Zurück zum Zitat Tian F, Byfield SD, Parks WT, Yoo S, Felici A, Tang B, et al. Reduction in Smad2/3 signaling enhances tumorigenesis but suppresses metastasis of breast cancer cell lines. Can Res. 2003;63(23):8284–92. Tian F, Byfield SD, Parks WT, Yoo S, Felici A, Tang B, et al. Reduction in Smad2/3 signaling enhances tumorigenesis but suppresses metastasis of breast cancer cell lines. Can Res. 2003;63(23):8284–92.
23.
Zurück zum Zitat Xie W, Rimm D, Lin Y, Shih W, Reiss M. Loss of Smad Signaling in Human Colorectal Cancer Is Associated with Advanced Disease and Poor Prognosis. Cancer journal (Sudbury, Mass). 2003;9:302–12. Xie W, Rimm D, Lin Y, Shih W, Reiss M. Loss of Smad Signaling in Human Colorectal Cancer Is Associated with Advanced Disease and Poor Prognosis. Cancer journal (Sudbury, Mass). 2003;9:302–12.
24.
Zurück zum Zitat Zhao G, Wojciechowski MC, Jee S, Boros J, McAvoy JW, Lovicu FJ. Negative regulation of TGFβ-induced lens epithelial to mesenchymal transition (EMT) by RTK antagonists. Exp Eye Res. 2015;132:9–16.PubMed Zhao G, Wojciechowski MC, Jee S, Boros J, McAvoy JW, Lovicu FJ. Negative regulation of TGFβ-induced lens epithelial to mesenchymal transition (EMT) by RTK antagonists. Exp Eye Res. 2015;132:9–16.PubMed
26.
Zurück zum Zitat Papageorgis P, Cheng K-H, Ozturk S, Gong Y, Lambert A, Mostafavi Abdolmaleky H, et al. Smad4 inactivation promotes malignancy and drug resistance of colon cancer. Can Res. 2011;71:998–1008. Papageorgis P, Cheng K-H, Ozturk S, Gong Y, Lambert A, Mostafavi Abdolmaleky H, et al. Smad4 inactivation promotes malignancy and drug resistance of colon cancer. Can Res. 2011;71:998–1008.
27.
Zurück zum Zitat Samanta D, Datta P. Alterations in the Smad pathway in human cancers. Front Biosci. 2012;17:1281–93. Samanta D, Datta P. Alterations in the Smad pathway in human cancers. Front Biosci. 2012;17:1281–93.
28.
Zurück zum Zitat Maitra A, Molberg K, Albores-Saavedra J, Lindberg G. Loss of Dpc4 expression in colonic adenocarcinomas correlates with the presence of metastatic disease. Am J Pathol. 2000;157:1105–11.PubMedPubMedCentral Maitra A, Molberg K, Albores-Saavedra J, Lindberg G. Loss of Dpc4 expression in colonic adenocarcinomas correlates with the presence of metastatic disease. Am J Pathol. 2000;157:1105–11.PubMedPubMedCentral
29.
Zurück zum Zitat Inamoto S, Itatani Y, Yamamoto T, Minamiguchi S, Hirai H, Iwamoto M, et al. Loss of SMAD4 promotes colorectal cancer progression by accumulation of myeloid-derived suppressor cells through CCL15-CCR1 Chemokine Axis. Clin Cancer Res. 2015;22:492–501.PubMed Inamoto S, Itatani Y, Yamamoto T, Minamiguchi S, Hirai H, Iwamoto M, et al. Loss of SMAD4 promotes colorectal cancer progression by accumulation of myeloid-derived suppressor cells through CCL15-CCR1 Chemokine Axis. Clin Cancer Res. 2015;22:492–501.PubMed
30.
Zurück zum Zitat Peyravian N, Nobili S, Pezeshkian Z, Olfatifar M, Moradi A, Baghaei K, et al. Increased Expression of VANGL1 is Predictive of Lymph Node metastasis in colorectal cancer: results from a 20-Gene expression signature. J Personalized Med. 2021;11(2):126. Peyravian N, Nobili S, Pezeshkian Z, Olfatifar M, Moradi A, Baghaei K, et al. Increased Expression of VANGL1 is Predictive of Lymph Node metastasis in colorectal cancer: results from a 20-Gene expression signature. J Personalized Med. 2021;11(2):126.
31.
Zurück zum Zitat Seshimo I, Yamamoto H, Mishima H, Kurata A, Suzuki R, Ezumi K, et al. Expression and mutation of SMAD4 in poorly differentiated carcinoma and signet-ring cell carcinoma of the colorectum. J Exp Clin Cancer Res. 2006;25:433–42.PubMed Seshimo I, Yamamoto H, Mishima H, Kurata A, Suzuki R, Ezumi K, et al. Expression and mutation of SMAD4 in poorly differentiated carcinoma and signet-ring cell carcinoma of the colorectum. J Exp Clin Cancer Res. 2006;25:433–42.PubMed
32.
Zurück zum Zitat Tsubakihara Y, Moustakas A. Epithelial-Mesenchymal Transition and Metastasis under the Control of Transforming Growth Factor β. Int J Mol Sci. 2018;19:3672.PubMedCentral Tsubakihara Y, Moustakas A. Epithelial-Mesenchymal Transition and Metastasis under the Control of Transforming Growth Factor β. Int J Mol Sci. 2018;19:3672.PubMedCentral
33.
Zurück zum Zitat Hao Y, Baker D, Dijke P. TGF-β-Mediated Epithelial-Mesenchymal Transition and Cancer Metastasis. Int J Mol Sci. 2019;20:2767.PubMedCentral Hao Y, Baker D, Dijke P. TGF-β-Mediated Epithelial-Mesenchymal Transition and Cancer Metastasis. Int J Mol Sci. 2019;20:2767.PubMedCentral
34.
Zurück zum Zitat Itatani Y, Kawada K, Sakai Y. Transforming growth Factor-β signaling pathway in colorectal cancer and its tumor microenvironment. Int J Mol Sci. 2019;20:5822.PubMedCentral Itatani Y, Kawada K, Sakai Y. Transforming growth Factor-β signaling pathway in colorectal cancer and its tumor microenvironment. Int J Mol Sci. 2019;20:5822.PubMedCentral
35.
Zurück zum Zitat Pradhan R, Ngo P, Martínez-Sánchez L, Neurath M, López PR. Rho GTPases as key molecular players within intestinal mucosa and GI Diseases. Cells. 2021;10:66.PubMedCentral Pradhan R, Ngo P, Martínez-Sánchez L, Neurath M, López PR. Rho GTPases as key molecular players within intestinal mucosa and GI Diseases. Cells. 2021;10:66.PubMedCentral
36.
Zurück zum Zitat Dopeso H, Rodrigues P, Bilic J, Bazzocco S, Carton-Garcia F, Macaya I, et al. Mechanisms of inactivation of the tumour suppressor gene RHOA in colorectal cancer. British J Cancer. 2017;118:106–16. Dopeso H, Rodrigues P, Bilic J, Bazzocco S, Carton-Garcia F, Macaya I, et al. Mechanisms of inactivation of the tumour suppressor gene RHOA in colorectal cancer. British J Cancer. 2017;118:106–16.
37.
Zurück zum Zitat Nowrin K, Sohal S, Peterson G, Patel R, Walters E. Epithelial-mesenchymal transition as a fundamental underlying pathogenic process in COPD airways: Fibrosis, remodeling and cancer. Expert Rev Respir Med. 2014;8:1–13. Nowrin K, Sohal S, Peterson G, Patel R, Walters E. Epithelial-mesenchymal transition as a fundamental underlying pathogenic process in COPD airways: Fibrosis, remodeling and cancer. Expert Rev Respir Med. 2014;8:1–13.
38.
Zurück zum Zitat Rice A, Cortes E, Lachowski D, Oertle P, Matellan C, Thorpe S, et al. GPER activation inhibits cancer cell mechanotransduction and basement membrane invasion via RhoA. Cancers. 2020;12:289.PubMedCentral Rice A, Cortes E, Lachowski D, Oertle P, Matellan C, Thorpe S, et al. GPER activation inhibits cancer cell mechanotransduction and basement membrane invasion via RhoA. Cancers. 2020;12:289.PubMedCentral
39.
Zurück zum Zitat Alazzouzi H, Alhopuro P, Salovaara R, Sammalkorpi H, Järvinen H, Mecklin J-P, et al. SMAD4 as a prognostic marker in colorectal cancer. Clin Cancer Res. 2005;11(7):2606–11.PubMed Alazzouzi H, Alhopuro P, Salovaara R, Sammalkorpi H, Järvinen H, Mecklin J-P, et al. SMAD4 as a prognostic marker in colorectal cancer. Clin Cancer Res. 2005;11(7):2606–11.PubMed
40.
Zurück zum Zitat Kriegl L, Horst D, Reiche J, Engel J, Kirchner T, Jung A. LEF-1 and TCF4 expression correlate inversely with survival in colorectal cancer. J Transl Med. 2010;8:123.PubMedPubMedCentral Kriegl L, Horst D, Reiche J, Engel J, Kirchner T, Jung A. LEF-1 and TCF4 expression correlate inversely with survival in colorectal cancer. J Transl Med. 2010;8:123.PubMedPubMedCentral
41.
Zurück zum Zitat Repetto L, Gianni W, Agliano A, Gazzaniga P. Impact of EGFR expression on colorectal cancer patient prognosis and survival: a response. Ann Oncol. 2005;16(9):1557.PubMed Repetto L, Gianni W, Agliano A, Gazzaniga P. Impact of EGFR expression on colorectal cancer patient prognosis and survival: a response. Ann Oncol. 2005;16(9):1557.PubMed
42.
Zurück zum Zitat Huang K, Gao N, Bian D, Zhai Q, Yang P, Li M, et al. Correlation between FAK and EGF-induced EMT in colorectal cancer cells. J Oncol. 2020;2020:1–10. Huang K, Gao N, Bian D, Zhai Q, Yang P, Li M, et al. Correlation between FAK and EGF-induced EMT in colorectal cancer cells. J Oncol. 2020;2020:1–10.
43.
Zurück zum Zitat Li D-Q, Pakala S, Nair S, Eswaran J, Kumar R. Metastasis-Associated Protein 1/Nucleosome Remodeling and Histone Deacetylase Complex in Cancer. Can Res. 2012;72:387–94. Li D-Q, Pakala S, Nair S, Eswaran J, Kumar R. Metastasis-Associated Protein 1/Nucleosome Remodeling and Histone Deacetylase Complex in Cancer. Can Res. 2012;72:387–94.
44.
Zurück zum Zitat Toh Y, Nicolson G. The role of the MTA family and their encoded proteins in human cancers: Molecular functions and clinical implications. Clin Exp Metas. 2009;26:215–27. Toh Y, Nicolson G. The role of the MTA family and their encoded proteins in human cancers: Molecular functions and clinical implications. Clin Exp Metas. 2009;26:215–27.
45.
Zurück zum Zitat Pakala SB, Singh K, Reddy SDN, Ohshiro K, Li D-Q, Mishra L, et al. TGF-β1 signaling targets metastasis-associated protein 1, a new effector in epithelial cells. Oncogene. 2011;30(19):2230–41.PubMedPubMedCentral Pakala SB, Singh K, Reddy SDN, Ohshiro K, Li D-Q, Mishra L, et al. TGF-β1 signaling targets metastasis-associated protein 1, a new effector in epithelial cells. Oncogene. 2011;30(19):2230–41.PubMedPubMedCentral
46.
Zurück zum Zitat Zhu W, Cai M-Y, Tong Z-T, Dong S-S, Mai S-J, Liao Y-J, et al. Overexpression of EIF5A2 promotes colorectal carcinoma cell aggressiveness by upregulating MTA1 through C-myc to induce epithelial–mesenchymaltransition. Gut. 2012;61(4):562–75.PubMed Zhu W, Cai M-Y, Tong Z-T, Dong S-S, Mai S-J, Liao Y-J, et al. Overexpression of EIF5A2 promotes colorectal carcinoma cell aggressiveness by upregulating MTA1 through C-myc to induce epithelial–mesenchymaltransition. Gut. 2012;61(4):562–75.PubMed
47.
Zurück zum Zitat Cagatay S, Cimen I, Savas B, Banerjee S. MTA-1 expression is associated with metastasis and epithelial to mesenchymal transition in colorectal cancer cells. Tumour Biol. 2013;34:1189–204. Cagatay S, Cimen I, Savas B, Banerjee S. MTA-1 expression is associated with metastasis and epithelial to mesenchymal transition in colorectal cancer cells. Tumour Biol. 2013;34:1189–204.
48.
Zurück zum Zitat Li J, Ye L, Sun P-H, Satherley L, Hargest R, Zhang Z, et al. MTA1 Is Up-regulated in colorectal cancer and is inversely correlated with lymphatic metastasis. Cancer Genomics Proteomics. 2015;12:339–45.PubMed Li J, Ye L, Sun P-H, Satherley L, Hargest R, Zhang Z, et al. MTA1 Is Up-regulated in colorectal cancer and is inversely correlated with lymphatic metastasis. Cancer Genomics Proteomics. 2015;12:339–45.PubMed
Metadaten
Titel
A linkage between effectual genes in progression of CRC through canonical and non-canonical TGF-β signaling pathways
verfasst von
Marzieh Shirin
Sajedeh Madadi
Noshad Peyravian
Zahra Pezeshkian
Leili Rejali
Masoumeh Hosseini
Afshin Moradi
Binazir Khanabadi
Ghazal Sherkat
Hamid Asadzadeh Aghdaei
Ehsan Nazemalhosseini-Mojarad
Publikationsdatum
01.04.2022
Verlag
Springer US
Erschienen in
Medical Oncology / Ausgabe 4/2022
Print ISSN: 1357-0560
Elektronische ISSN: 1559-131X
DOI
https://doi.org/10.1007/s12032-021-01634-3

Weitere Artikel der Ausgabe 4/2022

Medical Oncology 4/2022 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.