1
|
Kashkin K, Kondratyeva L, Kopantzev E, Abramov I, Zhukova L, Chernov I. Deciphering of SOX9 Functions in Pancreatic Cancer Cells. Int J Mol Sci 2025; 26:2652. [PMID: 40141294 PMCID: PMC11941869 DOI: 10.3390/ijms26062652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
SOX9 is widely regarded as a key master regulator of gene transcription, responsible for the development and differentiation programs within tissue and organogenesis, particularly in the pancreas. SOX9 overexpression has been observed in multiple tumor types, including pancreatic cancer, and is discussed as a prognostic marker. In order to gain a more profound understanding of the role of SOX9 in pancreatic cancer, we have performed SOX9 knockdown in the COLO357 and PANC-1 cells using RNA interference, followed by full-transcriptome analysis of the siRNA-transfected cells. The molecular pathway enrichment analysis between SOX9-specific siRNA-transfected cells and control cells reveals the activation of processes associated with cellular signaling, cell differentiation, transcription, and methylation, alongside the suppression of genes involved in various stages of the cell cycle and apoptosis, upon the SOX9 knockdown. Alterations of the expression of transcription factors, epithelial-mesenchymal transition markers, oncogenes, tumor suppressor genes, and drug resistance-related genes upon SOX9 knockdown in comparison of primary and metastatic pancreatic cancer cells are discovered. The expression levels of genes comprising prognostic signatures for pancreatic cancer were also evaluated following SOX9 knockdown. Additional studies are needed to assess the properties and prognostic significance of SOX9 in pancreatic cancer using other biological models.
Collapse
Affiliation(s)
- Kirill Kashkin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, 117997 Moscow, Russia; (E.K.); (I.C.)
| | - Liya Kondratyeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, 117997 Moscow, Russia; (E.K.); (I.C.)
| | - Eugene Kopantzev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, 117997 Moscow, Russia; (E.K.); (I.C.)
| | - Ivan Abramov
- GBUZ Moscow Clinical Scientific and Practical Center Named After A.S. Loginov MHD (MCSC), 111123 Moscow, Russia; (I.A.); (L.Z.)
| | - Lyudmila Zhukova
- GBUZ Moscow Clinical Scientific and Practical Center Named After A.S. Loginov MHD (MCSC), 111123 Moscow, Russia; (I.A.); (L.Z.)
| | - Igor Chernov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, 117997 Moscow, Russia; (E.K.); (I.C.)
| |
Collapse
|
2
|
Phan T, Fan D, Melstrom LG. Developing Vaccines in Pancreatic Adenocarcinoma: Trials and Tribulations. Curr Oncol 2024; 31:4855-4884. [PMID: 39329989 PMCID: PMC11430674 DOI: 10.3390/curroncol31090361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024] Open
Abstract
Pancreatic adenocarcinoma represents one of the most challenging malignancies to treat, with dismal survival rates despite advances in therapeutic modalities. Immunotherapy, particularly vaccines, has emerged as a promising strategy to harness the body's immune system in combating this aggressive cancer. This abstract reviews the trials and tribulations encountered in the development of vaccines targeting pancreatic adenocarcinoma. Key challenges include the immunosuppressive tumor microenvironment, the heterogeneity of tumor antigens, and a limited understanding of immune evasion mechanisms employed by pancreatic cancer cells. Various vaccine platforms, including peptide-based, dendritic cell-based, and viral vector-based vaccines, have been explored in preclinical and clinical settings. However, translating promising results from preclinical models to clinical efficacy has proven elusive. In recent years, mRNA vaccines have emerged as a promising immunotherapeutic strategy in the fight against various cancers, including pancreatic adenocarcinoma. We will discuss the potential applications, opportunities, and challenges associated with mRNA vaccines in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Thuy Phan
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Darrell Fan
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Laleh G. Melstrom
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA;
| |
Collapse
|
3
|
Urolithins increased anticancer effects of chemical drugs, ionizing radiation and hyperthermia on human esophageal carcinoma cells in vitro. Tissue Cell 2022; 77:101846. [DOI: 10.1016/j.tice.2022.101846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/21/2022] [Accepted: 05/26/2022] [Indexed: 12/24/2022]
|
4
|
Neo SP, Alli-Shaik A, Wee S, Lim Z, Gunaratne J. Englerin A Rewires Phosphosignaling via Hsp27 Hyperphosphorylation to Induce Cytotoxicity in Renal Cancer Cells. J Proteome Res 2022; 21:1948-1960. [PMID: 35838755 DOI: 10.1021/acs.jproteome.2c00248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Englerin A (EA) is a small-molecule natural product with selective cytotoxicity against renal cancer cells. EA has been shown to induce apoptosis and cell death through cell-cycle arrest and/or insulin signaling pathways. However, its biological mode of action or targets in renal cancer remains enigmatic. In this study, we employed advanced mass spectrometry-based phosphoproteomics approaches to identify EA's functional roles in renal cancer. We identified 10,940 phosphorylation sites, of which 706 sites exhibited EA-dependent phosphorylation changes. Integrated analysis of motifs and interaction networks suggested activation of stress-activated kinases including p38 upon EA treatment. Of note, a downstream target of p38, Hsp27, was found to be hyperphosphorylated on multiple sites upon EA treatment. Among these, a novel site Ser65 on Hsp27, which was further validated by targeted proteomics, was shown to be crucial for EA-induced cytotoxicity in renal cancer cells. Taken together, these data reveal the complex signaling cascade that is induced upon EA treatment and importantly provide insights into its effects on downstream molecular signaling.
Collapse
Affiliation(s)
- Suat Peng Neo
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Asfa Alli-Shaik
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Sheena Wee
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Zijie Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Jayantha Gunaratne
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
| |
Collapse
|
5
|
Sottile ML, Cuello-Carrión FD, Gómez LC, Semino S, Ibarra J, García MB, Gonzalez L, Vargas-Roig LM, Nadin SB. DNA Damage Repair Proteins, HSP27, and Phosphorylated-HSP90α as Predictive/Prognostic Biomarkers of Platinum-based Cancer Chemotherapy: An Exploratory Study. Appl Immunohistochem Mol Morphol 2022; 30:425-434. [PMID: 35639358 DOI: 10.1097/pai.0000000000001037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 04/25/2022] [Indexed: 11/27/2022]
Abstract
Platinum analogs are commonly used for cancer treatment. There is increasing interest in finding biomarkers which could predict and overcome resistance, because to date there is no reliable predictive/prognostic marker for these compounds. Here we studied the immunohistochemical expression of proteins involved in DNA damage response and repair (γH2AX, 53BP1, ERCC1, MLH1, and MSH2) in primary tumor tissues from patients treated with platinum-based chemotherapy. Levels and localization of Heat Shock Protein (HSP)27 and phospho-(Thr5/7)-HSP90α (p-HSP90α) were also determined. The implications in clinical response, disease-free survival and overall survival were analyzed. High γH2AX and 53BP1 expressions were associated with poor clinical response. Nuclear p-HSP90α, as well as nuclear absence and low cytoplasmic expression of HSP27 correlated with good response. Patients with high γH2AX and high cytoplasmic HSP27 expressions had shorter overall survival and disease-free survival. MLH1, MSH2, or ERCC1 were not associated with clinical response or survival. We report the potential utility of p-HSP90α, HSP27, γH2AX, and 53BP1 as predictive/prognostic markers for platinum-based chemotherapy. We present the first study that evaluates the predictive and prognostic value of p-HSP90α in primary tumors. Our research opens new possibilities for clinical oncology and shows the usefulness of immunohistochemistry for predicting chemotherapy response and prognosis in cancer.
Collapse
Affiliation(s)
- Mayra L Sottile
- Tumor Biology Laboratory
- Medical Sciences School, Mendoza University
| | | | - Laura C Gómez
- Tumor Biology Laboratory
- Medical Sciences School, Mendoza University
| | | | - Jorge Ibarra
- Regional Integration Cancer Center, Mendoza, Argentina
| | | | | | | | | |
Collapse
|
6
|
Trilla-Fuertes L, Gámez-Pozo A, Lumbreras-Herrera MI, López-Vacas R, Heredia-Soto V, Ghanem I, López-Camacho E, Zapater-Moros A, Miguel M, Peña-Burgos EM, Palacios E, de Uribe M, Guerra L, Dittmann A, Mendiola M, Fresno Vara JÁ, Feliu J. Identification of Carcinogenesis and Tumor Progression Processes in Pancreatic Ductal Adenocarcinoma Using High-Throughput Proteomics. Cancers (Basel) 2022; 14:cancers14102414. [PMID: 35626021 PMCID: PMC9139847 DOI: 10.3390/cancers14102414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/05/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with an overall 5-year survival rate of just 5%. A better understanding of the carcinogenesis processes and the mechanisms of the progression of PDAC is mandatory. Fifty-two PDAC patients treated with surgery and adjuvant therapy, with available primary tumors, normal tissue, preneoplastic lesions (PanIN), and/or lymph node metastases, were selected for the study. Proteins were extracted from small punches and analyzed by LC-MS/MS using data-independent acquisition. Proteomics data were analyzed using probabilistic graphical models, allowing functional characterization. Comparisons between groups were made using linear mixed models. Three proteomic tumor subtypes were defined. T1 (32% of patients) was related to adhesion, T2 (34%) had metabolic features, and T3 (34%) presented high splicing and nucleoplasm activity. These proteomics subtypes were validated in the PDAC TCGA cohort. Relevant biological processes related to carcinogenesis and tumor progression were studied in each subtype. Carcinogenesis in the T1 subtype seems to be related to an increase of adhesion and complement activation node activity, whereas tumor progression seems to be related to nucleoplasm and translation nodes. Regarding the T2 subtype, it seems that metabolism and, especially, mitochondria act as the motor of cancer development. T3 analyses point out that nucleoplasm, mitochondria and metabolism, and extracellular matrix nodes could be involved in T3 tumor carcinogenesis. The identified processes were different among proteomics subtypes, suggesting that the molecular motor of the disease is different in each subtype. These differences can have implications for the development of future tailored therapeutic approaches for each PDAC proteomics subtype.
Collapse
Affiliation(s)
- Lucía Trilla-Fuertes
- Molecular Oncology & Pathology Laboratory, Instituto de Genética Médica y Molecular-INGEMM, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain; (L.T.-F.); (A.G.-P.); (M.I.L.-H.); (R.L.-V.); (J.Á.F.V.)
| | - Angelo Gámez-Pozo
- Molecular Oncology & Pathology Laboratory, Instituto de Genética Médica y Molecular-INGEMM, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain; (L.T.-F.); (A.G.-P.); (M.I.L.-H.); (R.L.-V.); (J.Á.F.V.)
| | - María Isabel Lumbreras-Herrera
- Molecular Oncology & Pathology Laboratory, Instituto de Genética Médica y Molecular-INGEMM, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain; (L.T.-F.); (A.G.-P.); (M.I.L.-H.); (R.L.-V.); (J.Á.F.V.)
| | - Rocío López-Vacas
- Molecular Oncology & Pathology Laboratory, Instituto de Genética Médica y Molecular-INGEMM, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain; (L.T.-F.); (A.G.-P.); (M.I.L.-H.); (R.L.-V.); (J.Á.F.V.)
| | - Victoria Heredia-Soto
- Molecular Pathology and Therapeutic Targets Group, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain; (V.H.-S.); (M.M.); (M.M.)
- Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, 28029 Madrid, Spain
| | - Ismael Ghanem
- Medical Oncology Service, Hospital Universitario La Paz, 28046 Madrid, Spain;
| | | | | | - María Miguel
- Molecular Pathology and Therapeutic Targets Group, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain; (V.H.-S.); (M.M.); (M.M.)
| | - Eva M. Peña-Burgos
- Pathology Department, Hospital Universitario La Paz, 28046 Madrid, Spain; (E.M.P.-B.); (E.P.); (M.d.U.); (L.G.)
| | - Elena Palacios
- Pathology Department, Hospital Universitario La Paz, 28046 Madrid, Spain; (E.M.P.-B.); (E.P.); (M.d.U.); (L.G.)
| | - Marta de Uribe
- Pathology Department, Hospital Universitario La Paz, 28046 Madrid, Spain; (E.M.P.-B.); (E.P.); (M.d.U.); (L.G.)
| | - Laura Guerra
- Pathology Department, Hospital Universitario La Paz, 28046 Madrid, Spain; (E.M.P.-B.); (E.P.); (M.d.U.); (L.G.)
| | - Antje Dittmann
- Functional Genomics Center Zurich, University of Zurich/ETH Zurich, 8057 Zurich, Switzerland;
| | - Marta Mendiola
- Molecular Pathology and Therapeutic Targets Group, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain; (V.H.-S.); (M.M.); (M.M.)
| | - Juan Ángel Fresno Vara
- Molecular Oncology & Pathology Laboratory, Instituto de Genética Médica y Molecular-INGEMM, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain; (L.T.-F.); (A.G.-P.); (M.I.L.-H.); (R.L.-V.); (J.Á.F.V.)
- Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, 28029 Madrid, Spain
| | - Jaime Feliu
- Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, 28029 Madrid, Spain
- Medical Oncology Service, Hospital Universitario La Paz, 28046 Madrid, Spain;
- Cátedra UAM-ANGEM, Faculty of Medicine, Universidad Autónoma de Madrid, 28046 Madrid, Spain
- Correspondence:
| |
Collapse
|
7
|
Curcumin and Ethanol Effects in Trembler-J Schwann Cell Culture. Biomolecules 2022; 12:biom12040515. [PMID: 35454103 PMCID: PMC9025918 DOI: 10.3390/biom12040515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 02/04/2023] Open
Abstract
Charcot-Marie-Tooth (CMT) syndrome is the most common progressive human motor and sensory peripheral neuropathy. CMT type 1E is a demyelinating neuropathy affecting Schwann cells due to peripheral-myelin-protein-22 (PMP22) mutations, modelized by Trembler-J mice. Curcumin, a natural polyphenol compound obtained from turmeric (Curcuma longa), exhibits dose- and time-varying antitumor, antioxidant and neuroprotective properties, however, the neurotherapeutic actions of curcumin remain elusive. Here, we propose curcumin as a possible natural treatment capable of enhancing cellular detoxification mechanisms, resulting in an improvement of the neurodegenerative Trembler-J phenotype. Using a refined method for obtaining enriched Schwann cell cultures, we evaluated the neurotherapeutic action of low dose curcumin treatment on the PMP22 expression, and on the chaperones and autophagy/mammalian target of rapamycin (mTOR) pathways in Trembler-J and wild-type genotypes. In wild-type Schwann cells, the action of curcumin resulted in strong stimulation of the chaperone and macroautophagy pathway, whereas the modulation of ribophagy showed a mild effect. However, despite the promising neuroprotective effects for the treatment of neurological diseases, we demonstrate that the action of curcumin in Trembler-J Schwann cells could be impaired due to the irreversible impact of ethanol used as a common curcumin vehicle necessary for administration. These results contribute to expanding our still limited understanding of PMP22 biology in neurobiology and expose the intrinsic lability of the neurodegenerative Trembler-J genotype. Furthermore, they unravel interesting physiological mechanisms of cellular resilience relevant to the pharmacological treatment of the neurodegenerative Tremble J phenotype with curcumin and ethanol. We conclude that the analysis of the effects of the vehicle itself is an essential and inescapable step to comprehensibly assess the effects and full potential of curcumin treatment for therapeutic purposes.
Collapse
|
8
|
Kalli M, Li R, Mills GB, Stylianopoulos T, Zervantonakis IK. Mechanical Stress Signaling in Pancreatic Cancer Cells Triggers p38 MAPK- and JNK-Dependent Cytoskeleton Remodeling and Promotes Cell Migration via Rac1/cdc42/Myosin II. Mol Cancer Res 2022; 20:485-497. [PMID: 34782370 PMCID: PMC8898300 DOI: 10.1158/1541-7786.mcr-21-0266] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 09/24/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022]
Abstract
Advanced or metastatic pancreatic cancer is highly resistant to existing therapies, and new treatments are urgently needed to improve patient outcomes. Current studies focus on alternative treatment approaches that target the abnormal microenvironment of pancreatic tumors and the resulting elevated mechanical stress in the tumor interior. Nevertheless, the underlying mechanisms by which mechanical stress regulates pancreatic cancer metastatic potential remain elusive. Herein, we used a proteomic assay to profile mechanical stress-induced signaling cascades that drive the motility of pancreatic cancer cells. Proteomic analysis, together with selective protein inhibition and siRNA treatments, revealed that mechanical stress enhances cell migration through activation of the p38 MAPK/HSP27 and JNK/c-Jun signaling axes, and activation of the actin cytoskeleton remodelers: Rac1, cdc42, and myosin II. In addition, mechanical stress upregulated transcription factors associated with epithelial-to-mesenchymal transition and stimulated the formation of stress fibers and filopodia. p38 MAPK and JNK inhibition resulted in lower cell proliferation and more effectively blocked cell migration under mechanical stress compared with control conditions. The enhanced tumor cell motility under mechanical stress was potently reduced by cdc42 and Rac1 silencing with no effects on proliferation. Our results highlight the importance of targeting aberrant signaling in cancer cells that have adapted to mechanical stress in the tumor microenvironment, as a novel approach to effectively limit pancreatic cancer cell migration. IMPLICATIONS Our findings highlight that mechanical stress activated the p38 MAPK and JNK signaling axis and stimulated pancreatic cancer cell migration via upregulation of the actin cytoskeleton remodelers cdc42 and Rac1.
Collapse
Affiliation(s)
- Maria Kalli
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Ruxuan Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Gordon B. Mills
- Knight Cancer Institute, Oregon Health Sciences University, Oregon, Pennsylvania
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Ioannis K. Zervantonakis
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| |
Collapse
|
9
|
Forika G, Kiss E, Petovari G, Danko T, Gellert AB, Krenacs T. Modulated Electro-Hyperthermia Supports the Effect of Gemcitabine Both in Sensitive and Resistant Pancreas Adenocarcinoma Cell Lines. Pathol Oncol Res 2021; 27:1610048. [PMID: 34955688 PMCID: PMC8702438 DOI: 10.3389/pore.2021.1610048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/17/2021] [Indexed: 12/09/2022]
Abstract
The poor prognosis of pancreatic ductal adenocarcinoma (PDAC) is frequently associated to high treatment resistance. Gemcitabine (GEM) alone or in combination is the most used chemotherapy for unresecable PDACs. Here we studied whether modulated electro-hyperthermia (mEHT), a non-invasive complementary treatment, can support the effect of GEM on PDAC cells in vitro. The LD20 for the GEM-resistant Panc1 cells proved to be 200× higher than for the drug-sensitive Capan1. The mEHT alone caused significant apoptosis in Capan1 cultures as confirmed by the elevated SubG1 phase cell fraction and increased number of cleaved Caspase-3 positive cells 48 h after treatment, with an additive effect when GEM was used after hyperthermia. These were accompanied by reduced number of G1, S, and G2/M phase cells and elevated expression of the cyclin-dependent kinase inhibitor p21waf1 protein. In GEM-resistant Panc1 cells, an initial apoptosis was detected by flow cytometry 24 h after mEHT ± GEM treatment, which however diminished by 48 h at persistent number of cleaved Caspase-3 positive tumor cells. Though GEM monotherapy reduced the number of tumor progenitor colonies in Capan1 cell line, an additive colony inhibitory effect of mEHT was observed after mEHT + GEM treatment. The heat shock induced Hsp27 and Hsp70 proteins, which are known to sensitize PDAC cells to GEM were upregulated in both Capan1 and Panc1 cells 24 h after mEHT treatment. The level of E-Cadherin, a cell adhesion molecule, increased in Capan1 cells after mEHT + GEM treatment. In conclusion, in GEM-sensitive PDAC cells mEHT treatment alone induced cell death and cell cycle inhibition and improved GEM efficiency in combination, which effects were milder and short-term up to 24 h in the GEM-resistant Panc1 cells. Our data further support the inclusion of hyperthermia, in particular of mEHT, into the traditional oncotherapy regimens of PDAC.
Collapse
Affiliation(s)
- Gertrud Forika
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Eva Kiss
- 1st Department of Internal Medicine and Oncology, Oncology Profile, Semmelweis University, Budapest, Hungary
| | - Gabor Petovari
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Titanilla Danko
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Aron Bertram Gellert
- Department of Thoracic Surgery, National Institute of Oncology and Semmelweis University, Budapest, Hungary
| | - Tibor Krenacs
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
- *Correspondence: Tibor Krenacs,
| |
Collapse
|
10
|
Jo JH, Kim SA, Lee JH, Park YR, Kim C, Park SB, Jung DE, Lee HS, Chung MJ, Song SY. GLRX3, a novel cancer stem cell-related secretory biomarker of pancreatic ductal adenocarcinoma. BMC Cancer 2021; 21:1241. [PMID: 34794402 PMCID: PMC8603516 DOI: 10.1186/s12885-021-08898-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/13/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are implicated in carcinogenesis, cancer progression, and recurrence. Several biomarkers have been described for pancreatic ductal adenocarcinoma (PDAC) CSCs; however, their function and mechanism remain unclear. METHOD In this study, secretome analysis was performed in pancreatic CSC-enriched spheres and control adherent cells for biomarker discovery. Glutaredoxin3 (GLRX3), a novel candidate upregulated in spheres, was evaluated for its function and clinical implication. RESULTS PDAC CSC populations, cell lines, patient tissues, and blood samples demonstrated GLRX3 overexpression. In contrast, GLRX3 silencing decreased the in vitro proliferation, migration, clonogenicity, and sphere formation of cells. GLRX3 knockdown also reduced tumor formation and growth in vivo. GLRX3 was found to regulate Met/PI3K/AKT signaling and stemness-related molecules. ELISA results indicated GLRX3 overexpression in the serum of patients with PDAC compared to that in healthy controls. The sensitivity and specificity of GLRX3 for PDAC diagnosis were 80.0 and 100%, respectively. When GLRX3 and CA19-9 were combined, sensitivity was significantly increased to 98.3% compared to that with GLRX3 or CA19-9 alone. High GLRX3 expression was also associated with poor disease-free survival in patients receiving curative surgery. CONCLUSION Overall, these results indicate GLRX3 as a novel diagnostic marker and therapeutic target for PDAC targeting CSCs.
Collapse
MESH Headings
- Animals
- CA-19-9 Antigen/metabolism
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/pathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Disease-Free Survival
- Gene Silencing
- Humans
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Neoplasm Proteins/metabolism
- Neoplastic Stem Cells/metabolism
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/mortality
- Pancreatic Neoplasms/pathology
- Phosphatidylinositol 3-Kinases/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-met/metabolism
- RNA, Small Interfering
- Secretome
- Sensitivity and Specificity
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
Collapse
Affiliation(s)
- Jung Hyun Jo
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Sun A Kim
- Cowell Biodigm Co., Ltd, Seoul, South Korea
| | - Jeong Hoon Lee
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Yu Rang Park
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Chanyang Kim
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Soo Been Park
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Dawoon E Jung
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Hee Seung Lee
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Si Young Song
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
11
|
Attaluri A, Kandala SK, Zhou H, Wabler M, DeWeese TL, Ivkov R. Magnetic nanoparticle hyperthermia for treating locally advanced unresectable and borderline resectable pancreatic cancers: the role of tumor size and eddy-current heating. Int J Hyperthermia 2021; 37:108-119. [PMID: 33426990 PMCID: PMC8363047 DOI: 10.1080/02656736.2020.1798514] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Purpose: Tumor volume largely determines the success of local control of borderline resectable and locally advanced pancreatic cancer with current therapy. We hypothesized that a tumor-mass normalized dose of magnetic nanoparticle hyperthermia (MNPH) with alternating magnetic fields (AMFs) reduces the effect of tumor volume for treatment. Methods: 18 female athymic nude mice bearing subcutaneous MiaPaCa02 human xenograft tumors were treated with MNPH following intratumor injections of 5.5 mg Fe/g tumor of an aqueous suspension of magnetic iron-oxide nanoparticles. Mice were randomly divided into control (n = 5) and treated groups having small (0.15 ± 0.03 cm3, n = 4) or large (0.30 ± 0.06 cm3, n = 5) tumors. We assessed the clinical feasibility of this approach and of pulsed AMF to minimize eddy current heating using a finite-element method to solve a bioheat equation for a human-scale multilayer model. Results: Compared to the control group, both small and large MiaPaCa02 subcutaneous tumors showed statistically significant growth inhibition. Conversely, there was no significant difference in tumor growth between large and small tumors. Both computational and xenograft models demonstrated higher maximum tumor temperatures for large tumors compared to small tumors. Computational modeling demonstrates that pulsed AMF can minimize nonspecific eddy current heating. Conclusions: MNPH provides an advantage to treat large tumors because the MION dose can be adjusted to increase power. Pulsed AMF, with adjusted treatment time, can enhance MNPH in challenging cases such as low MION dose in the target tissue and/or large patients by minimizing nonspecific eddy current heating without sacrificing thermal dose to the target. Nanoparticle heterogeneity in tumors remains a challenge for continued research.
Collapse
Affiliation(s)
- Anilchandra Attaluri
- Department of Mechanical Engineering, School of Science, Engineering, and Technology, The Pennsylvania State University - Harrisburg, Middletown, PA, USA.,Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sri Kamal Kandala
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Haoming Zhou
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michele Wabler
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Theodore L DeWeese
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
12
|
Ghasemi M, Sivaloganathan S. A computational study of combination HIFU-chemotherapy as a potential means of overcoming cancer drug resistance. Math Biosci 2020; 329:108456. [PMID: 32841615 DOI: 10.1016/j.mbs.2020.108456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 08/14/2020] [Accepted: 08/14/2020] [Indexed: 11/18/2022]
Abstract
The application of local hyperthermia, particularly in conjunction with other treatment strategies (like chemotherapy and radiotherapy) has been known to be a useful means of enhancing tumor treatment outcomes. However, to our knowledge, there has been no mathematical model designed to capture the impact of the combination of hyperthermia and chemotherapies on tumor growth and control. In this study, we propose a nonlinear Partial Differential Equation (PDE) model which describes the tumor response to chemotherapy, and use the model to study the effects of hyperthermia on the response of prototypical tumor to the generic chemotherapeutic agent. Ultrasound energy is delivered to the tumor through High Intensity Focused Ultrasound (HIFU), as a noninvasive technique to elevate the tumor temperature in a controlled manner. The proposed tumor growth model is coupled with the nonlinear density dependent Westervelt and Penne's bio-heat equations, used to calculate the net delivered energy and temperature of the tumor and its surrounding normal tissue. The tumor is assumed to be composed of two species: drug-sensitive and drug-resistant. The central assumption underlying our model is that the drug-resistant species is converted to a drug-sensitive type when the tumor temperature is elevated above a certain threshold temperature. The "in silico" results obtained, confirm that hyperthermia can result in less aggressive tumor development and emphasize the importance of designing an optimized thermal dose strategy. Furthermore, our results suggest that increasing the length of the on/off cycle of the transducer is an efficient approach to treatment scheduling in the sense of optimizing tumor eradication.
Collapse
Affiliation(s)
- Maryam Ghasemi
- Department of Applied Mathematics, Univ. Waterloo, Waterloo, ON, Canada, N2L 3G1.
| | | |
Collapse
|
13
|
Significance of unphosphorylated and phosphorylated heat shock protein 27 as a prognostic biomarker in pancreatic ductal adenocarcinoma. J Cancer Res Clin Oncol 2020; 146:1125-1137. [PMID: 32200459 PMCID: PMC7142055 DOI: 10.1007/s00432-020-03175-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/02/2020] [Indexed: 12/28/2022]
Abstract
Purpose Few studies reported about the potential of unphosphorylated heat shock protein 27 (HSP27) and phosphorylated heat shock protein 27 (pHSP27) as a predictor for survival and gemcitabine resistance in pancreatic ductal adenocarcinoma (PDAC). In this study, we analysed the expression patterns of pHSP27 and HSP27 in a patient population after surgery and correlated the immunohistochemical results with clinicopathological data and long-term outcome of the patients. Methods HSP27 and pHSP27 (Ser-15, Ser-78 and Ser-82) protein expression were analysed by immunohistochemistry using the immunoreactive score (IRS) from paraffin-embedded tissue of 106 patients with PDAC who underwent surgery. Immunohistochemical results were correlated with clinicopathological data, disease-free (DFS) and overall survival (OS). Results HSP27 expression was significantly lower in patients with a shorter OS (p = 0.006) and DFS (p < 0.0001). A higher HSP27 expression was associated with a better response to gemcitabine in the resected, non-metastasised patients group (p = 0.001). Furthermore, HSP27 was downregulated in patients suffering from metastases at time of surgery (p < 0.001) and in undifferentiated tumours (p = 0.007). In contrast, pHSP27-Ser15, -Ser78 and -Ser82 were not associated with any survival data of the study population. Conclusion HSP27 seems to be a strong indicator for the prediction of OS and DFS. Moreover, HSP27 could play a role in the formation and migration of liver metastases of PDAC.
Collapse
|
14
|
Wang X, Shan YQ, Tan QQ, Tan CL, Zhang H, Liu JH, Ke NW, Chen YH, Liu XB. MEX3A knockdown inhibits the development of pancreatic ductal adenocarcinoma. Cancer Cell Int 2020; 20:63. [PMID: 32140076 PMCID: PMC7048143 DOI: 10.1186/s12935-020-1146-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/19/2020] [Indexed: 02/08/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDA) is one of the most serious causes of death in the world due to its high mortality and inefficacy treatments. MEX3A was first identified in nematodes and was associated with tumor formation and may promote cell proliferation and tumor metastasis. So far, nothing is known about the relationship between MEX3A and PDA. Methods In this study, the expression level of MEX3A in PDA tissues was measured by immunohistochemistry. The qRT-PCR and western blot were used to identify the constructed MEX3A knockdown cell lines, which was further used to construct mouse xenotransplantation models. Cell proliferation, colony formation, cell apoptosis and migration were detected by MTT, colony formation, flow cytometry and Transwell. Results This study showed that MEX3A expression is significantly upregulated in PDA and associated with tumor grade. Loss-of-function studies showed that downregulation of MEX3A could inhibit cell growth in vitro and in vivo. Moreover, it was demonstrated that knockdown of MEX3A in PDA cells promotes apoptosis by regulating apoptosis-related factors, and inhibits migration through influencing EMT. At the same time, the regulation of PDA progression by MEX3A involves changes in downstream signaling pathways including Akt, p-Akt, PIK3CA, CDK6 and MAPK9. Conclusions We proposed that MEX3A is associated with the prognosis and progression of PDA,which can be used as a potential therapeutic target.
Collapse
Affiliation(s)
- Xing Wang
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Yu-Qiang Shan
- 2Department of Hangzhou First People's Hospital, No. 261, Huansha Road, Hangzhou, 310006 Zhejiang China
| | - Qing-Quan Tan
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Chun-Lu Tan
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Hao Zhang
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Jin-Heng Liu
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Neng-Wen Ke
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Yong-Hua Chen
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Xu-Bao Liu
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| |
Collapse
|
15
|
Cui SJ, Tang TY, Zou XW, Su QM, Feng L, Gong XY. Role of imaging biomarkers for prognostic prediction in patients with pancreatic ductal adenocarcinoma. Clin Radiol 2020; 75:478.e1-478.e11. [PMID: 32037002 DOI: 10.1016/j.crad.2019.12.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/30/2019] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive tumours. PDAC has a poor prognosis; therefore, it is necessary to perform further risk stratification. Identifying prognostic factors before treatment might help to implement suitable and personalised treatment for individuals and avoid side effects. Conventional staging systems and tumour biomarkers are fundamental to establish prognosis; however, they have obvious limitations. Novel imaging biomarkers extracted from advanced imaging techniques offer opportunities to evaluate underlying tumour physiological characteristics, such as mutational status, cellular composition, local microenvironment, tumour metabolism, and biological behaviour. Thus, imaging biomarkers might help the decision making of oncologists and surgeons. The present review discusses the functions of imaging biomarkers for prognostic prediction in patients with PDAC and their potential value for further translation in clinical practice.
Collapse
Affiliation(s)
- S-J Cui
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 310053, Hangzhou, China; Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, 310013, Hangzhou, China
| | - T-Y Tang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - X-W Zou
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Q-M Su
- Department of General Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - L Feng
- Department of Nuclear Medicine, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - X-Y Gong
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, 310013, Hangzhou, China; Institute of Artificial Intelligence and Remote Imaging, Hangzhou Medical College, 310000, Hangzhou, China.
| |
Collapse
|
16
|
Ye W, Li Z, Tang T, Du J, Zhou X, Wu H, Li X, Qin G. ERp29 downregulation enhances lung adenocarcinoma cell chemosensitivity to gemcitabine by upregulating HSP27 phosphorylation. Exp Ther Med 2018; 17:817-823. [PMID: 30651868 DOI: 10.3892/etm.2018.7040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 10/26/2018] [Indexed: 12/23/2022] Open
Abstract
The aim of the current study was to assess the underlying mechanism of endoplasmic reticulum protein 29 (ERp29) in lung adenocarcinoma chemosensitivity to gemcitabine. Western blot analysis was performed to detect ERp29 expression following lung adenocarcinoma cell treatment with gemcitabine. The effects of gemcitabine in combination with ERp29 siRNA on cell apoptosis, cell cycle and heat shock protein 27 (HSP27) expression were assessed. The results demonstrated that ERp29 expression was increased on exposure to gemcitabine. The apoptotic rate of lung adenocarcinoma cells were also increased following gemcitabine treatment and the combined application of gemcitabine and ERp29 siRNA synergistically increased apoptotic rates further. It was also revealed that gemcitabine and ERp29 siRNA synergistically increased the ratio of phosphorylated to total HSP27 protein. In addition, downregulation of HSP27 significantly reduced lung adenocarcinoma chemosensitivity to gemcitabine. These data indicate that ERp29 affects lung adenocarcinoma cell chemosensitivity to gemcitabine by regulating phosphorylated HSP27. ERp29 is a novel target, which may be used to enhance the therapeutic effect of lung adenocarcinoma treatment with gemcitabine.
Collapse
Affiliation(s)
- Wu Ye
- Department of Respiratory Diseases, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Zhijun Li
- Department of Respiratory Diseases, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Tingyu Tang
- Department of Respiratory Diseases, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Jianzong Du
- Department of Respiratory Diseases, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Xiaoxi Zhou
- Department of Respiratory Diseases, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Haiyan Wu
- Department of Respiratory Diseases, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Xuefang Li
- Department of Cardiovascular Medicine, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Guangyue Qin
- Department of Respiratory Diseases, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|
17
|
Kawano M, Kaino S, Amano S, Shinoda S, Suenaga S, Sen-Yo M, Sakaida I. Heat Shock Protein 27 Expression in EUS-FNA Samples Can Predict Gemcitabine Sensitivity in Pancreatic Cancer. ACTA ACUST UNITED AC 2018; 32:637-642. [PMID: 29695571 DOI: 10.21873/invivo.11286] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/02/2018] [Accepted: 03/06/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND/AIM Gemcitabine (GEM) sensitivity can help select the appropriate treatment for pancreatic cancer. We examined the association between HSP27 expression and GEM sensitivity. MATERIALS AND METHODS A total of 19 patients with unresectable pancreatic cancer who underwent endoscopic ultrasonography-guided fine needle aspiration (EUS-FNA) were enrolled and treated with GEM alone. We measured the expression of heat shock protein 27 (HSP27) and phosphorylated HSP27(p-HSP27) in EUS-FNA samples and evaluated the effects of GEM treatment. RESULTS The rate of GEM resistance was significantly higher in patients who showed overexpression of p-HSP27 (p<0.05). When we set the cut-off p-HSP27 (Ser82) detection rate at 51.6%, the group with a detection rate of >51.6% showed a significantly lower survival rate, and GEM was administered for a shorter period of time (p<0.05). CONCLUSION It was suggested that the HSP27 expression in EUS-FNA samples was useful for predicting GEM sensitivity.
Collapse
Affiliation(s)
- Michitaka Kawano
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Seiji Kaino
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Shogo Amano
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Shuhei Shinoda
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Shigeyuki Suenaga
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Manabu Sen-Yo
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Isao Sakaida
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| |
Collapse
|
18
|
KAWANO MICHITAKA, KAINO SEIJI, AMANO SHOGO, SHINODA SHUHEI, SHIGEYUKI SUENAGA, SEN-YO MANABU, SAKAIDA ISAO. Heat Shock Protein 27 Expression in EUS-FNA Samples Can Predict Gemcitabine Sensitivity in Pancreatic Cancer. In Vivo 2018; 32. [PMID: 29695571 PMCID: PMC6000801 DOI: 10.21873/invivo.112286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND/AIM Gemcitabine (GEM) sensitivity can help select the appropriate treatment for pancreatic cancer. We examined the association between HSP27 expression and GEM sensitivity. MATERIALS AND METHODS A total of 19 patients with unresectable pancreatic cancer who underwent endoscopic ultrasonography-guided fine needle aspiration (EUS-FNA) were enrolled and treated with GEM alone. We measured the expression of heat shock protein 27 (HSP27) and phosphorylated HSP27(p-HSP27) in EUS-FNA samples and evaluated the effects of GEM treatment. RESULTS The rate of GEM resistance was significantly higher in patients who showed overexpression of p-HSP27 (p<0.05). When we set the cut-off p-HSP27 (Ser82) detection rate at 51.6%, the group with a detection rate of >51.6% showed a significantly lower survival rate, and GEM was administered for a shorter period of time (p<0.05). CONCLUSION It was suggested that the HSP27 expression in EUS-FNA samples was useful for predicting GEM sensitivity.
Collapse
|
19
|
Ge H, Yan Y, Guo L, Tian F, Wu D. Prognostic role of HSPs in human gastrointestinal cancer: a systematic review and meta-analysis. Onco Targets Ther 2018; 11:351-359. [PMID: 29391812 PMCID: PMC5774472 DOI: 10.2147/ott.s155816] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background Heat shock proteins (HSPs) have been reported to be overexpressed in a wide range of human tumors. It has been shown that HSPs act as an oncogenic regulator and are involved in tumorigenesis. The clinical and prognostic significance of HSPs in gastrointestinal cancers (GICs) remains controversial. The aim of this study was to conduct a meta-analysis to assess the prognostic value of HSPs in GICs. Materials and methods A literature search was performed in PubMed, Cochrane Library, Web of Science, and Embase databases. Data on the relationship between expression of HSPs and survival outcomes were extracted. Pooled hazard ratios (HRs) with 95% CI were calculated. Results The expression of HSPs was not associated with the overall survival (OS) of GIC patients; however, it was significantly associated with worse OS for gastric cancer (GC) and colorectal cancer (CRC) patients. Conclusion Current evidence suggests that a high level of HSPs may not be a potential marker to predict the survival rate for every type of GICs. However, the expression of HSPs may predict a poor prognosis for GC and CRC patients.
Collapse
Affiliation(s)
- Hua Ge
- Department of Gastrointestinal Surgery
| | - Yan Yan
- Quality Control Department, The First People's Hospital of Zunyi, Zunyi Medical University, Huichuan, Zunyi, Guizhou, China
| | | | - Fei Tian
- Department of Gastrointestinal Surgery
| | - Di Wu
- Department of Gastrointestinal Surgery
| |
Collapse
|
20
|
Moussavi M, Haddad F, Matin MM, Iranshahi M, Rassouli FB. Efficacy of hyperthermia in human colon adenocarcinoma cells is improved by auraptene. Biochem Cell Biol 2017; 96:32-37. [PMID: 28915362 DOI: 10.1139/bcb-2017-0146] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Colon adenocarcinoma is one of the most common cancers worldwide, and resistance to current therapeutic modalities is a serious drawback in its treatment. Auraptene is a natural coumarin with considerable anticancer effects. The goal of this study was to introduce a novel combinatorial approach for treatment against colon adenocarcinoma cells. To do so, HT29 cells were pretreated with nontoxic auraptene and then hyperthermia was induced. Afterwards, the viability of the cells was assessed, changes induced in the cell cycle were analyzed, and the expression patterns of candidate genes were studied. Results from the MTT assay demonstrated significant (p < 0.01) decreases in cell viability when 20 μg/mL auraptene was used for 72 h, heat shock was induced, and cells were allowed to recover for 24 h. Flow cytometry analysis also indicated considerable changes in the distribution of cells between the sub-G1/G1 and G2/M phases of cell cycle after the combinatorial treatment. Real-time RT-PCR studies revealed significant (p < 0.01) up-regulation of P21 in the cells pretreated with auraptene after heat shock, whereas no significant change was observed in HSP27 expression. Our findings not only indicate, for the first time, that the efficacy of hyperthermia was improved by auraptene pretreatment, but also suggest that this coumarin could be used in the future to achieve more effective therapeutic outcomes.
Collapse
Affiliation(s)
- Mahdi Moussavi
- a Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Farhang Haddad
- a Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maryam M Matin
- a Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.,b Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mehrdad Iranshahi
- c Department of Pharmacognosy and Biotechnology, Biotechnology Research Center, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh B Rassouli
- a Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.,b Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
21
|
Ooi M, Phan A, Nguyen NQ. Future role of endoscopic ultrasound in personalized management of pancreatic cancer. Endosc Ultrasound 2017; 6:300-307. [PMID: 29063873 PMCID: PMC5664850 DOI: 10.4103/eus.eus_84_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 09/08/2017] [Indexed: 12/27/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is aggressive and lethal with the majority of cases presenting with advanced unresectable disease due to delayed diagnosis. Despite improvement in surgery, chemotherapies, and intensive care medicine, the outcome of PDAC remains poor, which may relate to the tumor biology. Recent data suggest that PDAC is a "systemic cancer" with complex molecular or genomics derangement with marked heterogeneity. The ability to characterize the PDAC better by detailed evaluation of tissue biomarkers or genomics allows for improved prediction of prognosis and stratification of treatment, a concept known as "personalized cancer therapy." Using tissue from resected PDAC specimens has several weaknesses and is only possible in 20% of patients with PDAC. Endoscopic ultrasound (EUS)-guided biopsy overcomes these weaknesses, and with recent advancements in needle technology, tissue can be obtained for personalized cancer therapy for all patients with PDAC. This review aims to outline our current understanding of the molecular biology of PDAC specifically focusing on how EUS-guided biopsy may play a fundamental role in tissue acquisition, allowing for assessment and stratify therapy according to the individual cancer biology as we move toward the era of precision medicine.
Collapse
Affiliation(s)
- Marie Ooi
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, Australia
| | - An Phan
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Nam Q. Nguyen
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| |
Collapse
|
22
|
Li M, Radvanyi L, Yin B, Rycaj K, Li J, Chivukula R, Lin K, Lu Y, Shen J, Chang DZ, Li D, Johanning GL, Wang-Johanning F. Downregulation of Human Endogenous Retrovirus Type K (HERV-K) Viral env RNA in Pancreatic Cancer Cells Decreases Cell Proliferation and Tumor Growth. Clin Cancer Res 2017; 23:5892-5911. [PMID: 28679769 DOI: 10.1158/1078-0432.ccr-17-0001] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 05/09/2017] [Accepted: 06/29/2017] [Indexed: 12/23/2022]
Abstract
Purpose: We investigated the role of the human endogenous retrovirus type K (HERV-K) envelope (env) gene in pancreatic cancer.Experimental Design: shRNA was employed to knockdown (KD) the expression of HERV-K in pancreatic cancer cells.Results: HERV-K env expression was detected in seven pancreatic cancer cell lines and in 80% of pancreatic cancer patient biopsies, but not in two normal pancreatic cell lines or uninvolved normal tissues. A new HERV-K splice variant was discovered in several pancreatic cancer cell lines. Reverse transcriptase activity and virus-like particles were observed in culture media supernatant obtained from Panc-1 and Panc-2 cells. HERV-K viral RNA levels and anti-HERV-K antibody titers were significantly higher in pancreatic cancer patient sera (N = 106) than in normal donor sera (N = 40). Importantly, the in vitro and in vivo growth rates of three pancreatic cancer cell lines were significantly reduced after HERV-K KD by shRNA targeting HERV-K env, and there was reduced metastasis to lung after treatment. RNA-Seq results revealed changes in gene expression after HERV-K env KD, including RAS and TP53. Furthermore, downregulation of HERV-K Env protein expression by shRNA also resulted in decreased expression of RAS, p-ERK, p-RSK, and p-AKT in several pancreatic cancer cells or tumors.Conclusions: These results demonstrate that HERV-K influences signal transduction via the RAS-ERK-RSK pathway in pancreatic cancer. Our data highlight the potentially important role of HERV-K in tumorigenesis and progression of pancreatic cancer, and indicate that HERV-K viral proteins may be attractive biomarkers and/or tumor-associated antigens, as well as potentially useful targets for detection, diagnosis, and immunotherapy of pancreatic cancer. Clin Cancer Res; 23(19); 5892-911. ©2017 AACR.
Collapse
Affiliation(s)
- Ming Li
- Viral Oncology Program, Center for Cancer and Metabolism, SRI International, Menlo Park, California
| | - Laszlo Radvanyi
- EMD Serono Research and Development Institute, Billerica, Massachusetts
| | - Bingnan Yin
- Department of Inflammation and Epigenetics, Methodist Research Institute, Houston, Texas
| | | | - Jia Li
- Viral Oncology Program, Center for Cancer and Metabolism, SRI International, Menlo Park, California
| | - Raghavender Chivukula
- Viral Oncology Program, Center for Cancer and Metabolism, SRI International, Menlo Park, California
| | - Kevin Lin
- Department of Epigenetics and Molecular Carcinogenesis, Science Park, the University of Texas MD Anderson Cancer Center, Smithville, Texas
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, Science Park, the University of Texas MD Anderson Cancer Center, Smithville, Texas
| | - JianJun Shen
- Department of Epigenetics and Molecular Carcinogenesis, Science Park, the University of Texas MD Anderson Cancer Center, Smithville, Texas
| | - David Z Chang
- Virginia Oncology Associates, Newport News, Virginia
| | - Donghui Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gary L Johanning
- Viral Oncology Program, Center for Cancer and Metabolism, SRI International, Menlo Park, California
| | - Feng Wang-Johanning
- Viral Oncology Program, Center for Cancer and Metabolism, SRI International, Menlo Park, California.
| |
Collapse
|
23
|
Totti S, Vernardis SI, Meira L, Pérez-Mancera PA, Costello E, Greenhalf W, Palmer D, Neoptolemos J, Mantalaris A, Velliou EG. Designing a bio-inspired biomimetic in vitro system for the optimization of ex vivo studies of pancreatic cancer. Drug Discov Today 2017; 22:690-701. [PMID: 28153670 DOI: 10.1016/j.drudis.2017.01.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/16/2016] [Accepted: 01/18/2017] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer is one of the most aggressive and lethal human malignancies. Drug therapies and radiotherapy are used for treatment as adjuvants to surgery, but outcomes remain disappointing. Advances in tissue engineering suggest that 3D cultures can reflect the in vivo tumor microenvironment and can guarantee a physiological distribution of oxygen, nutrients, and drugs, making them promising low-cost tools for therapy development. Here, we review crucial structural and environmental elements that should be considered for an accurate design of an ex vivo platform for studies of pancreatic cancer. Furthermore, we propose environmental stress response biomarkers as platform readouts for the efficient control and further prediction of the pancreatic cancer response to the environmental and treatment input.
Collapse
Affiliation(s)
- Stella Totti
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK
| | - Spyros I Vernardis
- Biological Systems Engineering Laboratory (BSEL), Department of Chemical Engineering, Imperial College London, SW7 2AZ London, UK
| | - Lisiane Meira
- Department of Clinical and Experimental Medicine, University of Surrey, Guildford GU2 7XH, UK
| | - Pedro A Pérez-Mancera
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool,Daulby Street, Liverpool L69 3GA, UK
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool,Daulby Street, Liverpool L69 3GA, UK; NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool,Daulby Street, Liverpool L69 3GA, UK
| | - William Greenhalf
- NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool,Daulby Street, Liverpool L69 3GA, UK
| | - Daniel Palmer
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool,Daulby Street, Liverpool L69 3GA, UK
| | - John Neoptolemos
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool,Daulby Street, Liverpool L69 3GA, UK; NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool,Daulby Street, Liverpool L69 3GA, UK
| | - Athanasios Mantalaris
- Biological Systems Engineering Laboratory (BSEL), Department of Chemical Engineering, Imperial College London, SW7 2AZ London, UK
| | - Eirini G Velliou
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK.
| |
Collapse
|
24
|
Systematic review of peri-operative prognostic biomarkers in pancreatic ductal adenocarcinoma. HPB (Oxford) 2016; 18:652-63. [PMID: 27485059 PMCID: PMC4972371 DOI: 10.1016/j.hpb.2016.05.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 05/09/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) continues to be associated with a poor prognosis. This systematic review aimed to summarize the literature regarding potential prognostic biomarkers to facilitate validation studies and clinical application. METHODS A systematic review was performed (2004-2014) according to PRISMA guidelines. Studies were ranked using REMARK criteria and the following outcomes were examined: overall/disease free survival, nodal involvement, tumour characteristics, metastasis, recurrence and resectability. RESULTS 256 biomarkers were identified in 158 studies. 171 biomarkers were assessed with respect to overall survival: urokinase-type plasminogen activator receptor, atypical protein kinase C and HSP27 ranked the highest. 33 biomarkers were assessed for disease free survival: CD24 and S100A4 were the highest ranking. 17 biomarkers were identified for lymph node involvement: Smad4/Dpc4 and FOXC1 ranked highest. 13 biomarkers were examined for tumour grade: mesothelin and EGFR were the highest ranking biomarkers. 10 biomarkers were identified for metastasis: p16 and sCD40L were the highest ranking. 4 biomarkers were assessed resectability: sCD40L, s100a2, Ca 19-9, CEA. CONCLUSION This review has identified and ranked specific biomarkers that should be a primary focus of ongoing validation and clinical translational work in PDAC.
Collapse
|
25
|
The Clinical Significance of Phosphorylated Heat Shock Protein 27 (HSPB1) in Pancreatic Cancer. Int J Mol Sci 2016; 17:ijms17010137. [PMID: 26805817 PMCID: PMC4730376 DOI: 10.3390/ijms17010137] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/14/2016] [Accepted: 01/15/2016] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer is one of most aggressive forms of cancer. After clinical detection it exhibits fast metastatic growth. Heat shock protein 27 (HSP27; HSPB1) has been characterized as a molecular chaperone which modifies the structures and functions of other proteins in cells when they are exposed to various stresses, such as chemotherapy. While the administration of gemcitabine, an anti-tumor drug, has been the standard treatment for patients with advanced pancreatic cancer, accumulating evidence shows that HSP27 plays a key role in the chemosensitivity to gemcitabine. In addition, phosphorylated HSP27 induced by gemcitabine has been associated with the inhibition of pancreatic cancer cell growth. In this review, we summarize the role of phosphorylated HSP27, as well as HSP27, in the regulation of chemosensitivity in pancreatic cancer.
Collapse
|
26
|
Abstract
OBJECTIVES To evaluate the regulation mechanism of heat shock protein 27 (HSP27) on gemcitabine (GEM) resistance of pancreatic cancer cell. METHODS The expression vectors pEGFP-C1-HSP27 and the vectors of MicroRNA targeting Snail were introduced into GEM-sensitive pancreatic cancer SW1990 cells, and the vectors of small hairpin RNA targeting HSP27 were transfected into SW1990 and GEM-resistant SW1990/GEM cells. The expressions of HSP27, p-HSP27 (Ser82), Snail, ERCC1, and E-cadherin were evaluated by Western blotting. The sensitivity of transfected cells to GEM was detected by CCK-8 assay and Annexin V-FITC apoptosis assay. RESULTS As compared to SW1990, SW1990/GEM showed significantly increased expressions of HSP27, p-HSP27, Snail and ERCC1 with decreased expression of E-cadherin. By increasing HSP27 expression, we found increase of Snail and ERCC1 with reduction of E-cadherin expressions, while reduction of HSP27 expression caused reduction of Snail and ERCC1 but increase of E-cadherin expressions. Downregulation of Snail resulted in the reduction of ERCC1 expression and increase of E-cadherin. Furthermore, downregulation of HSP27 or snail caused increased GEM sensitivity of pancreatic cancer cells, and upregulation of HSP27 showed the opposite results. CONCLUSIONS There is an inverse correlation between HSP27 expression and GEM sensitivity of SW1990 cells, which might be realized by regulating E-cadherin and ERCC1 expressions through Snail.
Collapse
|
27
|
Kobayashi S, Ueno M, Irie K, Goda Y, Aoyama T, Morinaga S, Ohkawa S, Morimoto M. Potential prognostic significance of a new proteomic profile in patients with advanced pancreatic adenocarcinoma. Pancreatology 2015; 15:525-530. [PMID: 26255025 DOI: 10.1016/j.pan.2015.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 07/07/2015] [Accepted: 07/12/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Seven-signal proteomic approach has recently been developed as a new proteomic profile measured by matrix-assisted laser desorption/ionization mass spectrometry. The aim of this study was to evaluate prognostic significance of this proteomic value in patients with pancreatic adenocarcinoma. METHODS Blood samples from the patients with pancreatic adenocarcinoma were prospectively collected before treatments including surgical resection and systemic chemotherapies. The seven-signal proteomic profiles of the samples were measured, and the prognostic significance of the proteomic value was evaluated through comparison with other existing prognostic markers. RESULTS Cut-off value of the proteomic profiles at 52 stratified overall prognosis of the patients (6.5 months vs. 10.9 months with the values ≥52 vs. <52, p = 0.020). In subgroup analyses of inoperable cases with carcinoembryonic antigen level of <5 ng/ml or performance status of 0-1, the proteomic value at 52 stratified their prognosis (p = 0.002 and p = 0.006, respectively). CONCLUSIONS The new seven-signal proteomics showed useful prognostic significance for patients with pancreatic adenocarcinoma. Further studies with a large sample size would be required to evaluate whether this proteomic approach possibly complements the existing parameters, such as carcinoembryonic antigen and performance status.
Collapse
Affiliation(s)
- Satoshi Kobayashi
- Division of Hepatobiliary and Pancreatic Medical Oncology, Kanagawa Cancer Center, Japan.
| | - Makoto Ueno
- Division of Hepatobiliary and Pancreatic Medical Oncology, Kanagawa Cancer Center, Japan
| | - Kuniyasu Irie
- Division of Hepatobiliary and Pancreatic Medical Oncology, Kanagawa Cancer Center, Japan
| | - Yoshihiro Goda
- Division of Hepatobiliary and Pancreatic Medical Oncology, Kanagawa Cancer Center, Japan
| | - Toru Aoyama
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Japan
| | - Soichiro Morinaga
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Japan
| | - Shinichi Ohkawa
- Division of Hepatobiliary and Pancreatic Medical Oncology, Kanagawa Cancer Center, Japan
| | - Manabu Morimoto
- Division of Hepatobiliary and Pancreatic Medical Oncology, Kanagawa Cancer Center, Japan
| |
Collapse
|
28
|
Guo Y, Ziesch A, Hocke S, Kampmann E, Ochs S, De Toni EN, Göke B, Gallmeier E. Overexpression of heat shock protein 27 (HSP27) increases gemcitabine sensitivity in pancreatic cancer cells through S-phase arrest and apoptosis. J Cell Mol Med 2014; 19:340-50. [PMID: 25331547 PMCID: PMC4407596 DOI: 10.1111/jcmm.12444] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 08/25/2014] [Indexed: 02/06/2023] Open
Abstract
We previously established a role for HSP27 as a predictive marker for therapeutic response towards gemcitabine in pancreatic cancer. Here, we investigate the underlying mechanisms of HSP27-mediated gemcitabine sensitivity. Utilizing a pancreatic cancer cell model with stable HSP27 overexpression, cell cycle arrest and apoptosis induction were analysed by flow cytometry, nuclear staining, immunoblotting and mitochondrial staining. Drug sensitivity studies were performed by proliferation assays. Hyperthermia was simulated using mild heat shock at 41.8°C. Upon gemcitabine treatment, HSP27-overexpressing cells displayed an early S-phase arrest subsequently followed by a strongly increased sub-G1 fraction. Apoptosis was characterized by PARP-, CASPASE 3-, CASPASE 8-, CASPASE 9- and BIM- activation along with a mitochondrial membrane potential loss. It was reversible through chemical caspase inhibition. Importantly, gemcitabine sensitivity and PARP cleavage were also elicited by heat shock-induced HSP27 overexpression, although to a smaller extent, in a panel of pancreatic cancer cell lines. Finally, HSP27-overexpressing pancreatic cancer cells displayed an increased sensitivity also towards death receptor-targeting agents, suggesting another pro-apoptotic role of HSP27 along the extrinsic apoptosis pathway. Taken together, in contrast to the well-established anti-apoptotic properties of HSP27 in cancer, our study reveals novel pro-apoptotic functions of HSP27—mediated through both the intrinsic and the extrinsic apoptotic pathways—at least in pancreatic cancer cells. HSP27 could represent a predictive marker of therapeutic response towards specific drug classes in pancreatic cancer and provides a novel molecular rationale for current clinical trials applying the combination of gemcitabine with regional hyperthermia in pancreatic cancer patients.
Collapse
Affiliation(s)
- Yang Guo
- Department of Medicine II, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Marengo E, Robotti E. Biomarkers for pancreatic cancer: Recent achievements in proteomics and genomics through classical and multivariate statistical methods. World J Gastroenterol 2014; 20:13325-13342. [PMID: 25309068 PMCID: PMC4188889 DOI: 10.3748/wjg.v20.i37.13325] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 06/04/2014] [Accepted: 06/26/2014] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most aggressive and lethal neoplastic diseases. A valid alternative to the usual invasive diagnostic tools would certainly be the determination of biomarkers in peripheral fluids to provide less invasive tools for early diagnosis. Nowadays, biomarkers are generally investigated mainly in peripheral blood and tissues through high-throughput omics techniques comparing control vs pathological samples. The results can be evaluated by two main strategies: (1) classical methods in which the identification of significant biomarkers is accomplished by monovariate statistical tests where each biomarker is considered as independent from the others; and (2) multivariate methods, taking into consideration the correlations existing among the biomarkers themselves. This last approach is very powerful since it allows the identification of pools of biomarkers with diagnostic and prognostic performances which are superior to single markers in terms of sensitivity, specificity and robustness. Multivariate techniques are usually applied with variable selection procedures to provide a restricted set of biomarkers with the best predictive ability; however, standard selection methods are usually aimed at the identification of the smallest set of variables with the best predictive ability and exhaustivity is usually neglected. The exhaustive search for biomarkers is instead an important alternative to standard variable selection since it can provide information about the etiology of the pathology by producing a comprehensive set of markers. In this review, the most recent applications of the omics techniques (proteomics, genomics and metabolomics) to the identification of exploratory biomarkers for PC will be presented with particular regard to the statistical methods adopted for their identification. The basic theory related to classical and multivariate methods for identification of biomarkers is presented and then, the most recent applications in this field are discussed.
Collapse
|
30
|
Köpper F, Binkowski AM, Bierwirth C, Dobbelstein M. The MAPK-activated protein kinase 2 mediates gemcitabine sensitivity in pancreatic cancer cells. Cell Cycle 2014; 13:884-9. [PMID: 24556918 PMCID: PMC3984311 DOI: 10.4161/cc.28292] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/18/2014] [Accepted: 02/19/2014] [Indexed: 01/06/2023] Open
Abstract
Pancreatic carcinoma is the major clinical entity where the nucleoside analog gemcitabine is used for first-line therapy. Overcoming cellular resistance toward gemcitabine remains a major challenge in this context. This raises the need to identify factors that determine gemcitabine sensitivity in pancreatic carcinoma cells. We previously found the MAPK-activated protein kinase 2 (MK2), part of the p38/MK2 stress response pathway, to be required for DNA replication fork stalling when osteosarcoma-derived cells were treated with gemcitabine. As a consequence, inhibition or depletion of MK2 protects these cells from gemcitabine-induced death (Köpper, et al. Proc Natl Acad Sci USA 2013; 110:16856-61). Here, we addressed whether MK2 also determines the sensitivity of pancreatic cancer cells toward gemcitabine. We found that MK2 inhibition reduced the intensity of the DNA damage response and enhanced survival of the pancreatic cancer cell lines BxPC-3, MIA PaCa-2, and Panc-1, which display a moderate to strong sensitivity to gemcitabine. In contrast, MK2 inhibition only weakly attenuated the DNA damage response intensity and did not enhance long-term survival in the gemcitabine-resistant cell line PaTu 8902. Importantly, in BxPC-3 and MIA PaCa-2 cells, inhibition of MK2 also rescued increased H2AX phosphorylation caused by inhibition of the checkpoint kinase Chk1 in the presence of gemcitabine. These results indicate that MK2 mediates gemcitabine efficacy in pancreatic cancer cells that respond to the drug, suggesting that the p38/MK2 pathway represents a determinant of the efficacy by that gemcitabine counteracts pancreatic cancer.
Collapse
Affiliation(s)
- Frederik Köpper
- Institute of Molecular Oncology; Göttingen Centre of Molecular Biosciences (GZMB); Faculty of Medicine; University of Göttingen; Göttingen, Germany
| | - Anna Maria Binkowski
- Institute of Molecular Oncology; Göttingen Centre of Molecular Biosciences (GZMB); Faculty of Medicine; University of Göttingen; Göttingen, Germany
| | - Cathrin Bierwirth
- Institute of Molecular Oncology; Göttingen Centre of Molecular Biosciences (GZMB); Faculty of Medicine; University of Göttingen; Göttingen, Germany
| | - Matthias Dobbelstein
- Institute of Molecular Oncology; Göttingen Centre of Molecular Biosciences (GZMB); Faculty of Medicine; University of Göttingen; Göttingen, Germany
| |
Collapse
|
31
|
Arrigo AP, Gibert B. HspB1, HspB5 and HspB4 in Human Cancers: Potent Oncogenic Role of Some of Their Client Proteins. Cancers (Basel) 2014; 6:333-65. [PMID: 24514166 PMCID: PMC3980596 DOI: 10.3390/cancers6010333] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/03/2014] [Accepted: 01/17/2014] [Indexed: 12/20/2022] Open
Abstract
Human small heat shock proteins are molecular chaperones that regulate fundamental cellular processes in normal unstressed cells as well as in many cancer cells where they are over-expressed. These proteins are characterized by cell physiology dependent changes in their oligomerization and phosphorylation status. These structural changes allow them to interact with many different client proteins that subsequently display modified activity and/or half-life. Nowdays, the protein interactomes of small Hsps are under intense investigations and will represent, when completed, key parameters to elaborate therapeutic strategies aimed at modulating the functions of these chaperones. Here, we have analyzed the potential pro-cancerous roles of several client proteins that have been described so far to interact with HspB1 (Hsp27) and its close members HspB5 (αB-crystallin) and HspB4 (αA-crystallin).
Collapse
Affiliation(s)
- André-Patrick Arrigo
- Apoptosis, Cancer and Development Laboratory, Lyon Cancer Research Center, INSERM U1052-CNRS UMR5286, Claude Bernard University Lyon 1, Lyon 69008, France.
| | - Benjamin Gibert
- Apoptosis, Cancer and Development Laboratory, Lyon Cancer Research Center, INSERM U1052-CNRS UMR5286, Claude Bernard University Lyon 1, Lyon 69008, France.
| |
Collapse
|
32
|
Zhang P, Zou M, Wen X, Gu F, Li J, Liu G, Dong J, Deng X, Gao J, Li X, Jia X, Dong Z, Chen L, Wang Y, Tian Y. Development of serum parameters panels for the early detection of pancreatic cancer. Int J Cancer 2013; 134:2646-55. [PMID: 24615168 DOI: 10.1002/ijc.28584] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 10/24/2013] [Accepted: 10/25/2013] [Indexed: 12/24/2022]
Abstract
Early detection of pancreatic cancer is promising for improving clinical outcome; however, no effective biomarker has yet been identified. Here, we detected 61 clinical serum parameters in 200 healthy controls (Ctrls), 163 pancreatic ductal adenocarcinoma (PDAC) patients and 109 benign pancreatitis patients (Benign) in the training group. A metropolis algorithm with Monte Carlo simulation was used for identifying parameter panels. Sera from 183 Ctrl, 129 PDAC and 95 Benign individuals were used for cross-validation. Samples from 77 breast, 72 cervical, 101 colorectal, 138 gastric, 108 prostate and 132 lung cancer patients were collected for evaluating cancer selectivity. A panel consisting of carbohydrate antigen (CA)19-9, albumin (ALB), C-reactive protein (CRP) and interleukin (IL)-8 had the highest diagnostic value for discriminating between PDAC and Ctrl. The sensitivity (SN) was 99.39% for all-stage, 96.10% for early-stage and 98.80% for advanced-stage PDAC at 90% specificity (SP). In the validation group, the sensitivities were 93.80, 93.10 and 94.40%, respectively, at 90% SP. This panel also identified 80.52% of the breast cancer, 66.67% cervical cancer, 86.14% colorectal cancer, 89.86% gastric cancer, 71.30% prostate cancer and 93.85% lung cancer samples as non-PDAC. The panel consisting of CA19-9, carbon dioxide, CRP and IL-6 panel had the highest diagnostic value for discriminating between PDAC and Benign. The SN was 74.23% for all-stage, 75.30% for early-stage and 74.40% for advanced-stage PDAC at 90% SP. In the validation group, the sensitivities were 72.10, 76.10 and 67.20%, respectively, at 90% SP. Our parameter panels may aid in the early detection of PDAC to improve clinical outcome.
Collapse
Affiliation(s)
- Pengjun Zhang
- Department of Clinical Biochemistry, State Key Laboratory of Kidney Disease, Chinese PLA General Hospital, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Li M, Yu X, Guo H, Sun L, Wang A, Liu Q, Wang X, Li J. Bufalin exerts antitumor effects by inducing cell cycle arrest and triggering apoptosis in pancreatic cancer cells. Tumour Biol 2013. [PMID: 24218335 DOI: 10.1007/s13227-013-1326-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
As one of the most aggressive human malignancies, pancreatic cancer is a leading cause of cancer-related deaths worldwide and only about 4% of patients will live 5 years after diagnosis. Eighty to approximately eighty-five percent of patients are diagnosed with an unresectable or metastatic disease, which is correlated with poor prognosis and low survival rate. Therefore, it is tremendously significant to exploit novel chemicals to prevent and treat pancreatic cancer. Previous research and clinical studies have demonstrated that many natural products derived from traditional Chinese medicine (TCM) such as camptothecin derivatives and vinca alkaloids could be effective antitumor compounds, hinting that TCM is a promising source for developing new antitumor drugs. In this report, we investigated the effects of bufalin, a primary active ingredient of the traditional Chinese medicine Chan-Su, on pancreatic cancer cell lines PANC-1 and CFPAC-1 and studied the underlying molecular mechanism. We found that exposure to bufalin could suppress the proliferation of pancreatic cancer cells time and dose dependently. We used flow cytometry to study the effects of bufalin on apoptosis and cell cycle distribution in PANC-1 and CFPAC-1 cells. The results indicated that bufalin could significantly induce both apoptosis and G2/M cell cycle arrest in pancreatic cancer cells. With western blotting, we found that the expression level of an antiapoptotic protein heat shock protein 27 (Hsp27) and its partner molecule p-Akt was decreased upon the treatment with bufalin. Besides, bufalin activated pro-caspase-3 and pro-caspase-9 and modulated the expression level of Bcl-2 and Bax. These data suggested that bufalin may trigger apoptosis by targeting Hsp27, which could inhibit apoptosis by interfering with key apoptotic proteins. The influence on the level of cylinB1, CDK1, and p21 was also observed after bufalin treatment, and the relationship between Hsp27 and the cell cycle-related proteins mentioned above deserves much more research. In addition, our data showed that bufalin could enhance the growth inhibition effect of gemcitabine in above pancreatic cancer cells. Taken together, bufalin might be worthy of further study for its potential as a therapeutic agent for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Meiying Li
- Department of Medical Oncology, Cancer Center, Qilu Hospital, Shandong University, Jinan, 250012, China
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Bufalin exerts antitumor effects by inducing cell cycle arrest and triggering apoptosis in pancreatic cancer cells. Tumour Biol 2013; 35:2461-71. [PMID: 24218335 DOI: 10.1007/s13277-013-1326-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 10/14/2013] [Indexed: 12/12/2022] Open
Abstract
As one of the most aggressive human malignancies, pancreatic cancer is a leading cause of cancer-related deaths worldwide and only about 4% of patients will live 5 years after diagnosis. Eighty to approximately eighty-five percent of patients are diagnosed with an unresectable or metastatic disease, which is correlated with poor prognosis and low survival rate. Therefore, it is tremendously significant to exploit novel chemicals to prevent and treat pancreatic cancer. Previous research and clinical studies have demonstrated that many natural products derived from traditional Chinese medicine (TCM) such as camptothecin derivatives and vinca alkaloids could be effective antitumor compounds, hinting that TCM is a promising source for developing new antitumor drugs. In this report, we investigated the effects of bufalin, a primary active ingredient of the traditional Chinese medicine Chan-Su, on pancreatic cancer cell lines PANC-1 and CFPAC-1 and studied the underlying molecular mechanism. We found that exposure to bufalin could suppress the proliferation of pancreatic cancer cells time and dose dependently. We used flow cytometry to study the effects of bufalin on apoptosis and cell cycle distribution in PANC-1 and CFPAC-1 cells. The results indicated that bufalin could significantly induce both apoptosis and G2/M cell cycle arrest in pancreatic cancer cells. With western blotting, we found that the expression level of an antiapoptotic protein heat shock protein 27 (Hsp27) and its partner molecule p-Akt was decreased upon the treatment with bufalin. Besides, bufalin activated pro-caspase-3 and pro-caspase-9 and modulated the expression level of Bcl-2 and Bax. These data suggested that bufalin may trigger apoptosis by targeting Hsp27, which could inhibit apoptosis by interfering with key apoptotic proteins. The influence on the level of cylinB1, CDK1, and p21 was also observed after bufalin treatment, and the relationship between Hsp27 and the cell cycle-related proteins mentioned above deserves much more research. In addition, our data showed that bufalin could enhance the growth inhibition effect of gemcitabine in above pancreatic cancer cells. Taken together, bufalin might be worthy of further study for its potential as a therapeutic agent for pancreatic cancer treatment.
Collapse
|
35
|
B7H4, HSP27 and DJ-1 molecular markers as prognostic factors in pancreatic cancer. Pancreatology 2013; 13:564-9. [PMID: 24280570 DOI: 10.1016/j.pan.2013.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/01/2013] [Accepted: 10/14/2013] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Pancreatic cancer (PC) is one of the most lethal tumors of the gastrointestinal tract. The ability to predict which patients would benefit most from surgical intervention and chemotherapy would be a great clinical tool. A large number of potential markers have been identified lately in pancreatic cancer and their clinical utilities as prognostic tools are under investigation. METHODS We recruited 41 patients who had undergone radical surgical resection for PC between 2003 and 2010. To investigate the prognostic factors, we evaluated 3 possible markers: B7H4, HSP27 and DJ-1 protein expressions in the tissue specimens of these 41 patients by immunohistochemistry and analyzed the clinical and pathological features of these specimens. RESULTS The expression of the three antigens was independently associated with a negative impact of chemotherapy with gemcitabine on patient's survival. Moreover, patients who overexpressed B7H4 had worse prognosis than the ones who did not. CONCLUSIONS B7H4, DJ-1 and HSP27 may be used in the future as prognostic markers that express resistance of pancreatic cancer patients to chemotherapy with gemcitabine.
Collapse
|
36
|
Gallmeier E, Bader DC, Kriegl L, Berezowska S, Seeliger H, Göke B, Kirchner T, Bruns C, De Toni EN. Loss of TRAIL-receptors is a recurrent feature in pancreatic cancer and determines the prognosis of patients with no nodal metastasis after surgery. PLoS One 2013; 8:e56760. [PMID: 23460812 PMCID: PMC3584093 DOI: 10.1371/journal.pone.0056760] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 01/14/2013] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Agonistic antibodies targeting TRAIL-receptors 1 and 2 (TRAIL-R1 and TRAIL-R2) are being developed as a novel therapeutic approach in cancer therapy including pancreatic cancer. However, the cellular distribution of these receptors in primary pancreatic cancer samples has not been sufficiently investigated and no study has yet addressed the issue of their prognostic significance in this tumor entity. AIMS AND METHODS Applying tissue microarray (TMA) analysis, we performed an immunohistochemical assessment of TRAIL-receptors in surgical samples from 84 consecutive patients affected by pancreatic adenocarcinoma and in 26 additional selected specimens from patients with no lymph nodes metastasis at the time of surgery. The prognostic significance of membrane staining and staining intensity for TRAIL-receptors was evaluated. RESULTS The fraction of pancreatic cancer samples with positive membrane staining for TRAIL-R1 and TRAIL-R2 was lower than that of cells from surrounding non-tumor tissues (TRAIL-R1: p<0.001, TRAIL-R2: p = 0.006). In addition, subgroup analyses showed that loss of membrane staining for TRAIL-R2 was associated with poorer prognosis in patients without nodal metastases (multivariate Cox regression analysis, Hazard Ratio: 0.44 [95% confidence interval: 0.22-0.87]; p = 0.019). In contrast, analysis of decoy receptors TRAIL-R3 and -R4 in tumor samples showed an exclusively cytoplasmatic staining pattern and no prognostic relevance. CONCLUSION This is a first report on the prognostic significance of TRAIL-receptors expression in pancreatic cancer showing that TRAIL-R2 might represent a prognostic marker for patients with early stage disease. In addition, our data suggest that loss of membrane-bound TRAIL-receptors could represent a molecular mechanism for therapeutic failure upon administration of TRAIL-receptors-targeting antibodies in pancreatic cancer. This hypothesis should be evaluated in future clinical trials.
Collapse
Affiliation(s)
- Eike Gallmeier
- Department of Medicine 2, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Dominik C. Bader
- Department of Medicine 2, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Lydia Kriegl
- Institute of Pathology, University of Munich, Munich, Germany
| | | | - Hendrik Seeliger
- Department of Surgery, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Burkhard Göke
- Department of Medicine 2, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Thomas Kirchner
- Institute of Pathology, University of Munich, Munich, Germany
| | - Christiane Bruns
- Department of Surgery, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Enrico N. De Toni
- Department of Medicine 2, University Hospital Grosshadern, University of Munich, Munich, Germany
| |
Collapse
|
37
|
Storr SJ, Zaitoun AM, Arora A, Durrant LG, Lobo DN, Madhusudan S, Martin SG. Calpain system protein expression in carcinomas of the pancreas, bile duct and ampulla. BMC Cancer 2012; 12:511. [PMID: 23140395 PMCID: PMC3542103 DOI: 10.1186/1471-2407-12-511] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 11/06/2012] [Indexed: 12/22/2022] Open
Abstract
Background Pancreatic cancer, including cancer of the ampulla of Vater and bile duct, is very aggressive and has a poor five year survival rate; improved methods of patient stratification are required. Methods We assessed the expression of calpain-1, calpain-2 and calpastatin in two patient cohorts using immunohistochemistry on tissue microarrays. The first cohort was composed of 68 pancreatic adenocarcinomas and the second cohort was composed of 120 cancers of the bile duct and ampulla. Results In bile duct and ampullary carcinomas an association was observed between cytoplasmic calpastatin expression and patient age (P = 0.036), and between nuclear calpastatin expression and increased tumour stage (P = 0.026) and the presence of vascular invasion (P = 0.043). In pancreatic cancer, high calpain-2 expression was significantly associated with improved overall survival (P = 0.036), which remained significant in multivariate Cox-regression analysis (hazard ratio = 0.342; 95% confidence interva l = 0.157-0.741; P = 0.007). In cancers of the bile duct and ampulla, low cytoplasmic expression of calpastatin was significantly associated with poor overall survival (P = 0.012), which remained significant in multivariate Cox-regression analysis (hazard ratio = 0.595; 95% confidence interval = 0.365-0.968; P = 0.037). Conclusion The results suggest that calpain-2 and calpastatin expression is important in pancreatic cancers, influencing disease progression. The findings of this study warrant a larger follow-up study.
Collapse
Affiliation(s)
- Sarah J Storr
- Academic Oncology, University of Nottingham, School of Molecular Medical Sciences, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, UK
| | | | | | | | | | | | | |
Collapse
|
38
|
Ciocca DR, Arrigo AP, Calderwood SK. Heat shock proteins and heat shock factor 1 in carcinogenesis and tumor development: an update. Arch Toxicol 2012; 87:19-48. [PMID: 22885793 DOI: 10.1007/s00204-012-0918-z] [Citation(s) in RCA: 194] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 07/25/2012] [Indexed: 12/20/2022]
Abstract
Heat shock proteins (HSP) are a subset of the molecular chaperones, best known for their rapid and abundant induction by stress. HSP genes are activated at the transcriptional level by heat shock transcription factor 1 (HSF1). During the progression of many types of cancer, this heat shock transcriptional regulon becomes co-opted by mechanisms that are currently unclear, although evidently triggered in the emerging tumor cell. Concerted activation of HSF1 and the accumulation of HSPs then participate in many of the traits that permit the malignant phenotype. Thus, cancers of many histologies exhibit activated HSF1 and increased HSP levels that may help to deter tumor suppression and evade therapy in the clinic. We review here the extensive work that has been carried out and is still in progress aimed at (1) understanding the oncogenic mechanisms by which HSP genes are switched on, (2) determining the roles of HSF1/HSP in malignant transformation and (3) discovering approaches to therapy based on disrupting the influence of the HSF1-controlled transcriptome in cancer.
Collapse
Affiliation(s)
- Daniel R Ciocca
- Oncology Laboratory, Institute of Experimental Medicine and Biology of Cuyo (IMBECU), Scientific and Technological Center (CCT), CONICET, 5500 Mendoza, Argentina.
| | - Andre Patrick Arrigo
- Apoptosis Cancer and Development, Cancer Research Center of Lyon (CRCL), UMR INSERM 1052-CNRS 5286, Claude Bernard University, Lyon-1, Cheney A Building, Centre Regional Léon Bérard, 28, rue Laennec 69008 LYON, France. ;
| | - Stuart K Calderwood
- Molecular and Cellular Radiation Oncology, Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, MA02215
| |
Collapse
|