1
|
Dawson LA, Winter KA, Knox JJ, Zhu AX, Krishnan S, Guha C, Kachnic LA, Gillin MT, Hong TS, Craig TD, Williams TM, Hosni A, Chen E, Noonan AM, Koay EJ, Sinha R, Lock MI, Ohri N, Dorth JA, Delouya G, Swaminath A, Moughan J, Crane CH. Stereotactic Body Radiotherapy vs Sorafenib Alone in Hepatocellular Carcinoma: The NRG Oncology/RTOG 1112 Phase 3 Randomized Clinical Trial. JAMA Oncol 2025; 11:136-144. [PMID: 39699905 PMCID: PMC11843352 DOI: 10.1001/jamaoncol.2024.5403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/12/2024] [Indexed: 12/20/2024]
Abstract
Importance Most patients with locally advanced hepatocellular carcinoma (HCC) recur within the liver following systemic therapy. Objective To determine whether stereotactic body radiation therapy (SBRT) improves outcomes in patients with locally advanced HCC compared with sorafenib alone. Design, Setting, and Participants This multicenter phase 3 randomized clinical trial randomized patients with HCC 1:1 to sorafenib or SBRT followed by sorafenib, stratified by performance status, liver function, degree of metastases, and macrovascular invasion. Eligible patients had HCC unsuitable for or refractory to standard local-regional therapies and were candidates for first-line systemic therapy. Data were collected from April 2013 to March 2021, and data were analyzed from July 2022 to August 2023. Intervention Personalized SBRT, 27.5 to 50 Gy in 5 fractions. Main Outcomes and Measures The primary end point was overall survival (OS). Secondary end points were progression-free survival (PFS), adverse events, and quality of life. Results Of 193 patients randomized, 177 were eligible. Accrual was stopped early due to a change in standard-of-care systemic therapy. Of 177 included patients, 150 (84.7%) were male, and the median (IQR) age was 66 (60-72) years. Macrovascular invasion was seen in 131 (74.0%). As of July 1, 2022, the median OS was 12.3 months (90% CI, 10.6-14.3) with sorafenib vs 15.8 months (90% CI, 11.4-19.2) following SBRT and sorafenib (hazard ratio [HR], 0.77; 90% CI, 0.59-1.01; 1-sided P = .06). Adjusting for stratification factors, OS was improved with SBRT (HR, 0.72; 95% CI, 0.52-0.99; 2-sided P = .04). Median PFS was improved from 5.5 months (95% CI, 3.4-6.3) with sorafenib to 9.2 months (95% CI, 7.5-11.9) with SBRT and sorafenib (HR, 0.55; 95% CI, 0.40-0.75; 2-sided P < .001). Treatment-related grade 3 or higher adverse events were seen in 37 of 88 (42%) and 39 of 83 (47%) of patients treated with sorafenib vs SBRT and sorafenib, respectively (P = .52). There were 2 treatment-related deaths in the sorafenib group (death not otherwise specified and liver failure) and 1 in the SBRT and sorafenib group (lung infection). At 6 months, improved quality of life was seen in 2 of 20 (10%) and 6 of 17 (35%) of patients treated with sorafenib and SBRT and sorafenib, respectively. Conclusions and Relevance In this phase 3 randomized clinical trial, among patients with locally advanced HCC, SBRT was associated with a clinically important but not statistically significant improved overall survival compared with sorafenib alone. Trial Registration ClinicalTrials.gov Identifier: NCT01730937.
Collapse
Affiliation(s)
- Laura A. Dawson
- University Health Network–Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Kathryn A. Winter
- NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania
- American College of Radiology, Philadelphia, Pennsylvania
| | - Jennifer J. Knox
- University Health Network–Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Andrew X. Zhu
- Massachusetts General Hospital Cancer Center, Boston
| | | | | | - Lisa A. Kachnic
- NYP/Columbia University Medical Center/Herbert Irving Comprehensive Cancer Center, New York, New York
| | | | | | - Timothy D. Craig
- University Health Network–Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | | | - Ali Hosni
- University Health Network–Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Eric Chen
- University Health Network–Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Anne M. Noonan
- Ohio State University Comprehensive Cancer Center, Columbus
| | | | - Rishi Sinha
- Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | | | - Nitin Ohri
- Montefiore Medical Center, Bronx, New York
| | | | - Guila Delouya
- CHUM-Centre Hospitalier de l’Universite de Montreal, Montreal, Quebec, Canada
| | - Anand Swaminath
- Juravinski Cancer Centre at Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Jennifer Moughan
- NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania
- American College of Radiology, Philadelphia, Pennsylvania
| | | |
Collapse
|
2
|
Mehanna MG, El-Halawany AM, Al-Abd AM, Alqurashi MM, Bukhari HA, Kazmi I, Al-Qahtani SD, Bawadood AS, Anwar F, Al-Abbasi FA. 6-Shogaol improves sorafenib efficacy in colorectal cancer cells by modulating its cellular accumulation and metabolism. Pathol Res Pract 2024; 262:155520. [PMID: 39217771 DOI: 10.1016/j.prp.2024.155520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024]
Abstract
Carcinoma of the colon and rectum, also known as colorectal cancer, ranks as the third most frequently diagnosed malignancy globally. Sorafenib exhibits broad-spectrum antitumor activity against Raf, VEGF, and PDGF pathways in hepatocellular, thyroid, and renal cancers, but faces resistance in colorectal malignancies. 6-Shogaol, a prominent natural compound found in Zingiberaceae, exhibits antioxidant, anti-inflammatory, anticancer, and antiemetic properties. We investigated the influence of 6-shogaol on sorafenib's cytotoxic profile against colorectal cancer cell lines (HT-29, HCT-116, CaCo-2, and LS174T) through its effects on cellular accumulation and metabolism. Cytotoxicity was assessed using the sulpharodamine B assay, caspase-3 and c-PARP cleavage, cell cycle distribution analysis, and P-gp efflux activity. 6-Shogoal showed considerable cytotoxicity with decreased IC50 in colorectal cancer cell lines. Combining sorafenib and 6-shogaol increased c-PARP and pro-caspase-3 concentrations in HCT-116 cells compared to sorafenib alone. In combination, pro-caspase-3 concentrations were decreased in CaCo-2 cells compared to alone. Sorafenib combinations with 6-shogaol showed a significant drop in cell cycle distribution from 16.96±1.10 % to 9.16±1.85 %, respectively. At 100 µM, sorafenib and 6-shogaol showed potent and significant activity with intra-cellular rhodamine concentration on P-gp efflux activity in CRC cell lines. In conclusion, 6-shogaol substantially improved the cytotoxic profile of sorafenib by affecting its cellular uptake and metabolism. Future research should focus on dosage optimization and formulation and evaluate the efficacy and safety of the combination in animal models with colorectal cancer.
Collapse
Affiliation(s)
- Mohamed G Mehanna
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Ali M El-Halawany
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr-El-Ainy Street, Cairo 11562, Egypt.
| | - Ahmed M Al-Abd
- Pharmacology Department, Medical and Clinical Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt.
| | - May M Alqurashi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Hussam A Bukhari
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia; King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Salwa D Al-Qahtani
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia.
| | - Azizah Salim Bawadood
- Basic Medical Sciences Department, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia.
| | - Firoz Anwar
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
3
|
Anim MT, Tuffour I, Willis R, Schell M, Ostlund T, Mahnashi MH, Halaweish F, Willand-Charnley R. Deacetylated Sialic Acid Sensitizes Lung and Colon Cancers to Novel Cucurbitacin-Inspired Estrone Epidermal Growth Factor Receptor (EGFR) Inhibitor Analogs. Molecules 2023; 28:6257. [PMID: 37687086 PMCID: PMC10488366 DOI: 10.3390/molecules28176257] [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: 07/27/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Cancers utilize sugar residues such as sialic acids (Sia) to improve their ability to survive. Sia presents a variety of functional group alterations, including O-acetylation on the C6 hydroxylated tail. Previously, sialylation has been reported to suppress EGFR activation and increase cancer cell sensitivity to Tyrosine Kinase Inhibitors (TKIs). In this study, we report on the effect of deacetylated Sia on the activity of three novel EGFR-targeting Cucurbitacin-inspired estrone analogs (CIEAs), MMA 294, MMA 321, and MMA 320, in lung and colon cancer cells. Acetylation was modulated by the removal of Sialate O-Acetyltransferase, also known as CAS1 Domain-containing protein (CASD1) gene via CRISPR-Cas9 gene editing. Using a variety of cell-based approaches including MTT cell viability assay, flow cytometry, immunofluorescence assay and in-cell ELISA we observed that deacetylated Sia-expressing knockout cells (1.24-6.49 μM) were highly sensitive to all CIEAs compared with the control cells (8.82-20.97 μM). Apoptosis and varied stage cell cycle arrest (G0/G1 and G2/M) were elucidated as mechanistic modes of action of the CIEAs. Further studies implicated overexpression of CIEAs' cognate protein target, phosphorylated EGFR, in the chemosensitivity of the deacetylated Sia-expressing knockout cells. This observation correlated with significantly decreased levels of key downstream proteins (phosphorylated ERK and mTOR) of the EGFR pathway in knockout cells compared with controls when treated with CIEAs. Collectively, our findings indicate that Sia deacetylation renders lung and colon cancer cells susceptible to EGFR therapeutics and provide insights for future therapeutic interventions.
Collapse
Affiliation(s)
- Mathias T. Anim
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| | - Isaac Tuffour
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| | - Rylan Willis
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| | - Matthew Schell
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| | - Trevor Ostlund
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| | - Mater H. Mahnashi
- Department of Pharmaceutical Chemistry, Najran University, Najran P.O. Box 1988, Saudi Arabia;
| | - Fathi Halaweish
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| | - Rachel Willand-Charnley
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| |
Collapse
|
4
|
Li W, Wang M, Ma W, Liu P, Zhang M, He J, Cui Y. Temozolomide protects against the progression of glioblastoma via SOX4 downregulation by inhibiting the LINC00470-mediated transcription factor EGR2. CNS Neurosci Ther 2023; 29:2292-2307. [PMID: 36987665 PMCID: PMC10352878 DOI: 10.1111/cns.14181] [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: 08/11/2022] [Revised: 02/17/2023] [Accepted: 03/05/2023] [Indexed: 03/30/2023] Open
Abstract
OBJECTIVE Temozolomide is extensively applied in chemotherapy for glioblastoma with unclear exact action mechanisms. This article seeks to address the potential molecular mechanisms in temozolomide therapy for glioblastoma involving LINC00470. METHODS Bioinformatics analysis was conducted to predict the potential mechanism of LINC00470 in glioblastoma, which was validated by dual-luciferase reporter, RIP, ChIP, and RNA pull-down assays. LINC00470 expression and the predicted downstream transcription factor early growth response 2 (EGR2) were detected in the collected brain tissues from glioblastoma patients. Following temozolomide treatment and/or gain- and loss-of-function approaches in glioblastoma cells, cell viability, invasion, migration, cycle distribution, angiogenesis, autophagy, and apoptosis were measured. In addition, the expression of mesenchymal surface marker proteins was assessed by western blot. Tumor xenograft in nude mice was conducted for in vivo validation. RESULTS Mechanistic analysis and bioinformatics analysis revealed that LINC00470 transcriptionally activated SRY-related high-mobility-group box 4 (SOX4) through the transcription factor EGR2. LINC00470 and EGR2 were highly expressed in brain tissues of glioblastoma patients. LINC00470 and EGR2 mRNA expression gradually decreased with increasing concentrations of temozolomide in glioblastoma cells, and SOX4 expression was reduced in cells by temozolomide and LINC00470 knockdown. Temozolomide treatment induced cell cycle arrest, diminished cell viability, migration, invasion, and angiogenesis, and increased apoptosis and autophagy in glioblastoma, which was counteracted by overexpressing LINC00470 or SOX4 but was further promoted by LINC00470 knockdown. Temozolomide restrained glioblastoma growth and angiogenesis in vivo, while LINC00470 or SOX4 overexpression nullified but LINC00470 knockdown further facilitated these trends. CONCLUSION Conclusively, temozolomide repressed glioblastoma progression by repressing the LINC00470/EGR2/SOX4 axis.
Collapse
Affiliation(s)
- Wenyang Li
- Department of NeurosurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Ming Wang
- Department of NeurosurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Wenjia Ma
- Department of NeurosurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Ping Liu
- Department of OncologyThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Mingming Zhang
- Department of NeurosurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Jiarong He
- Department of NeurosurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Yan Cui
- Department of NeurosurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| |
Collapse
|
5
|
Bahman A, Abaza MS, Khoushaish S, Al-Attiyah RJ. Therapeutic efficacy of sorafenib and plant-derived phytochemicals in human colorectal cancer cells. BMC Complement Med Ther 2023; 23:210. [PMID: 37365571 DOI: 10.1186/s12906-023-04032-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 06/09/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND The present study aimed to investigate the sequence-dependent anticancer effects of combined treatment with sorafenib (Sora), a Food and Drug Administration-approved multikinase inhibitor drug, and plant-derived phytochemicals (PPCs) on human colorectal cancer (CRC) cell growth, and proteins associated with the control of cell cycle and apoptosis. METHODS The cytotoxic effects of 14 PPCs on CRL1554 fibroblast cells were determined using an MTT assay. Moreover, the cytotoxicity of Sora, PPCs, and a combination of both on CRC cells were also investigated. Cell cycle analysis was performed using flow cytometry, and cell apoptosis was investigated using DNA fragmentation, Annexin V/propidium iodide double staining, and mitochondrial membrane potential analyses. The cell cycle- and apoptosis-associated protein expression levels were analysed using western blotting. RESULTS Based on their low levels of cytotoxicity in CRL1554 cells at ≤ 20%, curcumin, quercetin, kaempferol, and resveratrol were selected for use in subsequent experiments. The combined treatment of sora and PPCs caused levels of CRC cytotoxicity in a dose-, cell type-, and schedule-dependent manner. Moreover, the combined treatment of CRC cells arrested cell growth at the S and G2/M phases, induced apoptotic cell death, caused extensive mitochondrial membrane damage, and altered the expression of the cell cycle and apoptotic proteins. CONCLUSIONS Results of the present study highlighted a difference in the level of sora efficacy in CRC cells when combined with PPCs. Further in vivo and clinical studies using the combined treatment of sora and PPCs are required to determine their potential as a novel therapeutic strategy for CRCs.
Collapse
Affiliation(s)
- Abdulmajeed Bahman
- Department of Biological Sciences, Molecular Biology Program, Faculty of Science, Kuwait University, P.O. Box 5969, 13060, Safat, Kuwait
| | - Mohamed-Salah Abaza
- Department of Biological Sciences, Molecular Biology Program, Faculty of Science, Kuwait University, P.O. Box 5969, 13060, Safat, Kuwait.
| | - Sarah Khoushaish
- Department of Biological Sciences, Molecular Biology Program, Faculty of Science, Kuwait University, P.O. Box 5969, 13060, Safat, Kuwait
| | - Rajaa J Al-Attiyah
- Department of Microbiology and Immunology, Faculty of Medicine, Kuwait University, P.O. Box 24923, 13110, Safat, Kuwait
| |
Collapse
|
6
|
Williams KS, Secomb TW, El-Kareh AW. An autonomous mathematical model for the mammalian cell cycle. J Theor Biol 2023; 569:111533. [PMID: 37196820 DOI: 10.1016/j.jtbi.2023.111533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 04/04/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023]
Abstract
A mathematical model for the mammalian cell cycle is developed as a system of 13 coupled nonlinear ordinary differential equations. The variables and interactions included in the model are based on detailed consideration of available experimental data. A novel feature of the model is inclusion of cycle tasks such as origin licensing and initiation, nuclear envelope breakdown and kinetochore attachment, and their interactions with controllers (molecular complexes involved in cycle control). Other key features are that the model is autonomous, except for a dependence on external growth factors; the variables are continuous in time, without instantaneous resets at phase boundaries; mechanisms to prevent rereplication are included; and cycle progression is independent of cell size. Eight variables represent cell cycle controllers: the Cyclin D1-Cdk4/6 complex, APCCdh1, SCFβTrCP, Cdc25A, MPF, NuMA, the securin-separase complex, and separase. Five variables represent task completion, with four for the status of origins and one for kinetochore attachment. The model predicts distinct behaviors corresponding to the main phases of the cell cycle, showing that the principal features of the mammalian cell cycle, including restriction point behavior, can be accounted for in a quantitative mechanistic way based on known interactions among cycle controllers and their coupling to tasks. The model is robust to parameter changes, in that cycling is maintained over at least a five-fold range of each parameter when varied individually. The model is suitable for exploring how extracellular factors affect cell cycle progression, including responses to metabolic conditions and to anti-cancer therapies.
Collapse
Affiliation(s)
| | - Timothy W Secomb
- BIO5 Institute, University of Arizona, Tucson, AZ, USA; Department of Physiology, University of Arizona, Tucson, AZ, USA
| | | |
Collapse
|
7
|
Inhibition of USP1 activates ER stress through Ubi-protein aggregation to induce autophagy and apoptosis in HCC. Cell Death Dis 2022; 13:951. [PMID: 36357365 PMCID: PMC9649627 DOI: 10.1038/s41419-022-05341-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 10/07/2022] [Accepted: 10/12/2022] [Indexed: 11/12/2022]
Abstract
The deubiquitinating enzyme USP1 (ubiquitin-specific protease 1) plays a role in the progression of various tumors, emerging as a potential therapeutic target. This study aimed to determine the role of USP1 as a therapeutic target in hepatocellular carcinoma (HCC). We detected USP1 expression in the tumor and adjacent tissues of patients with HCC using immunohistochemical staining. We evaluated the effect of the USP1 inhibitor ML-323 on HCC cell proliferation and cell cycle using a CCK-8 cell-counting kit and plate cloning assays, and propidium iodide, respectively. Apoptosis was detected by annexin V-FITC/Propidium Iodide (PI) staining and caspase 3 (casp3) activity. Transmission electron microscopy and LC3B immunofluorescence were used to detect autophagy. Western blotting was used to detect the accumulation of ubiquitinated proteins, the expression of endoplasmic reticulum (ER) stress-related proteins, and the AMPK-ULK1/ATG13 signaling pathway. We demonstrated that ML-323 inhibits the growth of HCC cells and induces G1 phase cell cycle arrest by regulating cyclin expression. ML-323 treatment resulted in the accumulation of ubiquitinated proteins, induced ER stress, and triggered Noxa-dependent apoptosis, which was regulated by the Activating Transcription Factor 4(ATF4). Moreover, active ER stress induces protective autophagy by increasing AMPK phosphorylation; therefore, we inhibited ER stress using 4-Phenylbutyric acid (4-PBA), which resulted in ER stress reduction, apoptosis, and autophagy in ML-323-treated HCC cells. In addition, blocking autophagy using the AMPK inhibitor compound C (CC), chloroquine (CQ), or bafilomycin A1 (BafA1) enhanced the cytotoxic effect of ML-323. Our findings revealed that targeting USP1 may be a potential strategy for the treatment of HCC.
Collapse
|
8
|
Abdu S, Juaid N, Amin A, Moulay M, Miled N. Therapeutic Effects of Crocin Alone or in Combination with Sorafenib against Hepatocellular Carcinoma: In Vivo & In Vitro Insights. Antioxidants (Basel) 2022; 11:antiox11091645. [PMID: 36139719 PMCID: PMC9495549 DOI: 10.3390/antiox11091645] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/13/2022] [Accepted: 08/24/2022] [Indexed: 12/24/2022] Open
Abstract
This study investigated the therapeutic effects of the phytochemical crocin alone or in combination with sorafenib both in rats chemically induced with hepatocellular carcinoma (HCC) and in human liver cancer cell line (HepG2). Male rats were randomly divided into five groups, namely, control group, HCC induced group, and groups treated with sorafenib, crocin or both crocin and sorafenib. HCC was induced in rats with a single intraperitoneal injection of diethylnitrosamine (DEN), then 2-acetylaminofluorene (2-AAF). The HCC-induced rats showed a significant decrease in body weight compared to animals treated with either or both examined drugs. Serum inflammatory markers (C-reactive protein (CRP); interleukin-6 (IL-6); lactate dehydrogenase (LDH), and oxidative stress markers were significantly increased in the HCC group and were restored upon treatment with either or both of therapeutic molecules. Morphologically, the HCC-induced rats manifested most histopathological features of liver cancer. Treatment with either or both of crocin and sorafenib successfully restored normal liver architecture. The expression of key genes involved in carcinogenesis (TNFα, p53, VEGF and NF-κB) was highly augmented upon HCC induction and was attenuated post-treatment with either or both examined drugs. Treatment with both crocin and sorafenib improved the histopathological and inflammation parameters as compared to single treatments. The in vivo anti-cancer effects of crocin and/or sorafenib were supported by their respective cytotoxicity on HepG2 cells. Crocin and sorafenib displayed an anti-tumor synergetic effect on HepG2 cells. The present findings demonstrated that a treatment regimen with crocin and sorafenib reduced liver toxicity, impeded HCC development, and improved the liver functions.
Collapse
Affiliation(s)
- Suzan Abdu
- Department of Biological Sciences, University of Jeddah, Jeddah 23445, Saudi Arabia
| | - Nouf Juaid
- Department of Biological Sciences, University of Jeddah, Jeddah 23445, Saudi Arabia
- Correspondence: (N.J.); (N.M.)
| | - Amr Amin
- Biology Department, UAE University, Al Ain 15551, United Arab Emirates
- The College, The University of Chicago, Chicago, IL 60637, USA
| | - Mohamed Moulay
- Embryonic Stem Cell Research Unit, King Fahd Medical Research Center, King Abdul Aziz University, Jeddah 22252, Saudi Arabia
| | - Nabil Miled
- Department of Biological Sciences, University of Jeddah, Jeddah 23445, Saudi Arabia
- Functional Genomics and Plant Physiology Research Unit, Higher Institute of Biotechnology Sfax, University of Sfax, BP261 Road Soukra Km4, Sfax 3038, Tunisia
- Correspondence: (N.J.); (N.M.)
| |
Collapse
|
9
|
Tsai TH, Chen YJ, Wang LY, Hsieh CH. Impact of Local Liver Irradiation Concurrent Versus Sequential with Lenvatinib on Pharmacokinetics and Biodistribution. Cancers (Basel) 2021; 13:cancers13071598. [PMID: 33808407 PMCID: PMC8037784 DOI: 10.3390/cancers13071598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Lenvatinib is a systemic treatment for patients with advanced hepatocellular carcinoma (HCC). Stereotactic body radiation therapy (SBRT) is an advanced technique of hypofractionated external beam radiotherapy (EBRT) that can be applied in patients with HCC. The current study showed that the area under the concentration–time curve of lenvatinib concentration (AUClenvatinib) increased by 148.8% with radiotherapy (RT)2Gy×3f’x (EBRT for the whole liver), and 68.9% with RT9Gy×3f’× (SBRT targeting a 1.5 × 1.5 cm region in the center of the liver) in the sequential regimen compared to the concurrent regimen in rats. Additionally, the AUClenvatinib was decreased by 50% in the concurrent regimen of both RT techniques with lenvatinib compared to the control group. The biodistribution of lenvatinib in the organs at risk was markedly decreased in the concurrent regimens. The radiation–drug interactions were between lenvatinib and RT, and showed sequential preferably. Abstract Concurrent and sequential regimens involving radiotherapy (RT) and lenvatinib were designed with off-target or stereotactic body radiation therapy (SBRT) doses in a freely moving rat model to evaluate the effect of RT on the pharmacokinetics (PK) of lenvatinib. Liver RT concurrent with lenvatinib decreased the area under the concentration–time curve of lenvatinib concentration (AUClenvatinib) by 51.1% with three fractions of 2 Gy (RT2Gy×3f’x, p = 0.03), and 48.9% with RT9Gy×3f’x (p = 0.03). The AUClenvatinib increased by 148.8% (p = 0.008) with RT2Gy×3f’x, and 68.9% (p = 0.009) with RT9Gy×3f’x in the sequential regimen compared to the concurrent regimen. There were no differences in the AUClenvatinib between RT2Gy×3f’x and RT9Gy×3f’x in the concurrent or sequential regimen. Both the RT2Gy×3f’x and RT9Gy×3f’x concurrent regimens markedly decreased the biodistribution of lenvatinib in the heart, liver, lung, spleen, and kidneys, which ranged from 31% to 100% for RT2Gy×3f’x, and 11% to 100% for RT9Gy×3f’x, compared to the sham regimen. The PK and biodistribution of lenvatinib can be modulated by simultaneous off-target irradiation and SBRT doses. The timing of lenvatinib administration with respect to RT, impacted the PK and biodistribution of the drug. Additionally, off-target and SBRT doses had a similar ability to modulate the effect of systemic therapy.
Collapse
Affiliation(s)
- Tung-Hu Tsai
- Institute of Traditional Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (T.-H.T.); (Y.-J.C.)
| | - Yu-Jen Chen
- Institute of Traditional Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (T.-H.T.); (Y.-J.C.)
- Departments of Radiation Oncology, Mackay Memorial Hospital, Taipei 104, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan
- Department of Nursing, MacKay Junior College of Medicine, Nursing and Management, Taipei 112, Taiwan
| | - Li-Ying Wang
- School and Graduate Institute of Physical Therapy, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
- Physical Therapy Center, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chen-Hsi Hsieh
- Institute of Traditional Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (T.-H.T.); (Y.-J.C.)
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
- Correspondence: or or ; Tel.: +886-2-8966-7000 (ext. 1033); Fax: +886-2-8966-0906
| |
Collapse
|
10
|
Effect of radiotherapy on survival in advanced hepatocellular carcinoma patients treated with sorafenib: a nationwide cancer-registry-based study. Sci Rep 2021; 11:1614. [PMID: 33452421 PMCID: PMC7810734 DOI: 10.1038/s41598-021-81176-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/01/2021] [Indexed: 12/18/2022] Open
Abstract
Sorafenib is the standard treatment for advanced hepatocellular carcinoma (HCC) patients. This study aims to determine whether combining radiotherapy with sorafenib administration increases its efficacy. The study cohort included 4763 patients with diagnosed advanced HCC who received sorafenib between January 2012 and December 2015, as reported in medical records in the Taiwan Cancer Registry database. The effect of sorafenib with or without radiotherapy on survival was calculated using the Kaplan–Meier method and compared using the log-rank test. A Cox proportional hazards model was used for multivariate analysis. Patients receiving sorafenib plus radiotherapy had greater 1-year survival than did those receiving sorafenib alone (P < 0.001). Uni- and multivariate analyses also showed that radiotherapy increased survival after adjusting for confounders (adjusted HR 0.57; 95% CI 0.51–0.63). Further stratified analysis according to the timing of radiotherapy relative to sorafenib treatment revealed that patients who underwent radiotherapy after sorafenib had greater 1-year survival than did those undergoing radiotherapy within sorafenib use or sorafenib alone (adjusted HR 0.39; 95% CI 0.27–0.54). Combined treatment with sorafenib and radiotherapy results in greater HCC patient survival and should be considered an option for treating this challenging disease.
Collapse
|
11
|
Deciphering Antitumor Mechanism of Pien Tze Huang in Mice of Hepatocellular Carcinoma Based on Proteomics. J Immunol Res 2020; 2020:4876251. [PMID: 33344655 PMCID: PMC7728492 DOI: 10.1155/2020/4876251] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 11/05/2020] [Accepted: 11/19/2020] [Indexed: 12/24/2022] Open
Abstract
The Chinese formula Pien Tze Huang (PZH) has been used to treat hepatocellular carcinoma (HCC) and showed positive clinical effects. However, the antitumor mechanism of PZH in HCC remains unclear. In this study, HCC xenograft Balb/c mice were treated with PZH; then, proteomics detection and Ingenuity Pathway Analysis (IPA) were used to analyze the differentiated phosphorylated proteins in tumor tissues. The results indicated that PZH could inhibit tumor weight by 50.76%. Eighty-four upregulated and 11 downregulated phosphorylated proteins were identified in PZH-treated mice. Twenty signaling pathways were associated with inflammation (including the IL-6 and TNFR1/2 pathways), cancer growth (including the p53 and FAK pathways), and the cell cycle (including the G2/M and G1/S checkpoint regulation pathways). Moreover, TNF-α, IL-6, and several typical differentially expressed phosphorylated proteins (such as p-CCNB1, p-FOXO3, and p-STAT3) in tumor tissues, tumor cell viability, and cell cycle arrest assay in vitro further verify the results of IPA. These results revealed that PZH achieved antitumor activity in HCC; the underlying mechanisms of which were mainly through regulating the inflammation-associated cytokine secretion, cancer growth pathways, and induction of G2/M arrest. These data provided the potential molecular basis for PZH to act as a therapeutic drug or a supplement to chemotherapy drugs for human HCC in the future.
Collapse
|
12
|
Goh MJ, Kang W, Sinn DH, Gwak GY, Paik YH, Choi MS, Lee JH, Koh KC, Paik SW. Advanced Stage Hepatocellular Carcinoma Successfully Treated with Transarterial Radioembolization and Multi-tyrosine Kinase Inhibitor Therapy. JOURNAL OF LIVER CANCER 2020; 20:160-166. [PMID: 37384324 PMCID: PMC10035672 DOI: 10.17998/jlc.20.2.160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/01/2020] [Accepted: 03/01/2020] [Indexed: 06/30/2023]
Abstract
Transarterial radioembolization (TARE) with yttrium-90 microspheres has become widely utilized in managing hepatocellular carcinoma (HCC). The utility of TARE is expanding with new insights through experiences from real-world practice and clinical trials, and recently published data suggest that TARE in combination with sorafenib may improve the overall survival in selected patients. Here, we report a case of advanced stage HCC that was successfully treated with TARE and sorafenib. The patient achieved complete response (CR) at 12 months after the initial treatment with TARE and sorafenib, followed by additional transarterial chemoembolization and proton beam therapy for local tumor recurrence at 19-month post-TARE. The patient was followed up every 3 months thereafter and still achieved CR both biochemically and radiologically for the following 12 months. A combination strategy of TARE and systemic therapy may be a useful alternative treatment option for selected patients with advanced stage HCC.
Collapse
Affiliation(s)
- Myung Ji Goh
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Wonseok Kang
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong Hyun Sinn
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Geum-Youn Gwak
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yong-Han Paik
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Moon Seok Choi
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joon Hyeok Lee
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kwang Cheol Koh
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seung Woon Paik
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Murakami Y, Kenjo M, Ishikawa K, Sakayauchi T, Itasaka S, Negoro Y, Jingu K, Nishimura Y, Nagata Y, Ogawa K. Risk factors for severe gastrointestinal toxicity in patients receiving palliative radiotherapy for metastatic bone tumors: association with the use of molecular-targeted agents. JOURNAL OF RADIATION RESEARCH 2020; 61:629-634. [PMID: 32567666 PMCID: PMC7336816 DOI: 10.1093/jrr/rraa035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/18/2020] [Indexed: 06/11/2023]
Abstract
This study aimed to investigate whether the use of molecular-targeted agents could affect gastrointestinal (GI) toxicity in palliative radiotherapy (RT) for metastatic bone tumors in the abdominopelvic region. We collected data of patients who received palliative RT for bone metastases in the abdominopelvic region between 2013 and 2014 from six institutions. Data of 395 patients were collected and184 patients received molecularly targeted therapy, of whom 80 received vascular endothelial growth factor (VEGF)-targeted agents. For 556 lesions, 410 sessions of irradiation were undergone. GI toxicity of ≥G3 was observed in 3.8% of patients. The incidence rates of ≥G3 GI toxicity in patients without targeted agents use, in those using VEGF-targeted agents and in those using non-VEGF-targeted agents were 3.8, 7.5 and 1.0%, respectively. Regarding risk factors of the occurrence of ≥G3 GI toxicity, univariate analysis in all patients showed that a history of abdominopelvic surgery was a significant risk factor (P = 0.01), and the use of VEGF-targeted agents showed a trend of high incidence (P = 0.06). In patients using VEGF-targeted agents, both univariate and multivariate analysis showed that combined anticoagulant use (P = 0.03 and 0.01) and agent use between 1 week before and after RT (P = 0.046 and 0.03) were significant risk factors. In conclusion, the history of abdominopelvic surgery was associated with ≥G3 GI toxicity and the use of VEGF-targeted agents showed a trend for high incidence. When using VEGF-targeted agents, caution should be exercised in the combined use of anticoagulants and in the agent use between 1 week before and after RT.
Collapse
Affiliation(s)
- Yuji Murakami
- Department of Radiation Oncology, Hiroshima University Hospital, Hiroshima, Japan
| | - Masahiro Kenjo
- Hiroshima High-Precision Radiotherapy Cancer Center, Hiroshima, Japan
| | - Kazuki Ishikawa
- Department of Radiation Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Toru Sakayauchi
- Department of Radiation Oncology, Osaki Citizen Hospital, Osaki, Japan
| | - Satoshi Itasaka
- Department of Radiation Oncology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Yoshiharu Negoro
- Department of Radiation Oncology, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
| | - Keiichi Jingu
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasumasa Nishimura
- Department of Radiation Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yasushi Nagata
- Department of Radiation Oncology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kazuhiko Ogawa
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
14
|
Hsieh CH, Chen YJ, Tsai TH, Wang LY, Tai HC, Huang HL, Huang YC. Robust combination of liver stereotactic body radiotherapy modulates pharmacokinetics of sorafenib toward preferable parameters. Sci Rep 2020; 10:9575. [PMID: 32533042 PMCID: PMC7293270 DOI: 10.1038/s41598-020-66583-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 05/21/2020] [Indexed: 01/15/2023] Open
Abstract
To evaluate the effect and mechanism of radiotherapy (RT)–sorafenib pharmacokinetics (PK) in different regimens with conventional or high dose irradiation. Between February 2012 and December 2018, 43 patients with portal vein tumor thrombosis treated with sorafenib plus conventional RT (58%) or stereotactic body radiation therapy (SBRT, 42%) were retrospectively reviewed. In vivo and in vitro studies of concurrent and sequential RT with sorafenib were designed. SBRT resulted in a 3-fold increase in complete recanalization compared to conventional RT group (28% vs. 8%, p = 0.014). Compared to the control group, the area under the concentration vs. time curve (AUC) of sorafenib was increased in the concurrent RT2Gy and RT9Gy groups and the sequential RT9Gy group by 132% (p = 0.046), 163% (p = 0.038) and 102% (p = 0.018), respectively; and was decreased by 59% in the sequential RT2Gy group (p = 0.036). Sequential RT2Gy and RT9Gy increased CYP3A4 activity by 82% (p = 0.028) and 203% (p = 0.0004), respectively, compared to that with the corresponding concurrent regimen. SBRT produced better recanalization than conventional RT with sorafenib. The AUC of sorafenib was modulated by RT. P-gp expression was not influenced by RT. The sequential RT regimen increased CYP3A4 activity that may increase the RT-sorafenib synergy effect and overall sorafenib activity. The biodistribution of sorafenib was modulated by local RT with the different regimens.
Collapse
Affiliation(s)
- Chen-Hsi Hsieh
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan. .,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan. .,Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, Taipei, Taiwan.
| | - Yu-Jen Chen
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Radiation Oncology, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| | - Tung-Hu Tsai
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Chemical Engineering, National United University, Miaoli, Taiwan
| | - Li-Ying Wang
- School and Graduate Institute of Physical Therapy, College of Medicine, National Taiwan University, Taipei, Taiwan.,Physical Therapy Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Hung-Chi Tai
- Department of Radiation Oncology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Hsiang-Ling Huang
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, Taipei, Taiwan
| | - Yu-Chuen Huang
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.,School of Chinese Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
15
|
Jakobi K, Beyer S, Koch A, Thomas D, Schwalm S, Zeuzem S, Pfeilschifter J, Grammatikos G. Sorafenib Treatment and Modulation of the Sphingolipid Pathway Affect Proliferation and Viability of Hepatocellular Carcinoma In Vitro. Int J Mol Sci 2020; 21:ijms21072409. [PMID: 32244391 PMCID: PMC7177910 DOI: 10.3390/ijms21072409] [Citation(s) in RCA: 9] [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: 03/07/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) shows a remarkable heterogeneity and is recognized as a chemoresistant tumor with dismal prognosis. In previous studies, we observed significant alterations in the serum sphingolipids of patients with HCC. This study aimed to investigate the in vitro effects of sorafenib, which is the most widely used systemic HCC medication, on the sphingolipid pathway as well as the effects of inhibiting the sphingolipid pathway in HCC. Huh7.5 and HepG2 cells were stimulated with sorafenib, and inhibitors of the sphingolipid pathway and cell proliferation, viability, and concentrations of bioactive metabolites were assessed. We observed a significant downregulation of cell proliferation and viability and a simultaneous upregulation of dihydroceramides upon sorafenib stimulation. Interestingly, fumonisin B1 (FB1) and the general sphingosine kinase inhibitor SKI II were able to inhibit cell proliferation more prominently in HepG2 and Huh7.5 cells, whereas there were no consistent effects on the formation of dihydroceramides, thus implying an involvement of distinct metabolic pathways. In conclusion, our study demonstrates a significant downregulation of HCC proliferation upon sorafenib, FB1, and SKI II treatment, whereas it seems they exert antiproliferative effects independently from sphingolipids. Certainly, further data would be required to elucidate the potential of FB1 and SKI II as putative novel therapeutic targets in HCC.
Collapse
Affiliation(s)
- Katja Jakobi
- Medizinische Klinik 1, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (K.J.); (S.Z.)
- Institut für Allgemeine Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (S.B.); (A.K.); (S.S.); (J.P.)
| | - Sandra Beyer
- Institut für Allgemeine Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (S.B.); (A.K.); (S.S.); (J.P.)
| | - Alexander Koch
- Institut für Allgemeine Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (S.B.); (A.K.); (S.S.); (J.P.)
| | - Dominique Thomas
- Institut für Klinische Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany;
| | - Stephanie Schwalm
- Institut für Allgemeine Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (S.B.); (A.K.); (S.S.); (J.P.)
| | - Stefan Zeuzem
- Medizinische Klinik 1, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (K.J.); (S.Z.)
| | - Josef Pfeilschifter
- Institut für Allgemeine Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (S.B.); (A.K.); (S.S.); (J.P.)
| | - Georgios Grammatikos
- Medizinische Klinik 1, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (K.J.); (S.Z.)
- Institut für Allgemeine Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (S.B.); (A.K.); (S.S.); (J.P.)
- St Luke’s Hospital, 55236 Thessaloniki, Greece
- Correspondence: ; Tel.: +30-2316-014-910
| |
Collapse
|
16
|
Varshosaz J, Sadri F, Rostami M, Mirian M, Taymouri S. Synthesis of pectin-deoxycholic acid conjugate for targeted delivery of anticancer drugs in hepatocellular carcinoma. Int J Biol Macromol 2019; 139:665-677. [DOI: 10.1016/j.ijbiomac.2019.07.225] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 12/22/2022]
|
17
|
Xu J, Huang F, Yao Z, Jia C, Xiong Z, Liang H, Lin N, Deng M. Inhibition of cyclin E1 sensitizes hepatocellular carcinoma cells to regorafenib by mcl-1 suppression. Cell Commun Signal 2019; 17:85. [PMID: 31349793 PMCID: PMC6660968 DOI: 10.1186/s12964-019-0398-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/15/2019] [Indexed: 12/22/2022] Open
Abstract
Background To clarify the effects of cylcin E1 expression on HCC tumor progression, we studied the expression of cyclin E1 and inhibitory efficacy of regorafenib and sorafenib in HCC cells, and investigated a potential therapy that combines regorafenib treatment with cyclin E1 inhibition. Methods Western blotting for caspase-3 and Hoechst 33225 staining was used to measure the expression level of apoptosis-related proteins under drug treatment. Results Our results showed that enhanced expression of cyclin E1 after transfection compromised apoptosis in HCC cells induced by regorafenib or sorafenib. Conversely, down-regulation of cyclin E1 gene expression or inhibition of cyclin E1 by the cyclin-dependent kinase (CDK) inhibitors dinaciclib (DIN) or flavopiridol sensitized HCC cells to regorafenib and sorafenib by inducing apoptosis. The expression of Mcl-1, which is modulated by STAT3, plays a key role in regulating the therapeutic effects of CDK inhibitors. Xenograft experiments conducted to test the efficacy of regorafenib combined with DIN showed dramatic tumor inhibitory effects due to induction of apoptosis. Our results suggested that the level of cyclin E1 expression in HCCs may be used as a pharmacodynamic biomarker to assess the antitumor effects of regorafenib or sorafenib. Conclusions Combining regorafenib and CDK inhibitors may enhance the clinical efficiency of the treatment of HCCs. Electronic supplementary material The online version of this article (10.1186/s12964-019-0398-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jianliang Xu
- Hepatobilliary Surgery Department, The Third affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe District, Guangzhou, Guangdong, China
| | - Fei Huang
- Anesthesiology Department, The third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhicheng Yao
- General surgery, The Third affiliated hospital of Sun Yat-sen University, No. 600, Tianhe District, Guangzhou, 510630, Guangdong, China.
| | - Changchang Jia
- Cell & Gene therapy center, The Third affiliated Hospital of Sun Yat-sen Uuniversity, Guangzhou, Guangdong, China
| | - Zhiyong Xiong
- General surgery, The Third affiliated hospital of Sun Yat-sen University, No. 600, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Hao Liang
- General surgery, The Third affiliated hospital of Sun Yat-sen University, No. 600, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Nan Lin
- Hepatobilliary Surgery Department, The Third affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe District, Guangzhou, Guangdong, China
| | - Meihai Deng
- Hepatobilliary Surgery Department, The Third affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe District, Guangzhou, Guangdong, China.
| |
Collapse
|
18
|
Shafie F, Nabavizadeh F, Shafie Ardestani M, Panahi M, Adeli S, Samandari H, Ashabi G. Sorafenib-loaded PAMAM dendrimer attenuates liver fibrosis and its complications in bile-duct-ligated rats. Can J Physiol Pharmacol 2019; 97:691-698. [PMID: 31071278 DOI: 10.1139/cjpp-2019-0141] [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] [Indexed: 11/22/2022]
Abstract
We assessed the effect of sorafenib-loaded polyamidoamine (PAMAM) dendrimer on liver fibrosis induced by bile duct ligation (BDL). Male Wistar rats were divided into 9 groups: intact, sham, DMSO + BDL, BDL, sorafenib (30 mg/kg), sorafenib (60 mg/kg), PAMAM + BDL, sorafenib (30 mg/kg) + PAMAM + BDL, sorafenib (60 mg/kg) + PAMAM + BDL. BDL was induced and then rats were treated daily with sorafenib and (or) PAMAM for 4 weeks. Improvement of liver was detected via assessment of ascites formation, collagen deposition, liver blood flow, vascular endothelial growth factor level, and blood cells count. Sorafenib-loaded PAMAM dendrimer in both 30 and 60 mg/kg doses reduced ascites formation, reduced collagen deposition, and improved drug-induced hematological side effects of sorafenib alone in comparison with sorafenib-alone treatment. Sorafenib-loaded PAMAM dendrimer increased liver blood flow compared with sorafenib-received groups. Sorafenib-loaded PAMAM dendrimer reduced BDL-induced liver injury compared with sorafenib-received groups. Moreover, sorafenib-loaded PAMAM dendrimer decreased vascular endothelial growth factor level in serum and liver tissue in comparison with sorafenib-received groups. Sorafenib-loaded PAMAM dendrimer profoundly improved the therapeutic effects of sorafenib in BDL rats.
Collapse
Affiliation(s)
- Fatemeh Shafie
- a Department of Physiology, Medical School, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nabavizadeh
- a Department of Physiology, Medical School, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Shafie Ardestani
- b Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahshid Panahi
- c Department of Pathology, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Soheila Adeli
- d Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hedayat Samandari
- a Department of Physiology, Medical School, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghorbangol Ashabi
- a Department of Physiology, Medical School, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Anuja K, Chowdhury AR, Saha A, Roy S, Rath AK, Kar M, Banerjee B. Radiation-induced DNA damage response and resistance in colorectal cancer stem-like cells. Int J Radiat Biol 2019; 95:667-679. [PMID: 30753097 DOI: 10.1080/09553002.2019.1580401] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Purpose: Radiation therapy is an integral part of current treatment modality for colorectal cancer. Recent studies have revealed the presence of cancer stem-like cells (CSCs) population, in different tumors are responsible for therapeutic resistance and disease relapse, including colorectal cancer with poorer survival rate. Hence, characterization of the effect of Ionizing Radiation (IR) in colorectal cancer may serve to explain possible mechanisms. Material and methods: Parental HCT116 and HCT-15 cells and derived colonospheres were irradiated and dose was optimized based on cell survival assay and cell cycle analysis. DNA damage response (DDR) was elucidated by γH2AX foci formation, COMET assay, and ATM, p-ATM, ERCC1 expression post-treatment. The expression level of developmental marker (β-catenin), CSC markers (CD44, KLF4) and telomeric components (TRF2, RAP1, hTERT) were evaluated. Results: We observed cell survival was more in colonospheres post-irradiation and also exhibited decreased γH2AX foci, olive tail moment, increased ERCC1, and p-ATM expression than its parental counterpart which corresponds to efficient DDR. Differential expression of developmental marker, CSC markers, and telomeric components were observed after irradiation. Conclusion: This study highlighted the presence of CSC phenotype in colonospheres having increased DNA repair capacity. Differential expression of developmental marker, CSC markers and telomeric components between parental and colonospheres may contribute in radio-resistance property of CSCs.
Collapse
Affiliation(s)
- Kumari Anuja
- a Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University , Bhubaneswar , India
| | - Amit Roy Chowdhury
- a Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University , Bhubaneswar , India
| | - Arka Saha
- a Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University , Bhubaneswar , India
| | - Souvick Roy
- a Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University , Bhubaneswar , India
| | | | - Madhabananda Kar
- c Department of Surgical Oncology , All India Institute of Medical Sciences (AIIMS) , Bhubaneswar , India
| | - Birendranath Banerjee
- a Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University , Bhubaneswar , India
| |
Collapse
|
20
|
Angiogenesis in Pancreatic Cancer: Pre-Clinical and Clinical Studies. Cancers (Basel) 2019; 11:cancers11030381. [PMID: 30889903 PMCID: PMC6468440 DOI: 10.3390/cancers11030381] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/11/2019] [Accepted: 03/14/2019] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis is a crucial event in tumor development and progression, occurring by different mechanisms and it is driven by pro- and anti-angiogenic molecules. Pancreatic cancer vascularization is characterized by a high microvascular density, impaired microvessel integrity and poor perfused vessels with heterogeneous distribution. In this review article, after a brief introduction on pancreatic cancer classification and on angiogenesis mechanisms involved in its progression, the pre-clinical and clinical trials conducted in pancreatic cancer treatment using anti-angiogenic inhibitors will be described. Finally, we will discuss the anti-angiogenic therapy paradox between the advantage to abolish vessel supply to block tumor growth and the disadvantage due to reduction of drug delivery at the same time. The purpose is to identify new anti-angiogenic molecules that may enhance treatment regimen.
Collapse
|
21
|
Abstract
For patients with unresectable or medically inoperable hepatocellular carcinoma, there are many local and regional therapies available, including stereotactic body radiotherapy, radiofrequency ablation, and transcatheter embolic approaches. This article will describe these treatment options and review the current comparative literature, suggesting that stereotactic body radiotherapy provides similar or better tumor control and a favorable side effect profile.
Collapse
Affiliation(s)
- Yao Yu
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA
| | - Mary Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA.
| |
Collapse
|
22
|
Jo Y, Kim EH, Sai S, Kim JS, Cho JM, Kim H, Baek JH, Kim JY, Hwang SG, Yoon M. Functional Biological Activity of Sorafenib as a Tumor-Treating Field Sensitizer for Glioblastoma Therapy. Int J Mol Sci 2018; 19:E3684. [PMID: 30469352 PMCID: PMC6274791 DOI: 10.3390/ijms19113684] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/09/2018] [Accepted: 11/16/2018] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma, the most common primary brain tumor in adults, is an incurable malignancy with poor short-term survival and is typically treated with radiotherapy along with temozolomide. While the development of tumor-treating fields (TTFields), electric fields with alternating low and intermediate intensity has facilitated glioblastoma treatment, clinical outcomes of TTFields are reportedly inconsistent. However, combinatorial administration of chemotherapy with TTFields has proven effective for glioblastoma patients. Sorafenib, an anti-proliferative and apoptogenic agent, is used as first-line treatment for glioblastoma. This study aimed to investigate the effect of sorafenib on TTFields-induced anti-tumor and anti-angiogenesis responses in glioblastoma cells in vitro and in vivo. Sorafenib sensitized glioblastoma cells to TTFields, as evident from significantly decreased post-TTFields cell viability (p < 0.05), and combinatorial treatment with sorafenib and TTFields accelerated apoptosis via reactive oxygen species (ROS) generation, as evident from Poly (ADP-ribose) polymerase (PARP) cleavage. Furthermore, use of sorafenib plus TTFields increased autophagy, as evident from LC3 upregulation and autophagic vacuole formation. Cell cycle markers accumulated, and cells underwent a G2/M arrest, with an increased G0/G1 cell ratio. In addition, the combinatorial treatment significantly inhibited tumor cell motility and invasiveness, and angiogenesis. Our results suggest that combination therapy with sorafenib and TTFields is slightly better than each individual therapy and could potentially be used to treat glioblastoma in clinic, which requires further studies.
Collapse
Affiliation(s)
- Yunhui Jo
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
- Department of Bio-Convergence Engineering, Korea University, Seoul 02842, Korea.
| | - Eun Ho Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Sei Sai
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, Chiba 263-0024, Japan.
| | - Jin Su Kim
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Jae-Min Cho
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Hyeongi Kim
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Jeong-Hwa Baek
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Jeong-Yub Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Sang-Gu Hwang
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Myonggeun Yoon
- Department of Bio-Convergence Engineering, Korea University, Seoul 02842, Korea.
| |
Collapse
|
23
|
Tanriverdi O, Erdogan U, Tanik C, Yilmaz I, Gunaldi O, Adilay HU, Arslanhan A, Eseoglu M. Impact of sorafenib on epidural fibrosis: An immunohistochemical study. World J Clin Cases 2018; 6:249-258. [PMID: 30211205 PMCID: PMC6134279 DOI: 10.12998/wjcc.v6.i9.249] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/23/2018] [Accepted: 08/01/2018] [Indexed: 02/05/2023] Open
Abstract
AIM To determine if sorafenib, an antineoplastic agent, could prevent the development of spinal epidural fibrosis (EF).
METHODS The study used CD105 and osteopontin antibodies in an immunohistochemical approach to quantify EF that occurred as a consequence of laminectomy in rats. Wistar albino rats (n = 16) were divided into two groups: control (L1-2 level laminectomy only) and sorafenib treatment (L1-2 level laminectomy + topical sorafenib). The animals were euthanatized after 6 wk, and the EF tissues were examined for histopathological changes after immunohistochemical staining. The EF grades were assigned to the tissues, and the treatment and control groups were compared.
RESULTS The EF thickness, inflammatory cell density, and arachnoid adherences determined by light microscopy were significantly higher in the control group compared to the sorafenib-treated group. Based on fibrosis scores, the extent of EF in the treatment group was significantly lower than in the controls. Immunohistochemical staining for CD105 to identify microvessels revealed that the EF grades based on vessel count were significantly lower in the treatment group. Staining for osteopontin did not show any significant differences between the groups in terms of the extent of EF. The staging of EF based on vascular counts observed after immunohistochemical staining for CD105, but not for osteopontin, was compatible with conventional staging methods. Neither toxic effects on tissues nor systemic side effects were observed with the use of sorafenib.
CONCLUSION Local administration of sorafenib significantly reduced post-laminectomy EF. Decreased neovascularization in spinal tissue may be due to the sorafenib-induced inhibition of vascular endothelial growth factor.
Collapse
Affiliation(s)
- Osman Tanriverdi
- Department of Neurosurgery and Psychiatry, University of Health Sciences, Bakırky Prof. Dr. Mazhar Osman Training and Research Hospital for Neurology, İstanbul 34303, Turkey
| | - Uzay Erdogan
- Department of Neurosurgery and Psychiatry, University of Health Sciences, Bakırky Prof. Dr. Mazhar Osman Training and Research Hospital for Neurology, İstanbul 34303, Turkey
| | - Canan Tanik
- Department of Pathology, University of Health Sciences, Şişli Hamidiye Etfal Training and Research Hospital, İstanbul 34303, Turkey
| | - Ilhan Yilmaz
- Department of Neurosurgery, University of Health Sciences, Şişli Hamidiye Etfal Training and Research Hospital, İstanbul 34303, Turkey
| | - Omur Gunaldi
- Department of Neurosurgery and Psychiatry, University of Health Sciences, Bakırky Prof. Dr. Mazhar Osman Training and Research Hospital for Neurology, İstanbul 34303, Turkey
| | - Huseyin Utku Adilay
- Department of Neurosurgery, Medical Faculty, Balıkesir University, Balıkesir 31300, Turkey
| | - Ayca Arslanhan
- Institute of Neurological Science, Marmara University, İstanbul 34303, Turkey
| | - Metehan Eseoglu
- Department of Neurosurgery, Medical Faculty, Medipol University, İstanbul 34303, Turkey
| |
Collapse
|
24
|
Hosaka S, Katagiri H, Niwakawa M, Harada H, Wasa J, Murata H, Takahashi M. Radiotherapy combined with zoledronate can reduce skeletal-related events in renal cell carcinoma patients with bone metastasis. Int J Clin Oncol 2018; 23:1127-1133. [DOI: 10.1007/s10147-018-1310-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/24/2018] [Indexed: 01/26/2023]
|
25
|
Wada Y, Takami Y, Matsushima H, Tateishi M, Ryu T, Yoshitomi M, Matsumura T, Saitsu H. The Safety and Efficacy of Combination Therapy of Sorafenib and Radiotherapy for Advanced Hepatocellular Carcinoma: A Retrospective Study. Intern Med 2018; 57:1345-1353. [PMID: 29279513 PMCID: PMC5995712 DOI: 10.2169/internalmedicine.9826-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Objective Sorafenib is a standard therapy for advanced hepatocellular carcinoma (HCC), whereas radiotherapy is effective for local control of extrahepatic spread (EHS) or macrovascular invasion (MVI). This study investigated the safety and efficacy of this combined therapy to treat advanced HCC. Methods This retrospective study reviewed 62 patients with advanced-stage HCC with EHS or MVI who received sorafenib therapy, excluding the patients with only lung metastases. Results Of the 62 patients, 15 were treated using the combined therapy of sorafenib and radiotherapy (group RS), and 47 were treated with sorafenib monotherapy (group S). In group RS, patients were treated using three-dimensional conformal radiotherapy with a total irradiation dose of 30-60 Gy (median, 50 Gy). Irradiation was targeted at the bone, lymph nodes, adrenal gland, and MVI in 6, 5, 1, and 4 patients, respectively. The overall incidence of adverse events was 93.3% in group RS and 91.5% in group S (p=N.S.). Incidences of thrombocytopenia, leukopenia, and skin reaction were significantly higher in group RS (73.3%, 40.0%, and 66.7%, respectively) than in group S (36.2%, 10.6%, and 27.7%, respectively, p=0.02, 0.02, and <0.01, respectively). The incidence of severe adverse events, however, was comparable in the 2 groups: 20% in group RS and 19.2% in group S. The median progression-free survival (PFS) of EHS or MVI, PFS of whole lesions, and overall survival were longer in group RS (13.5, 10.6, and 31.2 months, respectively) than in group S (3.3, 3.5, and 12.1 months, respectively) (p<0.01 for all). Conclusion Sorafenib in combination with radiotherapy is a feasible and tolerable treatment option for advanced HCC.
Collapse
Affiliation(s)
- Yoshiyuki Wada
- Department of Hepato-Biliary-Pancreatic Surgery, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Japan
| | - Yuko Takami
- Department of Hepato-Biliary-Pancreatic Surgery, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Japan
| | - Hajime Matsushima
- Department of Hepato-Biliary-Pancreatic Surgery, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Japan
| | - Masaki Tateishi
- Department of Hepato-Biliary-Pancreatic Surgery, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Japan
| | - Tomoki Ryu
- Department of Hepato-Biliary-Pancreatic Surgery, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Japan
| | - Munehiro Yoshitomi
- Department of Hepato-Biliary-Pancreatic Surgery, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Japan
| | - Taisei Matsumura
- Department of Radiology, National Hospital Organization Kyushu Medical Center, Japan
| | - Hideki Saitsu
- Department of Hepato-Biliary-Pancreatic Surgery, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Japan
| |
Collapse
|
26
|
Möckelmann N, Rieckmann T, Busch CJ, Becker B, Gleißner L, Hoffer K, Omniczynski M, Steinmeister L, Laban S, Grénman R, Petersen C, Rothkamm K, Dikomey E, Knecht R, Kriegs M. Effect of sorafenib on cisplatin-based chemoradiation in head and neck cancer cells. Oncotarget 2018; 7:23542-51. [PMID: 27015558 PMCID: PMC5029646 DOI: 10.18632/oncotarget.8275] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 03/01/2016] [Indexed: 11/25/2022] Open
Abstract
Despite aggressive chemoradiation (CRT) protocols in the treatment of patients with head and neck squamous cell carcinomas (HNSCC), the outcome is still unfavorable. To improve therapy efficacy we had already successfully tested the multikinase inhibitor sorafenib in combination with irradiation (IR) in previous studies on HNSCC cell lines. In this study we investigated its effect on combined CRT treatment using cisplatin.Radio- and chemosensitivity with and without sorafenib was measured in four HNSCC cell lines and normal fibroblasts (NF) by colony formation assay. Apoptosis and cell cycle analysis were performed by flow cytometry. In HNSCC cells, sorafenib enhanced the antiproliferative effect of cisplatin without affecting apoptosis induction and with only minor effects on cell inactivation. Sorafenib added prior to irradiation enhanced cellular radiosensitivity in three of the tested HNSCC cell lines and caused massive overall cell inactivation when combined with CRT. In contrast, sorafenib did not radiosensitize NF and reduced cisplatin-induced cell inactivation. Cell inactivation by IR and cisplatin is further increased by the addition of sorafenib in HNSCC, but not in NF cells. Therefore, sorafenib is a promising candidate to improve therapy efficacy for HNSCC.
Collapse
Affiliation(s)
- Nikolaus Möckelmann
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Rieckmann
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Chia-Jung Busch
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Becker
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa Gleißner
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konstantin Hoffer
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maria Omniczynski
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leonhard Steinmeister
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon Laban
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Reidar Grénman
- Department of Otorhinolaryngology - Head and Neck Surgery and Department of Medical Biochemistry and Genetics, Turku University and University Hospital of Turku, Turku, Finland
| | - Cordula Petersen
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Department of Radiotherapy and Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kai Rothkamm
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ekkehard Dikomey
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Department of Radiotherapy and Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rainald Knecht
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malte Kriegs
- Head and Neck Cancer Center of The University Cancer Center Hamburg (UCCH), Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
27
|
Jeong YK, Kim MS, Lee JY, Kim EH, Kim W, Ha H, Jeong JH. Sorafenib Acts Synergistically in Combination with Radiotherapy without Causing Intestinal Damage in Colorectal Cancer. TUMORI JOURNAL 2018; 99:176-82. [DOI: 10.1177/030089161309900210] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Aims and background Colorectal cancer is one of the commonest cancers. Chemoradiotherapy gives better results than radiotherapy or chemotherapy in colorectal cancer. To enhance radiosensitivity of tumor cells for chemoradiotherapy, targeted therapy drugs that act as radiosensitizers can be used. In the present study, we provide a scientific rationale for the clinical application of sorafenib as a radiosensitizer in colorectal cancer, without causing significant adverse effects on normal intestinal tissue. Methods Three human colorectal adenocarcinoma cell lines (HCT116, HT-29, and SW480) were treated with sorafenib alone, or radiation followed by sorafenib. In vitro tests were performed using colony forming assays, cell cycle analysis, and comet assays. In addition, the effects of sorafenib and radiation therapy on the inhibition HT-29 tumor growth and survival of intestinal jejunum crypts were examined in vivo. Results Sorafenib increased the radiosensitivity of tumor cells in human colon adenocarcinoma cell lines (HCT116, HT-29, and SW480), as well as in HT-29 xenograft animal models. Sorafenib, in combination with ionizing radiation, induced the accumulation of tumor cells in the G2-M phase and delayed the repair of DNA damage caused by ionizing radiation. The combination of sorafenib and ionizing radiation did not enhance the apoptosis of intestinal crypt cells, compared with the use of radiation alone. Conclusions We provide a scientific rationale for the use of sorafenib in combination with radiotherapy in colorectal cancer.
Collapse
Affiliation(s)
- Youn Kyoung Jeong
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences
- Division of Life and Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University
| | - Mi-Sook Kim
- Department of Radiation Oncology, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Ji Young Lee
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences
| | - Eun Ho Kim
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Wonwoo Kim
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences
| | - Hunjoo Ha
- Division of Life and Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University
| | - Jae-Hoon Jeong
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences
| |
Collapse
|
28
|
Pauty J, Usuba R, Cheng IG, Hespel L, Takahashi H, Kato K, Kobayashi M, Nakajima H, Lee E, Yger F, Soncin F, Matsunaga YT. A Vascular Endothelial Growth Factor-Dependent Sprouting Angiogenesis Assay Based on an In Vitro Human Blood Vessel Model for the Study of Anti-Angiogenic Drugs. EBioMedicine 2018; 27:225-236. [PMID: 29289530 PMCID: PMC5828365 DOI: 10.1016/j.ebiom.2017.12.014] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/06/2017] [Accepted: 12/14/2017] [Indexed: 10/26/2022] Open
Abstract
Angiogenesis is the formation of new capillaries from pre-existing blood vessels and participates in proper vasculature development. In pathological conditions such as cancer, abnormal angiogenesis takes place. Angiogenesis is primarily carried out by endothelial cells, the innermost layer of blood vessels. The vascular endothelial growth factor-A (VEGF-A) and its receptor-2 (VEGFR-2) trigger most of the mechanisms activating and regulating angiogenesis, and have been the targets for the development of drugs. However, most experimental assays assessing angiogenesis rely on animal models. We report an in vitro model using a microvessel-on-a-chip. It mimics an effective endothelial sprouting angiogenesis event triggered from an initial microvessel using a single angiogenic factor, VEGF-A. The angiogenic sprouting in this model is depends on the Notch signaling, as observed in vivo. This model enables the study of anti-angiogenic drugs which target a specific factor/receptor pathway, as demonstrated by the use of the clinically approved sorafenib and sunitinib for targeting the VEGF-A/VEGFR-2 pathway. Furthermore, this model allows testing simultaneously angiogenesis and permeability. It demonstrates that sorafenib impairs the endothelial barrier function, while sunitinib does not. Such in vitro human model provides a significant complimentary approach to animal models for the development of effective therapies.
Collapse
Affiliation(s)
- Joris Pauty
- Center for International Research on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan; LIMMS/CNRS-IIS UMI 2820, Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan; CNRS/IIS/COL/Université Lille 1 SMMiL-E project, CNRS Délégation Nord-Pas de Calais et Picardie, 2 rue de Canonniers, Lille, Cedex 59046, France
| | - Ryo Usuba
- Center for International Research on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Irene Gayi Cheng
- Department of Bioengineering, Clemson University, 118 Engineering Service Dr., Clemson, SC 29634, USA
| | - Louise Hespel
- Ecole Normale Supérieure-PSL Research University, Département de Chimie, Sorbonne Universités, UPMC Université Paris 06, CNRS UMR 8640 PASTEUR, 24 rue Lhomond, 75005 Paris, France
| | - Haruko Takahashi
- Center for International Research on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Keisuke Kato
- R&D Department 1, SCREEN Holdings Co., Ltd., 322 Furukawa-cho, Hazukashi, Fushimi-ku, Kyoto 612-8486, Japan
| | - Masayoshi Kobayashi
- R&D Department 1, SCREEN Holdings Co., Ltd., 322 Furukawa-cho, Hazukashi, Fushimi-ku, Kyoto 612-8486, Japan
| | - Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Eujin Lee
- Center for International Research on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Florian Yger
- LAMSADE, CNRS UMR 7243, Université Paris-Dauphine, PSL Research University, 75016 Paris, France
| | - Fabrice Soncin
- CNRS/IIS/COL/Université Lille 1 SMMiL-E project, CNRS Délégation Nord-Pas de Calais et Picardie, 2 rue de Canonniers, Lille, Cedex 59046, France; Université Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T, F-59000 Lille, France.
| | - Yukiko T Matsunaga
- Center for International Research on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan; LIMMS/CNRS-IIS UMI 2820, Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan; CNRS/IIS/COL/Université Lille 1 SMMiL-E project, CNRS Délégation Nord-Pas de Calais et Picardie, 2 rue de Canonniers, Lille, Cedex 59046, France.
| |
Collapse
|
29
|
Williams KS, Secomb TW, El-Kareh AW. Additive Damage Models for Cellular Pharmacodynamics of Radiation-Chemotherapy Combinations. Bull Math Biol 2017; 80:1236-1258. [PMID: 28849417 DOI: 10.1007/s11538-017-0316-z] [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: 12/09/2016] [Accepted: 07/03/2017] [Indexed: 11/28/2022]
Abstract
Many cancer patients receive combination treatments with radiation and chemotherapy. Available mathematical models for cellular pharmacodynamics have limited ability to represent observed in vitro responses to radiochemotherapy. Here, a family of additive damage models is proposed to describe cell kill resulting from radiochemotherapy with fixed schedule and variable doses. The pathways by which the agents produce cellular damage are assumed to converge in a single cell death process, so that survival depends on total damage, which can be represented as a sum of contributions from the various damage pathways. Heterogeneity in response across the cell population is ascribed to variations in the damage threshold for cell kill. The family of proposed models includes effects of one or two pathways of damage for each agent, saturation in drug responses, and cooperative or antagonistic interactions between agents. Models from this family with 4-7 unknown parameters are tested for their ability to fit 218 in vitro literature data sets for a range of drugs and cell lines. Overall, the additive damage models are found to outperform models based on the existing concept of independent cell kill, according to the corrected Akaike Information Criterion. The results are used to assess the importance of the various effects included in the models. These additive damage models have potential applications to the optimization of treatment and to the analysis and interpretation of in vitro screening data for new drug-radiation combinations.
Collapse
Affiliation(s)
| | - Timothy W Secomb
- Program in Applied Mathematics, University of Arizona, Tucson, AZ, USA.,Microcirculation Division, University of Arizona, Tucson, AZ, USA.,Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Ardith W El-Kareh
- Program in Applied Mathematics, University of Arizona, Tucson, AZ, USA. .,Microcirculation Division, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
30
|
Zhang X, Xu Y, Chen JM, Liu C, Du GL, Zhang H, Chen GF, Jiang SL, Liu CH, Mu YP, Liu P. Huang Qi Decoction Prevents BDL-Induced Liver Fibrosis Through Inhibition of Notch Signaling Activation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:85-104. [PMID: 28081630 DOI: 10.1142/s0192415x17500070] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Notch signaling has been demonstrated to be involved in ductular reactions and fibrosis. Previous studies have shown that Huang Qi Decoction (HQD) can prevent the progression of cholestatic liver fibrosis (CLF). However, whether HQD affects the Notch signaling pathway is unclear. In this study, CLF was established by common bile duct ligation (BDL) in rats. At the end of the first week, the rats were randomly divided into a model group (i.e., BDL), an HQD group, and a sorafenib positive control group (SORA) and were treated for 3 weeks. Bile duct proliferation and liver fibrosis were determined by tissue staining. Activation of the Notch signaling pathway was evaluated by analyzing expressions of Notch-1, -2, -3, and -4, Jagged (JAG) 1, and Delta like (DLL)-1, -3, and -4. The results showed that HQD significantly reduced the deposition of collagen and the Hyp content of liver tissue and inhibited the activation of HSCs compared with the BDL group. In addition, HQD significantly decreased the protein and mRNA expressions of TGF-[Formula: see text]1 and [Formula: see text]-SMA. In contrast, HQD significantly enhanced expression of the Smad 7 protein. HQD also reduced biliary epithelial cell proliferation, and reduced the mRNA levels of CK7, CK8, CK18, SRY-related high mobility group-box gene (SOX) 9, epithelial cell adhesion molecule (EpCAM) and the positive areas of CK19 and OV6. In addition, the mRNA and protein expressions of Notch-3, -4, JAG1, and DLL-1, -3 were significantly reduced in the HQD compared to the BDL group. These results demonstrated that HQD may prevent biliary liver fibrosis through inhibition of the Notch signaling pathway, and it may be a potential treatment for cholestatic liver disease.
Collapse
Affiliation(s)
- Xiao Zhang
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
- § Longhua Hospital Affiliated to Shanghai University of TCM, Shanghai, P.R. China
| | - Ying Xu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Jia-Mei Chen
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Cheng Liu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Guang-Li Du
- † E-institute of Shanghai Municipal Education Commission, Shanghai University of TCM, Shanghai, P.R. China
| | - Hua Zhang
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Gao-Feng Chen
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Shi-Li Jiang
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Cheng-Hai Liu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Yong-Ping Mu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Ping Liu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- † E-institute of Shanghai Municipal Education Commission, Shanghai University of TCM, Shanghai, P.R. China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| |
Collapse
|
31
|
Bossé D, Ng T, Ahmad C, Alfakeeh A, Alruzug I, Biagi J, Brierley J, Chaudhury P, Cleary S, Colwell B, Cripps C, Dawson LA, Dorreen M, Ferland E, Galiatsatos P, Girard S, Gray S, Halwani F, Kopek N, Mahmud A, Martel G, Robillard L, Samson B, Seal M, Siddiqui J, Sideris L, Snow S, Thirwell M, Vickers M, Goodwin R, Goel R, Hsu T, Tsvetkova E, Ward B, Asmis T. Eastern Canadian Gastrointestinal Cancer Consensus Conference 2016. ACTA ACUST UNITED AC 2016; 23:e605-e614. [PMID: 28050151 DOI: 10.3747/co.23.3394] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The annual Eastern Canadian Gastrointestinal Cancer Consensus Conference 2016 was held in Montreal, Quebec, 5-7 February. Experts in radiation oncology, medical oncology, surgical oncology, and infectious diseases involved in the management of patients with gastrointestinal malignancies participated in presentations and discussion sessions for the purpose of developing the recommendations presented here. This consensus statement addresses multiple topics: ■ Follow-up and survivorship of patients with resected colorectal cancer■ Indications for liver metastasectomy■ Treatment of oligometastases by stereotactic body radiation therapy■ Treatment of borderline resectable and unresectable pancreatic cancer■ Transarterial chemoembolization in hepatocellular carcinoma■ Infectious complications of antineoplastic agents.
Collapse
Affiliation(s)
- D Bossé
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - T Ng
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - C Ahmad
- Newfoundland and Labrador: Dr. H. Bliss Murphy Cancer Centre, St. John's (Ahmad, Seal, Siddiqui)
| | - A Alfakeeh
- Quebec: Hôpital Charles-LeMoyne Cancer Centre, Greenfield Park (Samson); McGill University Health Centre, Montreal (Alfakeeh, Alruzug, Chaudhury, Kopek, Thirlwell, Ward); Sir Mortimer B. Davis Jewish General Hospital (Galiatsatos); Centre Hospitalier Pierre-Boucher (Ferland); Centre Hospitalier Universitaire de Montréal (Girard, Sideris)
| | - I Alruzug
- Quebec: Hôpital Charles-LeMoyne Cancer Centre, Greenfield Park (Samson); McGill University Health Centre, Montreal (Alfakeeh, Alruzug, Chaudhury, Kopek, Thirlwell, Ward); Sir Mortimer B. Davis Jewish General Hospital (Galiatsatos); Centre Hospitalier Pierre-Boucher (Ferland); Centre Hospitalier Universitaire de Montréal (Girard, Sideris)
| | - J Biagi
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - J Brierley
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - P Chaudhury
- Quebec: Hôpital Charles-LeMoyne Cancer Centre, Greenfield Park (Samson); McGill University Health Centre, Montreal (Alfakeeh, Alruzug, Chaudhury, Kopek, Thirlwell, Ward); Sir Mortimer B. Davis Jewish General Hospital (Galiatsatos); Centre Hospitalier Pierre-Boucher (Ferland); Centre Hospitalier Universitaire de Montréal (Girard, Sideris)
| | - S Cleary
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - B Colwell
- Nova Scotia: QEII Health Sciences Centre, Halifax (Colwell, Dorreen, Snow)
| | - C Cripps
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - L A Dawson
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - M Dorreen
- Nova Scotia: QEII Health Sciences Centre, Halifax (Colwell, Dorreen, Snow)
| | - E Ferland
- Quebec: Hôpital Charles-LeMoyne Cancer Centre, Greenfield Park (Samson); McGill University Health Centre, Montreal (Alfakeeh, Alruzug, Chaudhury, Kopek, Thirlwell, Ward); Sir Mortimer B. Davis Jewish General Hospital (Galiatsatos); Centre Hospitalier Pierre-Boucher (Ferland); Centre Hospitalier Universitaire de Montréal (Girard, Sideris)
| | - P Galiatsatos
- Quebec: Hôpital Charles-LeMoyne Cancer Centre, Greenfield Park (Samson); McGill University Health Centre, Montreal (Alfakeeh, Alruzug, Chaudhury, Kopek, Thirlwell, Ward); Sir Mortimer B. Davis Jewish General Hospital (Galiatsatos); Centre Hospitalier Pierre-Boucher (Ferland); Centre Hospitalier Universitaire de Montréal (Girard, Sideris)
| | - S Girard
- Quebec: Hôpital Charles-LeMoyne Cancer Centre, Greenfield Park (Samson); McGill University Health Centre, Montreal (Alfakeeh, Alruzug, Chaudhury, Kopek, Thirlwell, Ward); Sir Mortimer B. Davis Jewish General Hospital (Galiatsatos); Centre Hospitalier Pierre-Boucher (Ferland); Centre Hospitalier Universitaire de Montréal (Girard, Sideris)
| | - S Gray
- New Brunswick: Saint John Regional Hospital, Saint John (Gray)
| | - F Halwani
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - N Kopek
- Quebec: Hôpital Charles-LeMoyne Cancer Centre, Greenfield Park (Samson); McGill University Health Centre, Montreal (Alfakeeh, Alruzug, Chaudhury, Kopek, Thirlwell, Ward); Sir Mortimer B. Davis Jewish General Hospital (Galiatsatos); Centre Hospitalier Pierre-Boucher (Ferland); Centre Hospitalier Universitaire de Montréal (Girard, Sideris)
| | - A Mahmud
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - G Martel
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - L Robillard
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - B Samson
- Quebec: Hôpital Charles-LeMoyne Cancer Centre, Greenfield Park (Samson); McGill University Health Centre, Montreal (Alfakeeh, Alruzug, Chaudhury, Kopek, Thirlwell, Ward); Sir Mortimer B. Davis Jewish General Hospital (Galiatsatos); Centre Hospitalier Pierre-Boucher (Ferland); Centre Hospitalier Universitaire de Montréal (Girard, Sideris)
| | - M Seal
- Newfoundland and Labrador: Dr. H. Bliss Murphy Cancer Centre, St. John's (Ahmad, Seal, Siddiqui)
| | - J Siddiqui
- Newfoundland and Labrador: Dr. H. Bliss Murphy Cancer Centre, St. John's (Ahmad, Seal, Siddiqui)
| | - L Sideris
- Quebec: Hôpital Charles-LeMoyne Cancer Centre, Greenfield Park (Samson); McGill University Health Centre, Montreal (Alfakeeh, Alruzug, Chaudhury, Kopek, Thirlwell, Ward); Sir Mortimer B. Davis Jewish General Hospital (Galiatsatos); Centre Hospitalier Pierre-Boucher (Ferland); Centre Hospitalier Universitaire de Montréal (Girard, Sideris)
| | - S Snow
- Nova Scotia: QEII Health Sciences Centre, Halifax (Colwell, Dorreen, Snow)
| | - M Thirwell
- Quebec: Hôpital Charles-LeMoyne Cancer Centre, Greenfield Park (Samson); McGill University Health Centre, Montreal (Alfakeeh, Alruzug, Chaudhury, Kopek, Thirlwell, Ward); Sir Mortimer B. Davis Jewish General Hospital (Galiatsatos); Centre Hospitalier Pierre-Boucher (Ferland); Centre Hospitalier Universitaire de Montréal (Girard, Sideris)
| | - M Vickers
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - R Goodwin
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - R Goel
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - T Hsu
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - E Tsvetkova
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| | - B Ward
- Quebec: Hôpital Charles-LeMoyne Cancer Centre, Greenfield Park (Samson); McGill University Health Centre, Montreal (Alfakeeh, Alruzug, Chaudhury, Kopek, Thirlwell, Ward); Sir Mortimer B. Davis Jewish General Hospital (Galiatsatos); Centre Hospitalier Pierre-Boucher (Ferland); Centre Hospitalier Universitaire de Montréal (Girard, Sideris)
| | - T Asmis
- Ontario: The Ottawa Hospital Cancer Centre, Ottawa (Asmis, Bossé, Cripps, Goel, Goodwin, Halwani, Hsu, Martel, Ng, Robillard, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Biagi); Princess Margaret Cancer Centre, Toronto (Brierley, Cleary, Dawson); Juravinski Cancer Centre, Hamilton (Tsvetkova); Cancer Centre of Southeastern Ontario, Kingston (Mahmud)
| |
Collapse
|
32
|
Salman A, Simoneau E, Hassanain M, Chaudhury P, Boucher LM, Valenti D, Cabrera T, Nudo C, Metrakos P. Combined sorafenib and yttrium-90 radioembolization for the treatment of advanced hepatocellular carcinoma. ACTA ACUST UNITED AC 2016; 23:e472-e480. [PMID: 27803608 DOI: 10.3747/co.23.2827] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS In this pilot study, we assessed the safety and tolerability of combining sorafenib with 90Y radioembolization for the treatment of unresectable hepatocellular carcinoma (hcc). METHODS The study, conducted prospectively during 2009-2012, included eligible patients with unresectable hcc and a life expectancy of at least 12 weeks. Each patient received sorafenib (400 mg twice daily) for 6-8 weeks before 90Y treatment. Safety and tolerability were assessed. RESULTS Of the 40 patients enrolled, 29 completed treatment (combined therapy). In the initial cohort, the most common cause of hcc was hepatitis C (32.5%), and most patients were staged Child A (82.5%). The 29 patients who completed the study had similar baseline characteristics. Grades 1 and 2 toxicities accounted for 77.8% of all adverse events reported. The most common toxicities reported were fatigue (19.0%), alteration in liver function (7.9%), and diarrhea (6.3%). There were 12 grade 3 and 2 grade 4 toxicity events reported. One patient died of liver failure within 30 days after treatment. During the study, the sorafenib dose was reduced in 6 patients (20.7%), and sorafenib had to be interrupted in 4 patients (13.8%) and discontinued in 4 patients (13.8%). The disease control rate was 72.4% per the modified Response Evaluation Criteria in Solid Tumors, and tumour necrosis was observed in 82.8% of patients. Overall survival in patients undergoing combined therapy was 12.4 months. CONCLUSIONS Preliminary results demonstrate the safety and tolerability of combining 90Y radioembolization and sorafenib for advanced hcc. A larger prospective study is needed to determine the extent of the survival benefit.
Collapse
Affiliation(s)
- A Salman
- Department of Surgery, Division of Hepatopancreatobiliary and Transplant Surgery, McGill University, Montreal, QC
| | - E Simoneau
- Department of Surgery, Division of Hepatopancreatobiliary and Transplant Surgery, McGill University, Montreal, QC
| | - M Hassanain
- Department of Oncology, McGill University, Montreal, QC; Department of Surgery, King Saud University, Riyadh, Saudi Arabia
| | - P Chaudhury
- Department of Surgery, Division of Hepatopancreatobiliary and Transplant Surgery, McGill University, Montreal, QC; Department of Oncology, McGill University, Montreal, QC
| | - L M Boucher
- Department of Radiology, Division of Interventional Radiology, McGill University, Montreal, QC
| | - D Valenti
- Department of Radiology, Division of Interventional Radiology, McGill University, Montreal, QC
| | - T Cabrera
- Department of Radiology, Division of Interventional Radiology, McGill University, Montreal, QC
| | - C Nudo
- Department of Medicine, Division of Hepatology, McGill University, Montreal, QC
| | - P Metrakos
- Department of Surgery, Division of Hepatopancreatobiliary and Transplant Surgery, McGill University, Montreal, QC; Department of Oncology, McGill University, Montreal, QC
| |
Collapse
|
33
|
Qu K, Liu T, Lin T, Zhang X, Cui R, Liu S, Meng F, Zhang J, Tai M, Wan Y, Liu C. Tyrosine kinase inhibitors: friends or foe in treatment of hepatic fibrosis? Oncotarget 2016; 7:67650-67660. [PMID: 27588502 PMCID: PMC5341902 DOI: 10.18632/oncotarget.11767] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 08/29/2016] [Indexed: 12/21/2022] Open
Abstract
Aberrant activity of tyrosine kinases has been proved to be associated with multiple diseases including fibrotic diseases. Tyrosine kinases inhibitors (TKIs) might be a novel approach to transform the anti-fibrotic treatment. However, both beneficial and adverse effects are observed by researchers when using these TKIs in either preclinical animal models or patients with hepatic fibrosis. Since hepatotoxicity of TKIs is the leading cause for drug withdrawals thus limits its application in anti-fibrosis, not only efficacy but also safety of TKIs should be paid great concerns. It has been observed in a few studies that TKIs could induce relatively high rate of hepatic biochemical markers elevations and even result in liver failure. Fortunately, several strategies have been adopt to handle with the hepatotoxicity. Accumulating evidences suggest that hepatic stellate cells (HSC) play a pivotal role in hepatic fibrogenesis, so it might be a good option to develop selective TKIs specifically targeting HSCs. The present review will briefly summarize the anti-fibrotic mechanism of TKIs, adverse effects of TKIs as well as the novel developed selective delivery of TKIs.
Collapse
Affiliation(s)
- Kai Qu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tian Liu
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ting Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Surgical Intensive Care Unit (SICU), The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xing Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ruixia Cui
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Sinan Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Surgical Intensive Care Unit (SICU), The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fandi Meng
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingyao Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Surgical Intensive Care Unit (SICU), The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Minghui Tai
- Department of Ultrasound, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yong Wan
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Surgical Intensive Care Unit (SICU), The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
34
|
Egger JV, Lane MV, Antonucci LA, Dedi B, Krucher NA. Dephosphorylation of the Retinoblastoma protein (Rb) inhibits cancer cell EMT via Zeb. Cancer Biol Ther 2016; 17:1197-1205. [PMID: 27645778 PMCID: PMC5137485 DOI: 10.1080/15384047.2016.1235668] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The tumor suppressor Retinoblastoma (Rb) protein is highly phosphorylated in cancer cells largely due to the overexpression of cyclins or the loss of expression of cyclin dependent kinase inhibitors (cdki). Hyperphosphorylation of Rb promotes proliferation, and plays a role in the regulation of apoptosis. Recently, inhibition of cyclin dependent activity toward Rb has been identified as a strategy that has shown clinical efficacy. We utilized a method to induce phosphatase activity toward Rb in cells by shRNA silencing of PNUTS (Phosphatase Nuclear Targeting Subunit) that regulates PP1-mediated dephosphorylation of Rb. In this study, the effect of Rb dephosphorylation on the epithelial to mesenchymal transition (EMT) was determined. The EMT transition is observed in cancer cells that have acquired invasive characteristics. In breast cancer cells grown in 3D Matrigel cultures, MCF7 cells undergo apoptosis in response to Rb dephosphorylation, whereas MDA-MB-231 and Hs578T cells exhibit a reduction in the EMT. Cells devoid of phosphorylated Rb (nontransformed MCF10A and Rb-null MDA-MB-468) lacked any response to PNUTS depletion, showing the effect is Rb-dependent. In addition, these studies showed that Rb dephosphorylation in 3D Matrigel cultures of highly invasive HT1080 cells led to the inhibition of the EMT. Furthermore we observed association between dephosphorylated Rb with ZEB1, a zinc-finger E-box-binding transcription factor that regulates expression of E- and N-cadherins. Finally Rb dephosphorylation led to inhibition of ZEB1 transcriptional activity, this data supports the notion that Rb dephosphorylation modulates the EMT. These studies suggest targeting Rb phosphorylation in mesenchymal cancer cells may decrease invasiveness.
Collapse
Affiliation(s)
- Jacklynn V Egger
- a Department of Biology , Dyson Hall, Pace University , Pleasantville , NY , USA
| | - Maria V Lane
- a Department of Biology , Dyson Hall, Pace University , Pleasantville , NY , USA
| | - Lisa A Antonucci
- a Department of Biology , Dyson Hall, Pace University , Pleasantville , NY , USA
| | - Brixhilda Dedi
- a Department of Biology , Dyson Hall, Pace University , Pleasantville , NY , USA
| | - Nancy A Krucher
- a Department of Biology , Dyson Hall, Pace University , Pleasantville , NY , USA
| |
Collapse
|
35
|
|
36
|
Qu K, Huang Z, Lin T, Liu S, Chang H, Yan Z, Zhang H, Liu C. New Insight into the Anti-liver Fibrosis Effect of Multitargeted Tyrosine Kinase Inhibitors: From Molecular Target to Clinical Trials. Front Pharmacol 2016; 6:300. [PMID: 26834633 PMCID: PMC4716646 DOI: 10.3389/fphar.2015.00300] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/08/2015] [Indexed: 12/13/2022] Open
Abstract
Tyrosine kinases (TKs) is a family of tyrosine protein kinases with important functions in the regulation of a broad variety of physiological cell processes. Overactivity of TK disturbs cellular homeostasis and has been linked to the development of certain diseases, including various fibrotic diseases. In regard to liver fibrosis, several TKs, such as vascular endothelial growth factor receptor, platelet-derived growth factor receptor, fibroblast growth factor receptor, and epidermal growth factor receptor kinases, have been identified as central mediators in collagen production and potential targets for anti-liver fibrosis therapies. Given the essential role of TKs during liver fibrogenesis, multitargeted inhibitors of aberrant TK activity, including sorafenib, erlotinib, imatinib, sunitinib, nilotinib, brivanib and vatalanib, have been shown to have potential for treating liver fibrosis. Beneficial effects are observed by researchers of this field using these multitargeted TK inhibitors in preclinical animal models and in patients with liver fibrosis. The present review will briefly summarize the anti-liver fibrosis effects of multitargeted TK inhibitors and molecular mechanisms.
Collapse
Affiliation(s)
- Kai Qu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an, China
| | - Zichao Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong UniversityXi'an, China; Department of General Surgery, Shaanxi Cancer HospitalXi'an, China
| | - Ting Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an, China
| | - Sinan Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an, China
| | - Hulin Chang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong UniversityXi'an, China; Department of Hepatobiliary Surgery, Shaanxi Provincial People's HospitalXi'an, China
| | - Zhaoyong Yan
- Department of Pain Treatment, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Hongxin Zhang
- Department of Pain Treatment, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Chang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an, China
| |
Collapse
|
37
|
Hsu C, Lin LI, Cheng YC, Feng ZR, Shao YY, Cheng AL, Ou DL. Cyclin E1 Inhibition can Overcome Sorafenib Resistance in Hepatocellular Carcinoma Cells Through Mcl-1 Suppression. Clin Cancer Res 2015; 22:2555-64. [PMID: 26603262 DOI: 10.1158/1078-0432.ccr-15-0499] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 10/27/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE To clarify the effects of cyclin E1 suppression on antitumor efficacy of sorafenib in hepatocellular carcinoma cells and to explore the potential of combining sorafenib with cyclin-dependent kinase (CDK) inhibition in therapy. EXPERIMENTAL DESIGN The effects of cyclin E1 suppression on sorafenib-induced apoptosis were tested in both sorafenib-sensitive (Huh-7 and HepG2, IC50 5-6 μmol/L) and sorafenib-resistant (Huh-7R and HepG2R, IC50 14-15 μmol/L) hepatocellular carcinoma cells. The activity of pertinent signaling pathways and the expression of cell cycle and apoptosis-related proteins were measured using Western blotting. Efficacy of sorafenib combined with the pan-CDK inhibitor flavopiridol was tested both in vitro and in xenograft experiments. The pertinent downstream mediators of antitumor efficacy were tested in transient transfection and RNA interference experiments. RESULTS Cyclin E1 mRNA and protein expressions were suppressed after sorafenib treatment in sorafenib-sensitive but not in sorafenib-resistant hepatocellular carcinoma cells. Changes in cyclin E2 or D1 were not correlated with sorafenib sensitivity. The knockdown of cyclin E1 expression reversed the resistance of hepatocellular carcinoma cells to sorafenib in terms of cell growth and apoptosis induction, whereas the overexpression of cyclin E1 increased the resistance to sorafenib. The growth-inhibitory and apoptosis-inducing effects of sorafenib were enhanced by flavopiridol, and Mcl-1 suppression was determined to play a critical role in mediating this enhancing effect. CONCLUSIONS The cyclin E1 suppression in hepatocellular carcinoma cells may serve as a pharmacodynamic biomarker for predicting sorafenib efficacy. The combination of sorafenib and CDK inhibitors may improve the efficacy of sorafenib in hepatocellular carcinoma. Clin Cancer Res; 22(10); 2555-64. ©2015 AACR.
Collapse
Affiliation(s)
- Chiun Hsu
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan. Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan. National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Liang-In Lin
- Graduate Institute of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Che Cheng
- Graduate Institute of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Zi-Rui Feng
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan. National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Yun Shao
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan. Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ann-Lii Cheng
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan. Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan. National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan. Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan. Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taiwan
| | - Da-Liang Ou
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan. National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
38
|
Liu C, Yang Z, Wang L, Lu Y, Tang B, Miao H, Xu Q, Chen X. Combination of sorafenib and gadolinium chloride (GdCl3) attenuates dimethylnitrosamine(DMN)-induced liver fibrosis in rats. BMC Gastroenterol 2015; 15:159. [PMID: 26572488 PMCID: PMC4647665 DOI: 10.1186/s12876-015-0380-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 10/19/2015] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND/AIMS Liver sinusoidal endothelial cells (SECs), hepatic stellate cells (HSCs) and Kupffer cells (KCs) are involved in the development of liver fibrosis and represent a potential therapeutic target. The therapeutic effects on liver fibrosis of sorafenib, a multiple tyrosine kinase inhibitor, and gadolinium chloride (GdCl3), which depletes KCs, were evaluated in rats. METHODS Liver fibrosis was induced in rats with dimethylnitrosamine, and the effects of sorafenib and/or GdCl3 in these rats were monitored. Interactions among ECs, HSCs and KCs were assessed by laser confocal microscopy. RESULTS The combination of sorafenib and GdCl3, but not each agent alone, attenuated liver fibrosis and significantly reduced liver function and hydroxyproline (Hyp). Sorafenib significantly inhibited the expression of angiogenesis-associated cell markers and cytokines, including CD31, von Willebrand factor (vWF), and vascular endothelial growth factor, whereas GdCl3 suppressed macrophage-related cell markers and cytokines, including CD68, tumor necrosis factor-α, interleukin-1β, and CCL2. Laser confocal microscopy showed that sorafenib inhibited vWF expression and GdCl3 reduced CD68 staining. Sorafenib plus GdCl3 suppressed the interactions of HSCs, ECs and KCs. CONCLUSION Sorafenib plus GdCl3 can suppress collagen accumulation, suggesting that this combination may be a potential therapeutic strategy in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Cheng Liu
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
- Laboratory of Molecular Pathology, Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Zongguo Yang
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| | - Lei Wang
- Department of Hepatology, Affiliated hospital of Shandong University of Trasitional Chinese Medicine, 250014, Jinan, China.
| | - Yunfei Lu
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| | - Bozong Tang
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| | - Hui Miao
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| | - Qingnian Xu
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| | - Xiaorong Chen
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| |
Collapse
|
39
|
Antonucci LA, Egger JV, Krucher NA. Phosphorylation of the Retinoblastoma protein (Rb) on serine-807 is required for association with Bax. Cell Cycle 2015; 13:3611-7. [PMID: 25483096 PMCID: PMC4614104 DOI: 10.4161/15384101.2014.964093] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The recent finding that the Retinoblastoma protein (Rb) is able to regulate apoptosis in a non-transcriptional manner directly at the mitochondria by interaction with the pro-apoptotic protein Bax prompted this investigation of the complex formed between Rb and Bax. Because the function of Rb in the cellular processes of proliferation, apoptosis, senescence and differentiation is regulated by phosphorylation we endeavored to elucidate the phosphorylation status of Rb with respect to its association with Bax and its role in apoptosis. In this study we found that Rb phosphorylated on at least 4 C-terminal phosphorylation sites (S608, S795, S807/S811, and T821) is present at the mitochondria under non-stressed cellular conditions. An in vitro binding assay showed that Bax binds to Rb phosphorylated at S807/S811 in 3 cancer cell types. Physiologically relevant association between Bax and Rb phosphorylated on S807/S811 was demonstrated by reciprocal co-immunoprecipitation experiments using antibodies specific for Rb phosphorylated on S807/S811 and Bax. Mutant Rb proteins expressed in Rb-null C33A cells showed that phosphorylation of S807 of Rb promotes association with Bax and that mimicking phosphorylation at S807 of Rb can block the induction of apoptosis due to PNUTS downregulation. Finally using siRNA to activate phosphatase activity in MCF7 cells, Rb is dephosphorylated at several sites including S807/S811, dissociates from Bax and apoptosis is triggered. These studies show that phosphorylation of Rb regulates its association with Bax and its role in apoptosis.
Collapse
Affiliation(s)
- Lisa A Antonucci
- a Department of Biology and Health Science ; Pace University ; Pleasantville , NY USA
| | | | | |
Collapse
|
40
|
Advani SJ, Camargo MF, Seguin L, Mielgo A, Anand S, Hicks AM, Aguilera J, Franovic A, Weis SM, Cheresh DA. Kinase-independent role for CRAF-driving tumour radioresistance via CHK2. Nat Commun 2015; 6:8154. [PMID: 26333361 PMCID: PMC4559870 DOI: 10.1038/ncomms9154] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 07/24/2015] [Indexed: 12/19/2022] Open
Abstract
Although oncology therapy regimens commonly include radiation and genotoxic drugs, tumour cells typically develop resistance to these interventions. Here we report that treatment of tumours with ionizing radiation or genotoxic drugs drives p21-activated kinase 1 (PAK1)-mediated phosphorylation of CRAF on Serine 338 (pS338) triggering a kinase-independent mechanism of DNA repair and therapeutic resistance. CRAF pS338 recruits CHK2, a cell cycle checkpoint kinase involved in DNA repair, and promotes CHK2 phosphorylation/activation to enhance the tumour cell DNA damage response. Accordingly, a phospho-mimetic mutant of CRAF (S338D) is sufficient to induce the CRAF/CHK2 association enhancing tumour radioresistance, while an allosteric CRAF inhibitor sensitizes tumour cells to ionizing radiation or genotoxic drugs. Our findings establish a role for CRAF in the DNA damage response that is independent from its canonical function as a kinase. Tumors hijack cellular pathways to evade the effects of cancer therapy. Here, Advani et al. show that DNA damage-induced phosphorylation of CRAF Serine 338 triggers DNA repair by recruiting CHK2, highlighting a role for CRAF independent from its canonical function as a kinase.
Collapse
Affiliation(s)
- Sunil J Advani
- Department of Radiation Medicine and Applied Sciences at the UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Maria Fernanda Camargo
- Department of Pathology at the UC San Diego Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, 3855 Health Science Drive, La Jolla, California 92037, USA
| | - Laetitia Seguin
- Department of Pathology at the UC San Diego Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, 3855 Health Science Drive, La Jolla, California 92037, USA
| | - Ainhoa Mielgo
- Department of Pathology at the UC San Diego Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, 3855 Health Science Drive, La Jolla, California 92037, USA
| | - Sudarshan Anand
- Department of Pathology at the UC San Diego Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, 3855 Health Science Drive, La Jolla, California 92037, USA
| | - Angel M Hicks
- Department of Radiation Medicine and Applied Sciences at the UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Joseph Aguilera
- Department of Radiation Medicine and Applied Sciences at the UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Aleksandra Franovic
- Department of Pathology at the UC San Diego Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, 3855 Health Science Drive, La Jolla, California 92037, USA
| | - Sara M Weis
- Department of Pathology at the UC San Diego Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, 3855 Health Science Drive, La Jolla, California 92037, USA
| | - David A Cheresh
- Department of Pathology at the UC San Diego Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, 3855 Health Science Drive, La Jolla, California 92037, USA
| |
Collapse
|
41
|
Zhai B, Jiang X, He C, Zhao D, Ma L, Xu L, Jiang H, Sun X. Arsenic trioxide potentiates the anti-cancer activities of sorafenib against hepatocellular carcinoma by inhibiting Akt activation. Tumour Biol 2015; 36:2323-2334. [PMID: 25416439 DOI: 10.1007/s13277-014-2839-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 11/11/2014] [Indexed: 01/05/2023] Open
Abstract
Sorafenib is the standard first-line systemic drug for advanced hepatocellular carcinoma (HCC), but it also induces the activation of Akt, which contributes to the mechanisms for the resistance to sorafenib. Arsenic trioxide (ATO) is a currently clinically used anticancer drug and displays its anticancer activities by inhibiting Akt activation. Therefore, we hypothesized that ATO may potentiate the anti-cancer activities of sorafenib against HCC. The results have demonstrated that ATO synergized with sorafenib to inhibit the proliferation and promote the apoptosis of HCC cells by diminishing the increased activation of Akt by sorafenib. ATO was shown to inhibit the expression or activation of Akt downstream factors, including glycogen synthase kinase (GSK)-3β, mammalian target of rapamycin (mTOR), ribosomal protein S6 kinase (S6K), and eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1), which regulate cell apoptosis and were upregulated or activated by sorafenib. Both sorafenib and ATO downregulated the expression of cyclin D1, resulting in HCC cells arrested at G0/G1 phase. ATO downregulated the expression of Bcl-2 and Bcl-xL and upregulated the expression of Bax, indicating that ATO could induce the apoptosis of HCC cells through the intrinsic pathways; but sorafenib showed little effects on these proteins of Bcl-2 family. ATO synergized with sorafenib to suppress the growth of HCC tumors established in mice by inhibiting the proliferation and inducing the apoptosis of HCC cells in situ. These results indicate that ATO may be a potential agent that given in combination with sorafenib acts synergistically for treating HCC.
Collapse
Affiliation(s)
- Bo Zhai
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Fumagalli I, Dugue D, Bibault JE, Clémenson C, Vozenin MC, Mondini M, Deutsch E. Cytotoxic effect of lapatinib is restricted to human papillomavirus-positive head and neck squamous cell carcinoma cell lines. Onco Targets Ther 2015; 8:335-45. [PMID: 25678800 PMCID: PMC4322874 DOI: 10.2147/ott.s68235] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Lapatinib is a dual epidermal growth factor receptor (EGFR) and HER2 inhibitor. Overexpression of these receptors is frequently observed in head and neck squamous cell carcinoma (HNSCC). As growing proportion of HNSCC is characterized by human papillomavirus (HPV) infection, we aimed at evaluating the efficacy of lapatinib as function of HPV status in HNSCC cell lines. Methods Two HPV-positive and two HPV-negative HNSCC cell lines were used. Proliferation, cell cycle, and Annexin V assays were performed to test their sensitivity to lapatinib. Combination of lapatinib and ionizing radiation was evaluated with clonogenic survival assays. Akt, EGFR and HER2, and E6/E7 expression and activation were analyzed by immunoblotting and quantitative reverse transcription polymerase chain reaction. Results Lapatinib reduced E6 and E7 expression and Akt phosphorylation, inhibited cell proliferation and induced cell death in HPV-positive cell lines. An additive effect of lapatinib with radiation was observed in these cells. Lapatinib had no effect on HPV-negative cells. Conclusion Lapatinib efficacy restricted to the HPV-positive cells suggests that HPV status could be a potential marker for enhanced response to lapatinib in HNSCC.
Collapse
Affiliation(s)
- Ingrid Fumagalli
- Inserm U1030, Molecular Radiotherapy, LABEX LERMIT, Gustave Roussy, University Paris XI, Villejuif, France ; Radiation Therapy Department, Oscar Lambret Comprehensive Cancer Center, Lille, France
| | - Delphine Dugue
- Inserm U1030, Molecular Radiotherapy, LABEX LERMIT, Gustave Roussy, University Paris XI, Villejuif, France
| | - Jean Emmanuel Bibault
- Inserm U1030, Molecular Radiotherapy, LABEX LERMIT, Gustave Roussy, University Paris XI, Villejuif, France ; Radiation Therapy Department, Oscar Lambret Comprehensive Cancer Center, Lille, France
| | - Céline Clémenson
- Inserm U1030, Molecular Radiotherapy, LABEX LERMIT, Gustave Roussy, University Paris XI, Villejuif, France
| | - Marie Catherine Vozenin
- Inserm U1030, Molecular Radiotherapy, LABEX LERMIT, Gustave Roussy, University Paris XI, Villejuif, France
| | - Michele Mondini
- Inserm U1030, Molecular Radiotherapy, LABEX LERMIT, Gustave Roussy, University Paris XI, Villejuif, France
| | - Eric Deutsch
- Inserm U1030, Molecular Radiotherapy, LABEX LERMIT, Gustave Roussy, University Paris XI, Villejuif, France ; Department of Radiation Oncology, Gustave Roussy, University Paris XI, Villejuif, France
| |
Collapse
|
43
|
Ricke J, Bulla K, Kolligs F, Peck-Radosavljevic M, Reimer P, Sangro B, Schott E, Schütte K, Verslype C, Walecki J, Malfertheiner P. Safety and toxicity of radioembolization plus Sorafenib in advanced hepatocellular carcinoma: analysis of the European multicentre trial SORAMIC. Liver Int 2015; 35:620-6. [PMID: 24930619 DOI: 10.1111/liv.12622] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 06/07/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS The benefits of combined systemic and liver-directed treatments in inoperable intermediate- or advanced-stage hepatocellular carcinoma (HCC) have yet to be defined. This article presents the planned safety analyses for the first 40 patients randomized to radioembolization with yttrium-90 ((90) Y) resin microspheres followed by sorafenib (n = 20) or sorafenib only (n = 20) in the SORAMIC study. METHODS Patients identified for palliative treatment who were poor candidates for transarterial (chemo)embolization (including those failing TACE) with preserved liver function (Child-Pugh ≤B7) and ECOG performance status <2 were screened. Radioembolization was administered using a sequential lobar approach. On day 3 after the last radioembolization procedure, sorafenib 200 mg twice daily was initiated escalating to 400 mg twice daily 1 week later; a matching sorafenib dose schedule was initiated in the control arm. RESULTS Patients were followed up for a median of 8.3 months. Median total implanted activity of (90) Y was 1.87 (range: 0.54-2.35) GBq. Patients received a similar intensity and duration of sorafenib in the combination-treatment arm (median daily dose 614 mg over 8.5 months) and control arm (557 mg over 9.6 months). The incidence of total (196 vs. 222) and grade ≥3 (43 vs. 47) adverse events was similar in combination-treatment arm and control arm respectively (P > 0.05). No significant differences in the number of total or grade 3/4 toxicities were recorded for: total bilirubin, albumin, liver enzymes, ascites, Child-Pugh, fatigue, hand-foot skin reaction, blood pressure or diarrhoea. CONCLUSIONS Radioembolization followed by sorafenib appears to be as well tolerated as sorafenib alone.
Collapse
Affiliation(s)
- Jens Ricke
- Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Li J, Chen Y, Wan J, Liu X, Yu C, Li W. ABT-263 enhances sorafenib-induced apoptosis associated with Akt activity and the expression of Bax and p21((CIP1/WAF1)) in human cancer cells. Br J Pharmacol 2015; 171:3182-95. [PMID: 24571452 DOI: 10.1111/bph.12659] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 02/16/2014] [Accepted: 02/21/2014] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Sorafenib, a potent inhibitor that targets several kinases associated with tumourigenesis and cell survival, has been approved for clinical treatment as a single agent. However, combining sorafenib with other agents improves its anti-tumour efficacy in various preclinical tumour models. ABT-263, a second-generation BH3 mimic, binds to the anti-apoptotic family members Bcl-2, Bcl-xL and Bcl-w, and has been demonstrated to enhance TNFSF10 (TRAIL)-induced apoptosis in human hepatocarcinoma cells. Hence, we investigated the effects of ABT-263 treatment combined with sorafenib. EXPERIMENTAL APPROACH The effects of ABT-263 combined with sorafenib were investigated in vitro, on cell viability, clone formation and apoptosis, and the mechanism examined using western blot and flow cytometry. This combination was also evaluated in vivo, in a mouse xenograft model; tumour growth, volume and weights were measured and a TUNEL assay performed. KEY RESULTS ABT-263 enhanced sorafenib-induced apoptosis while sparing non-tumourigenic cells. Although ABT-263 plus sorafenib significantly stimulated intracellular reactive oxygen species production and subsequent mitochondrial depolarization, this was not sufficient to trigger cell apoptosis. ABT-263 plus sorafenib significantly decreased Akt activity, which was, at least partly, involved in its effect on apoptosis. Bax and p21 (CIP1/WAF1) were shown to play a critical role in ABT-263 plus sorafenib-induced apoptosis. Combining sorafenib with ABT-263 dramatically increased its efficacy in vivo. CONCLUSION AND IMPLICATIONS The anti-tumour activity of ABT-263 plus sorafenib may involve the induction of intrinsic cell apoptosis via inhibition of Akt, and reduced Bax and p21 expression. Our findings offer a novel effective therapeutic strategy for tumour treatment.
Collapse
Affiliation(s)
- Jingru Li
- College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | | | | | | | | | | |
Collapse
|
45
|
Guan Z, Yu X, Wang H, Wang H, Zhang J, Li G, Cao J, Teng L. Advances in the targeted therapy of liposarcoma. Onco Targets Ther 2015; 8:125-36. [PMID: 25609980 PMCID: PMC4293924 DOI: 10.2147/ott.s72722] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Liposarcoma (LPS) is the most common type of soft-tissue sarcoma. Complete surgical resection is the only curative means for localized disease; however, both radiation and conventional cytotoxic chemotherapy remain controversial for metastatic or unresectable disease. An increasing number of trials with novel targeted therapy of LPS have provided encouraging data during recent years. This review will provide an overview of the advances in our understanding of LPS and summarize the results of recent trials with novel therapies targeting different genetic and molecular aberrations for different subtypes of LPS.
Collapse
Affiliation(s)
- Zhonghai Guan
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, People's Republic of China
| | - Xiongfei Yu
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, People's Republic of China
| | - Haohao Wang
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, People's Republic of China
| | - Haiyong Wang
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, People's Republic of China
| | - Jing Zhang
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, People's Republic of China
| | - Guangliang Li
- Department of Medicine Oncology, Zhejiang Cancer Hospital, Zhejiang, People's Republic of China
| | - Jiang Cao
- Clinical Research Center, The 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lisong Teng
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, People's Republic of China
| |
Collapse
|
46
|
Marks EI, Tan C, Zhang J, Zhou L, Yang Z, Scicchitano A, El-Deiry WS. Regorafenib with a fluoropyrimidine for metastatic colorectal cancer after progression on multiple 5-FU-containing combination therapies and regorafenib monotherapy. Cancer Biol Ther 2015; 16:1710-9. [PMID: 26561209 PMCID: PMC4847811 DOI: 10.1080/15384047.2015.1113355] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/09/2015] [Accepted: 10/22/2015] [Indexed: 12/19/2022] Open
Abstract
We present 2 patients with metastatic colorectal cancer who had progressed despite treatment with first-line FOLFOX and second-line FOLFIRI combination chemotherapy regimens. After failing these fluoropyrimidine-based regimens, both patients received additional cytotoxic and targeted therapies with eventual disease progression. These therapies included capecitabine plus dabrafenib and trametinib, regorafenib monotherapy, and regorafenib with panitumumab. After exhausting available options, both patients were offered regorafenib with either 5-fluorouracil (5-FU) or capecitabine. These therapies are individually approved for the treatment of colorectal cancer but have not yet been studied in combination. This regimen produced stable disease in both patients with acceptable toxicity. One patient continued therapy for 17 months. Although these patients previously progressed during treatment with regorafenib, capecitabine or 5-FU, the combination had some activity in both cases of refractory metastatic colorectal cancer and may be considered in the palliative setting. In bedside-to-bench cell culture experiments performed after the clinical observations, we observed sensitivity of human colorectal cancer cell lines (N = 4) to single agent regorafenib or 5-FU and evidence of synergy with the combination therapy. Synergistic effects were noted in colorectal cancer cells with KRAS mutation, BRAF mutation, and p53 mutation, as well as mismatch repair deficient cells. Regorafenib suppressed Mcl-1 and Bcl-XL in treated cancer cells that may have contributed to the anticancer efficacy including in combination with 5-FU. The safety and efficacy of regorafenib with 5-FU or capecitabine in combination should be further investigated as a therapy for patients with refractory metastatic colorectal cancer, including individuals who had progressed on regorafenib monotherapy.
Collapse
Affiliation(s)
- Eric I. Marks
- Penn State College of Medicine and Penn State Hershey Cancer Institute; Hershey, PA 17033, USA
| | - Carlyn Tan
- Department of Hematology/Oncology and Molecular Therapeutics Program; Fox Chase Cancer Center; Philadelphia, PA 19111, USA
| | - Jun Zhang
- Department of Hematology/Oncology and Molecular Therapeutics Program; Fox Chase Cancer Center; Philadelphia, PA 19111, USA
| | - Lanlan Zhou
- Department of Hematology/Oncology and Molecular Therapeutics Program; Fox Chase Cancer Center; Philadelphia, PA 19111, USA
| | - Zhaohai Yang
- Penn State College of Medicine and Penn State Hershey Cancer Institute; Hershey, PA 17033, USA
| | - Angelique Scicchitano
- Penn State College of Medicine and Penn State Hershey Cancer Institute; Hershey, PA 17033, USA
| | - Wafik S. El-Deiry
- Penn State College of Medicine and Penn State Hershey Cancer Institute; Hershey, PA 17033, USA
- Department of Hematology/Oncology and Molecular Therapeutics Program; Fox Chase Cancer Center; Philadelphia, PA 19111, USA
| |
Collapse
|
47
|
Teixeira C, Vale N, Pérez B, Gomes A, Gomes JRB, Gomes P. "Recycling" classical drugs for malaria. Chem Rev 2014; 114:11164-220. [PMID: 25329927 DOI: 10.1021/cr500123g] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Cátia Teixeira
- Centro de Investigação em Química da Universidade do Porto, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto , P-4169-007 Porto, Portugal.,CICECO, Departamento de Química, Universidade de Aveiro , P-3810-193 Aveiro, Portugal
| | - Nuno Vale
- Centro de Investigação em Química da Universidade do Porto, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto , P-4169-007 Porto, Portugal
| | - Bianca Pérez
- Centro de Investigação em Química da Universidade do Porto, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto , P-4169-007 Porto, Portugal
| | - Ana Gomes
- Centro de Investigação em Química da Universidade do Porto, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto , P-4169-007 Porto, Portugal
| | - José R B Gomes
- CICECO, Departamento de Química, Universidade de Aveiro , P-3810-193 Aveiro, Portugal
| | - Paula Gomes
- Centro de Investigação em Química da Universidade do Porto, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto , P-4169-007 Porto, Portugal
| |
Collapse
|
48
|
Canter RJ, Ames E, Mac S, Grossenbacher SK, Chen M, Li CS, Borys D, Smith RC, Tellez J, Sayers TJ, Monjazeb AM, Murphy WJ. Anti-proliferative but not anti-angiogenic tyrosine kinase inhibitors enrich for cancer stem cells in soft tissue sarcoma. BMC Cancer 2014; 14:756. [PMID: 25301268 PMCID: PMC4200119 DOI: 10.1186/1471-2407-14-756] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/02/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Increasing studies implicate cancer stem cells (CSCs) as the source of resistance and relapse following conventional cytotoxic therapies. Few studies have examined the response of CSCs to targeted therapies, such as tyrosine kinase inhibitors (TKIs). We hypothesized that TKIs would have differential effects on CSC populations depending on their mechanism of action (anti-proliferative vs. anti-angiogenic). METHODS We exposed human sarcoma cell lines to sorafenib, regorafenib, and pazopanib and assessed cell viability and expression of CSC markers (ALDH, CD24, CD44, and CD133). We evaluated survival and CSC phenotype in mice harboring sarcoma metastases after TKI therapy. We exposed dissociated primary sarcoma tumors to sorafenib, regorafenib, and pazopanib, and we used tissue microarray (TMA) and primary sarcoma samples to evaluate the frequency and intensity of CSC markers after neoadjuvant therapy with sorafenib and pazopanib. Parametric and non-parametric statistical analyses were performed as appropriate. RESULTS After functionally validating the CSC phenotype of ALDHbright sarcoma cells, we observed that sorafenib and regorafenib were cytotoxic to sarcoma cell lines (P < 0.05), with a corresponding 1.4 - 2.8 fold increase in ALDHbright cells from baseline (P < 0.05). In contrast, we observed negligible effects on viability and CSC sub-populations with pazopanib. At low doses, there was progressive CSC enrichment in vitro after longer term exposure to sorafenib although the anti-proliferative effects were attenuated. In vivo, sorafenib improved median survival by 11 days (P < 0.05), but enriched ALDHbright cells 2.5 - 2.8 fold (P < 0.05). Analysis of primary human sarcoma samples revealed direct cytotoxicity following exposure to sorafenib and regorafenib with a corresponding increase in ALDHbright cells (P < 0.05). Again, negligible effects from pazopanib were observed. TMA analysis of archived specimens from sarcoma patients treated with sorafenib demonstrated significant enrichment for ALDHbright cells in the post-treatment resection specimen (P < 0.05), whereas clinical specimens obtained longitudinally from a patient treated with pazopanib showed no enrichment for ALDHbright cells (P > 0.05). CONCLUSIONS Anti-proliferative TKIs appear to enrich for sarcoma CSCs while anti-angiogenic TKIs do not. The rational selection of targeted therapies for sarcoma patients may benefit from an awareness of the differential impact of TKIs on CSC populations.
Collapse
Affiliation(s)
- Robert J Canter
- Department of Surgery, Division of Surgical Oncology, University of California Davis Medical Center, 4501 X Street, Sacramento, CA 95817, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kim EH, Kim MS, Jung WG. The mechanisms responsible for the radiosensitizing effects of sorafenib on colon cancer cells. Oncol Rep 2014; 32:2421-8. [PMID: 25242034 DOI: 10.3892/or.2014.3497] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/18/2014] [Indexed: 11/06/2022] Open
Abstract
Colorectal cancer is one of the most common malignancies in the world, and is generally treated more effectively by chemoradiotherapy rather than radiotherapy or chemotherapy alone. Targeted radiosensitizers are often used in order to enhance the radiosensitivity of tumor cells. The aim of the present study was to identify the mechanism of radiosensitization by sorafenib in colorectal cancer. Three human colorectal adenocarcinoma cell lines (HCT116, HT29 and SW480) were treated with sorafenib alone or radiation followed by sorafenib. In vitro tests were performed using colony forming assays, FACS analysis, immunohistochemistry, tumor cell motility assays, invasion assays and endothelial tube formation assays. Sorafenib enhanced the anti-proliferative effects of radiation, reducing colony formation, increasing G2/M arrest and enhancing radiation-induced apoptosis by reactive oxygen species. Sorafenib also inhibited the repair of radiation-induced DNA damage by blocking the activation of DNA-dependent protein kinase. Combination treatment significantly inhibited tumor cell migration, tumor cell invasion and vascular endothelial growth factor-mediated angiogenesis in vitro. Taken together, our results provide a scientific rationale for the use of sorafenib with radiotherapy in colon cancer and suggest a clinical utility for this approach.
Collapse
Affiliation(s)
- Eun Ho Kim
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Mi-Sook Kim
- Department of Radiation Oncology, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Won-Gyun Jung
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| |
Collapse
|
50
|
Cuneo KC, Davis MA, Feng MU, Novelli PM, Ensminger WD, Lawrence TS. Low dose rate radiosensitization of hepatocellular carcinoma in vitro and in patients. Transl Oncol 2014; 7:472-8. [PMID: 24956939 PMCID: PMC4202782 DOI: 10.1016/j.tranon.2014.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/21/2014] [Indexed: 01/17/2023] Open
Abstract
Transarterial radioembolization (TARE) with 90Y microspheres delivers low dose rate radiation (LDR) to intrahepatic tumors. In the current study, we examined clonogenic survival, DNA damage, and cell cycle distribution in hepatocellular carcinoma (HCC) cell lines treated with LDR in combination with varying doses and schedules of 5-fluorouracil (5-FU), gemcitabine, and sorafenib. Radiosensitization was seen with 1 to 3 μM 5-FU (enhancement ratio 2.2–13.9) and 30 to 100 nM gemcitabine (enhancement ratio 1.9–2.9) administered 24 hours before LDR (0.26 Gy/h to 4.2 Gy). Sorafenib radiosensitized only at high concentrations (3–10 μM) when administered after LDR. For a given radiation dose, greater enhancement was seen with LDR compared to standard dose rate therapy. Summarizing our clinical experience with low dose rate radiosensitization, 13 patients (5 with HCC, 8 with liver metastases) were treated a total of 16 times with TARE and concurrent gemcitabine. Six partial responses and one complete response were observed with a median time to local failure of 7.1 months for all patients and 9.9 months for patients with HCC. In summary, HCC is sensitized to LDR with clinically achievable concentrations of gemcitabine and 5-FU in vitro. Encouraging responses were seen in a small cohort of patients treated with TARE and concurrent gemcitabine. Future studies are needed to validate the safety and efficacy of this approach.
Collapse
Affiliation(s)
- Kyle C Cuneo
- University of Michigan Medical Center, Department of Radiation Oncology, Ann Arbor, MI; Ann Arbor Veterans Affairs Hospital, Department of Radiation Oncology, Ann Arbor, MI.
| | - Mary A Davis
- University of Michigan Medical Center, Department of Radiation Oncology, Ann Arbor, MI
| | - Mary U Feng
- University of Michigan Medical Center, Department of Radiation Oncology, Ann Arbor, MI
| | - Paula M Novelli
- University of Michigan Medical Center, Department of Radiology, Ann Arbor, MI
| | - William D Ensminger
- University of Michigan Medical Center, Department of Internal Medicine, Ann Arbor, MI
| | - Theodore S Lawrence
- University of Michigan Medical Center, Department of Radiation Oncology, Ann Arbor, MI
| |
Collapse
|