1
|
El-Hazek RMM, Zaher NH, El-Gazzar MGM, Fadel NA, El-Sabbagh WA. Novel VEGFR2 inhibitors with thiazoloquinoxaline scaffold targeting hepatocellular carcinoma with lower cardiotoxic impact. Sci Rep 2023; 13:13907. [PMID: 37626064 PMCID: PMC10457369 DOI: 10.1038/s41598-023-40832-z] [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: 05/20/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a fatal tumor which is usually diagnosed at advanced stage. Molecular targeted drugs were used recently to treat HCC, however, due to serious side effects, mainly cardiotoxicity and emergence of resistance, there is demanding to explore new chemotherapeutics. 10 novel thiazoloquinoxaline derivatives coupled with different sulfonamide moieties 4(a-j) were designed and synthesized fulfilling pharmacophoric features of VEGFR-2 inhibition. Structures of all new compounds were verified via spectral and microanalytical data. After carrying in-vitro VEGFR-2 assay for compounds 4(a-j); sulfapyridine and sulfamethoxazole derivatives 4d and 4f showed potential inhibitory effect [61.04 and 83.35 nM], respectively, comparable to standard sorafenib [51.41 nM]. Both were then further evaluated for their cytocidal activity against HepG2 cell-line and against myocardium cells using H9C2 cell-line. As a result, only sulfapyridine derivative 4d exhibited a significant inhibition of HepG2 cells viability [IC50 = 4.31 μM]. Furthermore, it showed relatively lower cytotoxic impact against normal H9C2 myocardium cells [IC50, 33.47 μM] compared to that of sorafenib [IC50, 98.07 μM]. In-vivo study was carried out to determine myocardium safety of compound 4d on irradiated mice (8 Gy). In-vivo results of sulfapyridine derivative 4d showed normal cardiac enzyme function (CK) and serum catalase activity with significant reductions in LDH, cardiac TNF-α and caspase-9 levels, alongside with its efficacy in suppressing the expression of hepatic VEGF. In conclusion, sulfapyridine derivative 4d could be considered a promising candidate as VEGFR-2 inhibitor with less myocardium side effect.
Collapse
Affiliation(s)
- Reham M M El-Hazek
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Nashwa H Zaher
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt.
| | - Mostafa G M El-Gazzar
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Noha A Fadel
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Walaa A El-Sabbagh
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| |
Collapse
|
2
|
Ji X, Xu Z, Sun J, Li W, Duan X, Wang Q. Lenvatinib with or without stereotactic body radiotherapy for hepatocellular carcinoma with portal vein tumor thrombosis: a retrospective study. Radiat Oncol 2023; 18:101. [PMID: 37308914 DOI: 10.1186/s13014-023-02270-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 04/26/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Patients with hepatocellular carcinoma (HCC) involving portal vein tumor thrombosis (PVTT) are presently lacking effective treatment options. We aimed to compare the efficacy and safety of lenvatinib with or without SBRT for HCC with PVTT. MATERIALS AND METHODS This retrospective analysis included 37 patients treated with lenvatinib in combination with SBRT and 77 patients treated with lenvatinib alone from August 2018 to August 2021. Overall survival (OS), progression-free survival (PFS), intrahepatic PFS (IHPFS) and objective remission rate (ORR) were compared between the two groups, while adverse events (AEs) was analyzed between the two groups to assess safety profiles. RESULTS Median OS, PFS and IHPFS were significantly prolonged in the combination treatment group compared with the single treatment group (median OS, 19.3 vs. 11.2 months, p < 0.001; median PFS: 10.3 vs. 5.3 months, p < 0.001; median IHPFS, 10.7 vs. 5.3 months, p < 0.001). Moreover, a higher ORR (56.8% vs. 20.8%, P < 0.001) were observed in the lenvatinib combined with SBRT group. In subgroup analyses of Vp1-2 and Vp3-4 group, median OS, PFS and IHPFS were also significantly longer in the lenvatinib combined with SBRT group than those in the lenvatinib alone group. AEs in the combined therapy group were mostly manageable and the incidence was not statistically significant compared to the monotherapy group. CONCLUSION Lenvatinib plus SBRT had a significantly better survival benefit than lenvatinib monotherapy in the treatment of HCC patients with PVTT and was well tolerated.
Collapse
Affiliation(s)
- Xiaoquan Ji
- Department of Radiation Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhe Xu
- Treatment and Research Center for Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China
| | - Jing Sun
- Department of Radiation Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Wengang Li
- Department of Radiation Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Xuezhang Duan
- Department of Radiation Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| | - Quan Wang
- Department of Radiation Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China.
| |
Collapse
|
3
|
Malik IA, Rajput M, Werner R, Fey D, Salehzadeh N, von Arnim CAF, Wilting J. Differential in vitro effects of targeted therapeutics in primary human liver cancer: importance for combined liver cancer. BMC Cancer 2022; 22:1193. [PMCID: PMC9675209 DOI: 10.1186/s12885-022-10247-6] [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: 09/09/2022] [Accepted: 10/29/2022] [Indexed: 11/21/2022] Open
Abstract
The incidence of primary liver tumors, hepatocellular carcinoma (HCC), intrahepatic cholangiocellular carcinoma (ICC), and combined HCC/ICC (cHCC/CC) is increasing. For ICC, targeted therapy exists only for a small subpopulation of patients, while for HCC, Sorafenib and Lenvatinib are in use. Diagnosis of cHCC/CC is a great challenge and its incidence is underestimated, bearing the risk of unintended non-treatment of ICC. Here, we investigated effects of targeted inhibitors on human ICC cell lines (HUH28, RBE, SSP25), in comparison to extrahepatic (E)CC lines (EGI1, CCC5, TFK1), and HCC/hepatoblastoma cell lines (HEP3B, HUH7, HEPG2). Cells were challenged with: AKT inhibitor MK-2206; multikinase inhibitors Sorafenib, Lenvatinib and Dasatinib; PI3-kinase inhibitors BKM-120, Wortmannin, LY294002, and CAL-101; and mTOR inhibitor Rapamycin. Dosage of the substances was based on the large number of published data of recent years. Proliferation was analyzed daily for four days. All cell lines were highly responsive to MK-2206. Thereby, MK-2206 reduced expression of phospho(p)-AKT in all ICC, ECC, and HCC lines, which mostly corresponded to reduction of p-mTOR, whereas p-ERK1/2 was upregulated in many cases. Lenvatinib showed inhibitory effects on the two HCC cell lines, but not on HEPG2, ICCs and ECCs. Sorafenib inhibited proliferation of all cells, except the ECC line CCC5. However, at reduced dosage, we observed increased cell numbers in some ICC experiments. Dasatinib was highly effective especially in ICC cell lines. Inhibitory effects were observed with all four PI3-kinase inhibitors. However, cell type-specific differences were also evident here. Rapamycin was most effective in the two HCC cell lines. Our studies show that the nine inhibitors differentially target ICC, ECC, and HCC/hepatoblastoma lines. Caution should be taken with Lenvatinib and Sorafenib administration in patients with cHCC/CC as the drugs may have no effects on, or might even stimulate, ICC.
Collapse
Affiliation(s)
- Ihtzaz Ahmed Malik
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany
| | - Mansi Rajput
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany
| | - Rieke Werner
- grid.411984.10000 0001 0482 5331Department of Anatomy and Cell Biology, University Medical Center Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany
| | - Dorothea Fey
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany ,grid.411984.10000 0001 0482 5331Department of Anatomy and Cell Biology, University Medical Center Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany
| | - Niloofar Salehzadeh
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany ,grid.411984.10000 0001 0482 5331Department of Anatomy and Cell Biology, University Medical Center Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany
| | - Christine A. F. von Arnim
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany
| | - Jörg Wilting
- grid.411984.10000 0001 0482 5331Department of Anatomy and Cell Biology, University Medical Center Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany
| |
Collapse
|
4
|
Cho Y, Kim BH, Kim TH, Koh YH, Park JW. Sorafenib combined with radiation therapy for advanced hepatocellular carcinoma with portal and hepatic vein invasion extending to the inferior vena cava: a complete response case according to modified RECIST criteria. JOURNAL OF LIVER CANCER 2022; 22:63-68. [PMID: 37383539 PMCID: PMC10035710 DOI: 10.17998/jlc.2022.01.18] [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: 12/15/2021] [Revised: 01/13/2022] [Accepted: 01/18/2022] [Indexed: 06/30/2023]
Abstract
The prognosis of patients with advanced hepatocellular carcinoma (HCC) with tumor thrombus extending to the inferior vena cava (IVC) is extremely poor. Herein, we present a rare case of advanced HCC that was treated with sorafenib and radiotherapy, leading to complete remission. This patient had a 9 cm infiltrative HCC occupying almost the entire left lobe with a tumor thrombus extending through the hepatic vein, IVC, and left portal vein. The patient received 400 mg sorafenib twice daily. One year after the start of sorafenib, intensity-modulated radiation therapy for viable HCC and tumor thrombus was performed with a dose of 5,500 cGy. Twenty-seven months after the starting date of sorafenib, there was no intratumoral arterial enhancement, which suggested a complete response according to the modified RECIST criteria. This case suggests that the combination of sorafenib and radiotherapy might provide clinical benefits in patients with advanced HCC with IVC tumor thrombus.
Collapse
Affiliation(s)
- Yuri Cho
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang, Korea
| | - Bo Hyun Kim
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang, Korea
| | - Tae Hyun Kim
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang, Korea
- Center for Proton Therapy, National Cancer Center, Goyang, Korea
| | - Young Hwan Koh
- Department of Radiology, National Cancer Center, Goyang, Korea
| | - Joong-Won Park
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang, Korea
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| |
Collapse
|
5
|
Hassan SA, Ali AAH, Sohn D, Flögel U, Jänicke RU, Korf H, von Gall C. Does timing matter in radiotherapy of hepatocellular carcinoma? An experimental study in mice. Cancer Med 2021; 10:7712-7725. [PMID: 34545699 PMCID: PMC8559477 DOI: 10.1002/cam4.4277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/18/2021] [Accepted: 08/27/2021] [Indexed: 01/10/2023] Open
Abstract
This study investigates whether a chronotherapeutic treatment of hepatocellular carcinoma (HCC) may improve treatment efficacy and mitigate side effects on non-tumoral liver (NTL). HCC was induced in Per2::luc mice which were irradiated at four time points of the day. Proliferation and DNA-double strand breaks were analyzed in irradiated and nonirradiated animals by detection of Ki67 and γ-H2AX. Prior to whole animal experiments, organotypic slice cultures were investigated to determine the dosage to be used in whole animal experiments. Irradiation was most effective at the proliferation peaks in HCC at ZT02 (early inactivity phase) and ZT20 (late activity phase). Irradiation effects on NTL were minimal at ZT20. As compared with NTL, nonirradiated HCC revealed disruption in daily variation and downregulation of all investigated clock genes except Per1. Irradiation affected rhythmic clock gene expression in NTL and HCC at all ZTs except at ZT20 (late activity phase). Irradiation at ZT20 had no effect on total leukocyte numbers. Our results indicate ZT20 as the optimal time point for irradiation of HCC in mice at which the ratio between efficacy of tumor treatment and toxic side effects was maximal. Translational studies are now needed to evaluate whether the late activity phase is the optimal time point for irradiation of HCC in man.
Collapse
Affiliation(s)
- Soha A. Hassan
- Institute of Anatomy II, Medical FacultyHeinrich‐Heine‐UniversityDüsseldorfGermany
- Zoology DepartmentFaculty of ScienceSuez UniversitySuezEgypt
| | - Amira A. H. Ali
- Institute of Anatomy II, Medical FacultyHeinrich‐Heine‐UniversityDüsseldorfGermany
- Department of Anatomy and EmbryologyFaculty of MedicineMansoura UniversityMansouraEgypt
| | - Dennis Sohn
- Laboratory of Molecular Radiooncology, Clinic and Policlinic for Radiation Therapy and RadiooncologyMedical Faculty of Heinrich‐Heine‐UniversityDüsseldorfGermany
| | - Ulrich Flögel
- Department of Molecular CardiologyHeinrich‐Heine‐UniversityDüsseldorfGermany
| | - Reiner U. Jänicke
- Laboratory of Molecular Radiooncology, Clinic and Policlinic for Radiation Therapy and RadiooncologyMedical Faculty of Heinrich‐Heine‐UniversityDüsseldorfGermany
| | - Horst‐Werner Korf
- Institute of Anatomy IMedical FacultyHeinrich‐Heine‐UniversityDüsseldorfGermany
| | - Charlotte von Gall
- Institute of Anatomy II, Medical FacultyHeinrich‐Heine‐UniversityDüsseldorfGermany
| |
Collapse
|
6
|
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
|
7
|
Feng XE. miR-548b Suppresses Melanoma Cell Growth, Migration, and Invasion by Negatively Regulating Its Target Gene HMGB1. Cancer Biother Radiopharm 2021; 36:189-201. [PMID: 33750228 DOI: 10.1089/cbr.2019.3507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background: Melanoma is one of the most aggressive malignancies. Exploration of metastasis-related genes will improve the clinical outcomes of patients with melanoma. Recently, microRNAs (miRNAs) have been implicated in regulating the aggressiveness of melanoma. In the current study, the author demonstrated the expression of miR-548b and its functions in melanoma. Materials and Methods: The expression levels of miR-548b and high mobility group protein 1 (HMGB1) in melanoma specimens and adjacent normal tissues were examined using the quantitative real-time PCR method. The Cell Counting Kit-8 (CCK-8), wound healing test, and Transwell assays were conducted to examine the impact of miR-548b on aggressive phenotypes of melanoma cells. The protein expression of HMGB1 was detected by Western blot. The tumor growth of melanoma cells in vivo was analyzed using the transplanted tumor model. The expression of HMGB1 in vivo was assessed using immunohistochemistry assay. Results: miR-548b was significantly downregulated in the melanoma sample when compared with adjacent normal tissues. In addition, low levels of miR-548b were related to poor overall survival in patients with melanoma. As predicted, overexpression of miR-548b suppressed the growth and metastasis-associated traits of melanoma cells. Furthermore, the luciferase reporter gene assay and Western blotting revealed that HMGB1 was a target of miR-548b and its expression level was negatively modulated by miR-548b. Several rescue experiments indicated that reintroduction of HMGB1 abolished the inhibiting effects of miR-548b on melanoma cells. Finally, the author demonstrated that upregulation of miR-548b repressed melanoma cell growth in vivo. Conclusions: All these findings demonstrate that miR-548b serves as a cancer-suppressive miRNA in human melanoma by inhibiting HMGB1.
Collapse
Affiliation(s)
- Xi-En Feng
- Department of Dermatology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
| |
Collapse
|
8
|
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: 7] [Impact Index Per Article: 2.3] [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
|
9
|
Pérez-Romasanta LA, González-Del Portillo E, Rodríguez-Gutiérrez A, Matías-Pérez Á. Stereotactic Radiotherapy for Hepatocellular Carcinoma, Radiosensitization Strategies and Radiation-Immunotherapy Combination. Cancers (Basel) 2021; 13:cancers13020192. [PMID: 33430362 PMCID: PMC7825787 DOI: 10.3390/cancers13020192] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Radiotherapy is rapidly turning into a crucial component of multidisciplinary treatment for liver cancer because many patients are not surgical treatment candidates. Thanks to technical developments, radiotherapy have achieved high precision treatments, making it possible to eliminate tumor cells without severe damage to the liver and other organs. Stereotactic Body Radiation Therapy is an advanced radiotherapy technique able to eradicate malignant tumors wherever they are located in properly selected patients. The best use of radiotherapy, the most fruitful radiotherapy strategy, and the best way to combine it with other treatments for liver cancer are largely unknown. Radiosensitizers, agents that can potentiate radiotherapy, could broaden the radiotherapeutic landscape. Radiotherapy potentiation can be achieved with diverse treatments, not only drugs but also nanoparticles. In order to clear up the performance of radiotherapy in liver cancer management in the future and the best ways to potentiate its effects, considerable medical research is needed. Abstract Stereotactic body radiotherapy (SBRT) is an emerging ablative modality for hepatocellular carcinoma (HCC). Most patients with HCC have advanced disease at the time of diagnosis, and therefore, are not candidates for definitive-intent therapies such as resection or transplantation. For this reason, various alternative local and regional therapies have been used to prevent disease progression, palliate symptoms, and delay liver failure. Stereotactic body radiation therapy is a non-invasive technique of delivering ablative doses of radiation to tumors while sparing normal or non-tumor hepatic tissue. Incorporation of SBRT in multidisciplinary HCC management is gradual, initially applied when other liver-directed therapies have failed or are contraindicated, and tried in combination with other locoregional or systemic therapies for more unfavorable conditions by more experienced teams. In order to improve SBRT therapeutic ratio, there has been much interest in augmenting the effect of radiation on tumors by combining it with chemotherapy, molecularly targeted therapeutics, nanoparticles, and immunotherapy. This review aims to synthesize available evidence to evaluate the clinical feasibility and efficacy of SBRT for HCC, and to explore novel radio-potentiation concepts by combining SBRT with novel therapeutics. It is expected that those approaches would result in improved therapeutic outcomes, even though many questions remain with regard to the optimal way to assemble treatments. Further trials are needed to evaluate and consolidate these promising therapies for HCC.
Collapse
|
10
|
Hassan SA, Schmithals C, von Harten M, Piiper A, Korf HW, von Gall C. Time-dependent changes in proliferation, DNA damage and clock gene expression in hepatocellular carcinoma and healthy liver of a transgenic mouse model. Int J Cancer 2020; 148:226-237. [PMID: 32700769 DOI: 10.1002/ijc.33228] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/03/2020] [Accepted: 07/13/2020] [Indexed: 12/16/2022]
Abstract
Hepatocellular carcinoma (HCC) is highly resistant to anticancer therapy and novel therapeutic strategies are needed. Chronotherapy may become a promising approach because it may improve the efficacy of antimitotic radiation and chemotherapy by considering timing of treatment. To date little is known about time-of-day dependent changes of proliferation and DNA damage in HCC. Using transgenic c-myc/transforming growth factor (TGFα) mice as HCC animal model, we immunohistochemically demonstrated Ki67 as marker for proliferation and γ-H2AX as marker for DNA damage in HCC and surrounding healthy liver (HL). Core clock genes (Per1, Per2, Cry1, Cry2, Bmal 1, Rev-erbα and Clock) were examined by qPCR. Data were obtained from samples collected ex vivo at four different time points and from organotypic slice cultures (OSC). Significant differences were found between HCC and HL. In HCC, the number of Ki67 immunoreactive cells showed two peaks (ex vivo: ZT06 middle of day and ZT18 middle of night; OSC: CT04 and CT16). In ex vivo samples, the number of γ-H2AX positive cells in HCC peaked at ZT18 (middle of the night), while in OSC their number remained high during subjective day and night. In both HCC and HL, clock gene expression showed a time-of-day dependent expression ex vivo but no changes in OSC. The expression of Per2 and Cry1 was significantly lower in HCC than in HL. Our data support the concept of chronotherapy of HCC. OSC may become useful to test novel cancer therapies.
Collapse
Affiliation(s)
- Soha A Hassan
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.,Zoology Department, Faculty of Science, Suez University, Suez, Egypt
| | | | - Maike von Harten
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Albrecht Piiper
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Horst-Werner Korf
- Institute of Anatomy I, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.,Institute of Anatomy II, Goethe University, Frankfurt, Germany
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
11
|
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: 1.0] [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
|
12
|
Broad and Dynamic Diversification of Infectious Hepatitis C Virus in a Cell Culture Environment. J Virol 2020; 94:JVI.01856-19. [PMID: 31852791 DOI: 10.1128/jvi.01856-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
Previous studies documented that long-term hepatitis C virus (HCV) replication in human hepatoma Huh-7.5 cells resulted in viral fitness gain, expansion of the mutant spectrum, and several phenotypic alterations. In the present work, we show that mutational waves (changes in frequency of individual mutations) occurred continuously and became more prominent as the virus gained fitness. They were accompanied by an increasing proportion of heterogeneous genomic sites that affected 1 position in the initial HCV population and 19 and 69 positions at passages 100 and 200, respectively. Analysis of biological clones of HCV showed that these dynamic events affected infectious genomes, since part of the fluctuating mutations became incorporated into viable genomes. While 17 mutations were scored in 3 biological clones isolated from the initial population, the number reached 72 in 3 biological clones from the population at passage 200. Biological clones differed in their responses to antiviral inhibitors, indicating a phenotypic impact of viral dynamics. Thus, HCV adaptation to a specific constant environment (cell culture without external influences) broadens the mutant repertoire and does not focus the population toward a limited number of dominant genomes. A retrospective examination of mutant spectra of foot-and-mouth disease virus passaged in cell cultures suggests a parallel behavior here described for HCV. We propose that virus diversification in a constant environment has its basis in the availability of multiple alternative mutational pathways for fitness gain. This mechanism of broad diversification should also apply to other replicative systems characterized by high mutation rates and large population sizes.IMPORTANCE The study shows that extensive replication of an RNA virus in a constant biological environment does not limit exploration of sequence space and adaptive options. There was no convergence toward a restricted set of adapted genomes. Mutational waves and mutant spectrum broadening affected infectious genomes. Therefore, profound modifications of mutant spectrum composition and consensus sequence diversification are not exclusively dependent on environmental alterations or the intervention of population bottlenecks.
Collapse
|
13
|
Palomba F, Genovese D, Rampazzo E, Zaccheroni N, Prodi L, Morbidelli L. PluS Nanoparticles Loaded with Sorafenib: Synthetic Approach and Their Effects on Endothelial Cells. ACS OMEGA 2019; 4:13962-13971. [PMID: 31497714 PMCID: PMC6714606 DOI: 10.1021/acsomega.9b01699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/18/2019] [Indexed: 05/24/2023]
Abstract
Silica nanostructures are widely investigated for theranostic applications since relatively mild and easy synthetic methods allow the fabrication of multicompartment nanoparticles (NPs) and fine modulation of their properties. Here, we report the optimization of a synthetic strategy leading to brightly fluorescent silica NPs with a high loading ability, up to 45 molecules per NP, of Sorafenib, a small molecule acting as an antiangiogenic drug. We demonstrate that these NPs can efficiently release the drug and they are able to inhibit endothelial cell proliferation and migration and network formation. Their lyophilization can endow them with long shelf stability, whereas, once in solution, they show a much slower release compared to analogous micellar systems. Interestingly, Sorafenib released from Pluronic silica NPs completely prevented endothelial cell responses and postreceptor mitogen-activated protein kinase signaling ignited by vascular endothelial growth factor, one of the major players of tumor angiogenesis. Our results indicate that these theranostic systems represent a promising structure for anticancer applications since NPs alone have no cytotoxic effect on cultured endothelial cells, a cell type to which drugs and exogenous material are always in contact once delivered.
Collapse
Affiliation(s)
- Francesco Palomba
- Dipartimento
di Chimica “Giacomo Ciamician”, Alma Mater Studiorum, Università di Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Damiano Genovese
- Dipartimento
di Chimica “Giacomo Ciamician”, Alma Mater Studiorum, Università di Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Enrico Rampazzo
- Dipartimento
di Chimica “Giacomo Ciamician”, Alma Mater Studiorum, Università di Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Nelsi Zaccheroni
- Dipartimento
di Chimica “Giacomo Ciamician”, Alma Mater Studiorum, Università di Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Luca Prodi
- Dipartimento
di Chimica “Giacomo Ciamician”, Alma Mater Studiorum, Università di Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Lucia Morbidelli
- Dipartimento
di Scienze della Vita, Università
di Siena, Via A. Moro
2, 53100 Siena, Italy
| |
Collapse
|
14
|
Castro V, Ávila-Pérez G, Mingorance L, Gastaminza P. A Cell Culture Model for Persistent HCV Infection. Methods Mol Biol 2019; 1911:157-168. [PMID: 30593624 DOI: 10.1007/978-1-4939-8976-8_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chronic hepatitis C virus (HCV) infection affects millions of humans throughout the globe, causing liver disease and hepatocellular carcinoma. Persistence of the virus in the infected host can last for decades as a result of a faulty immune response that fails to clear the virus while constituting a major player in viral pathogenesis. In addition to evading immune responses, HCV has evolved intracellular survival strategies that enable persistent replication without directly killing the host cell.After the generation of cell culture infection models for HCV, the knowledge about this virus and host-virus interactions acquired in the last decade has been greatly increased. Interestingly, persistent infection can also be established in cell culture. This model recapitulates persistent HCV RNA replication and viral protein expression as well as infectious progeny virus assembly and secretion and may be used to study not only these aspects of the virus replication cycle but also to study host-virus interactions in a model of prolonged HCV infection. In this chapter, we describe a methodology to generate persistently HCV-infected cultures and to monitor viral load and progeny virus production. Also, we provide generic protocols to study the impact of chemical compounds and host-targeting shRNAs to illustrate the applications of this model in the study of HCV infection in cell culture.
Collapse
Affiliation(s)
- Victoria Castro
- Department of Molecular and Cellular Biology, Centro Nacional De Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Ginés Ávila-Pérez
- Department of Molecular and Cellular Biology, Centro Nacional De Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Lidia Mingorance
- Department of Molecular and Cellular Biology, Centro Nacional De Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Pablo Gastaminza
- Department of Molecular and Cellular Biology, Centro Nacional De Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain.
| |
Collapse
|
15
|
Bahman AA, Abaza MSI, Khoushiash SI, Al-Attiyah RJ. Sequence‑dependent effect of sorafenib in combination with natural phenolic compounds on hepatic cancer cells and the possible mechanism of action. Int J Mol Med 2018; 42:1695-1715. [PMID: 29901131 PMCID: PMC6089756 DOI: 10.3892/ijmm.2018.3725] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 05/21/2018] [Indexed: 12/19/2022] Open
Abstract
Sorafenib (Nexavar, BAY43-9006 or Sora) is the first molecular targeted agent that has exhibited significant therapeutic benefits in advanced hepatocellular carcinoma (HCC). However, not all HCC patients respond well to Sora and novel therapeutic strategies to optimize the efficacy of Sora are urgently required. Plant-based drugs have received increasing attention owing to their excellent chemotherapeutic and chemopreventive activities; they are also well tolerated, non-toxic, easily available and inexpensive. It is well known that certain biologically active natural products act synergistically with synthetic drugs used in clinical applications. The present study aimed to investigate whether a combination therapy with natural phenolic compounds (NPCs), including curcumin (Cur), quercetin (Que), kaempherol (Kmf) and resveratrol (Rsv), would allow a dose reduction of Sora without concomitant loss of its effectiveness. Furthermore, the possible molecular mechanisms of this synergy were assessed. The hepatic cancer cell lines Hep3b and HepG2 were treated with Sora alone or in combination with NPCs in concomitant, sequential, and inverted sequential regimens. Cell proliferation, cell cycle, apoptosis and expression of proteins associated with the cell cycle and apoptosis were investigated. NPCs markedly potentiated the therapeutic efficacy of Sora in a sequence-, type-, NPC dose- and cell line-dependent manner. Concomitant treatment with Sora and Cur [sensitization ratio (SR)=28], Kmf (SR=18) or Que (SR=8) was associated with the highest SRs in Hep3b cells. Rsv markedly potentiated the effect of Sora (SR=17) on Hep3b cells when administered in a reverse sequential manner. By contrast, Rsv and Que did not improve the efficacy of Sora against HepG2 cells, while concomitant treatment with Cur (SR=10) or Kmf (SR=4.01) potentiated the cytotoxicity of Sora. Concomitant treatment with Sora and Cur or Kmf caused S-phase and G2/M phase arrest of liver cancer cells and markedly induced apoptosis compared with mono-treatment with Sora, Cur or Kmf. Concomitant treatment with Sora and Cur reduced the protein levels of cyclins A, B2 and D1, phosphorylated retinoblastoma and B-cell lymphoma (Bcl) extra-large protein. By contrast, Sora and Cur co-treatment increased the protein levels of Bcl-2-associated X protein, cleaved caspase-3 and cleaved caspase-9 in a dose-dependent manner. In conclusion, concomitant treatment with Sora and Cur or Kmf appears to be a potent and promising therapeutic approach that may control hepatic cancer by triggering cell cycle arrest and apoptosis. Additional studies are required to examine the potential of combined treatment with Sora and NPCs in human hepatic cancer and other solid tumor types in vivo.
Collapse
Affiliation(s)
- Abdulmajeed A Bahman
- Molecular Biology Program, Department of Biological Sciences, Faculty of Science, Kuwait University, 13060 Safat, State of Kuwait
| | - Mohamed Salah I Abaza
- Molecular Biology Program, Department of Biological Sciences, Faculty of Science, Kuwait University, 13060 Safat, State of Kuwait
| | - Sarah I Khoushiash
- Molecular Biology Program, Department of Biological Sciences, Faculty of Science, Kuwait University, 13060 Safat, State of Kuwait
| | - Rajaa J Al-Attiyah
- Department of Microbiology and Immunology, Faculty of Medicine, Kuwait University, 13060 Safat, State of Kuwait
| |
Collapse
|
16
|
Wang F, Bank T, Malnassy G, Arteaga M, Shang N, Dalheim A, Ding X, Cotler SJ, Denning MF, Nishimura MI, Breslin P, Qiu W. Inhibition of insulin-like growth factor 1 receptor enhances the efficacy of sorafenib in inhibiting hepatocellular carcinoma cell growth and survival. Hepatol Commun 2018; 2:732-746. [PMID: 29881824 PMCID: PMC5983153 DOI: 10.1002/hep4.1181] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/01/2018] [Accepted: 03/08/2018] [Indexed: 01/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common primary cancer and second largest cause of cancer-related death worldwide. The first-line oral chemotherapeutic agent sorafenib only increases survival in patients with advanced HCC by less than 3 months. Most patients with advanced HCC have shown limited response rates and survival benefits with sorafenib. Although sorafenib is an inhibitor of multiple kinases, including serine/threonine-protein kinase c-Raf, serine/threonine-protein kinase B-Raf, vascular endothelial growth factor receptor (VEGFR)-1, VEGFR-2, VEGFR-3, and platelet-derived growth factor receptor β, HCC cells are able to escape from sorafenib treatment using other pathways that the drug insufficiently inhibits. The aim of this study was to identify and target survival and proliferation pathways that enable HCC to escape the antitumor activity of sorafenib. We found that insulin-like growth factor 1 receptor (IGF1R) remains activated in HCC cells treated with sorafenib. Knockdown of IGF1R sensitizes HCC cells to sorafenib treatment and decreases protein kinase B (AKT) activation. Overexpression of constitutively activated AKT reverses the effect of knockdown of IGF1R in sensitizing HCC cells to treatment with sorafenib. Further, we found that ceritinib, a drug approved by the U.S. Food and Drug Administration for treatment of non-small cell lung cancer, effectively inhibits the IGF1R/AKT pathway and enhances the inhibitory efficacy of sorafenib in human HCC cell growth and survival in vitro, in a xenograft mouse model and in the c-Met/β-catenin-driven HCC mouse model. Conclusion: Our study provides a biochemical basis for evaluation of a new combination treatment that includes IGF1R inhibitors, such as ceritinib and sorafenib, in patients with HCC. (Hepatology Communications 2018;2:732-746).
Collapse
Affiliation(s)
- Fang Wang
- Department of Surgery and Oncology Institute, Stritch School of MedicineLoyola University ChicagoMaywoodIL
| | - Thomas Bank
- Department of Surgery and Oncology Institute, Stritch School of MedicineLoyola University ChicagoMaywoodIL
| | - Gregory Malnassy
- Department of Surgery and Oncology Institute, Stritch School of MedicineLoyola University ChicagoMaywoodIL
| | - Maribel Arteaga
- Department of Surgery and Oncology Institute, Stritch School of MedicineLoyola University ChicagoMaywoodIL
| | - Na Shang
- Department of Surgery and Oncology Institute, Stritch School of MedicineLoyola University ChicagoMaywoodIL
| | - Annika Dalheim
- Department of Surgery and Oncology Institute, Stritch School of MedicineLoyola University ChicagoMaywoodIL
| | - Xianzhong Ding
- Pathology Department, Stritch School of MedicineLoyola University ChicagoMaywoodIL
| | - Scott J. Cotler
- Department of Medicine, Stritch School of MedicineLoyola University ChicagoMaywoodIL
| | - Mitchell F. Denning
- Pathology Department, Stritch School of MedicineLoyola University ChicagoMaywoodIL
| | - Michael I. Nishimura
- Department of Surgery and Oncology Institute, Stritch School of MedicineLoyola University ChicagoMaywoodIL
| | - Peter Breslin
- Department of Surgery and Oncology Institute, Stritch School of MedicineLoyola University ChicagoMaywoodIL
- Department of Molecular/Cellular Physiology, Stritch School of MedicineLoyola University ChicagoMaywoodIL
| | - Wei Qiu
- Department of Surgery and Oncology Institute, Stritch School of MedicineLoyola University ChicagoMaywoodIL
| |
Collapse
|
17
|
Deng M, Li L, Zhao J, Yuan S, Li W. Antitumor activity of the microtubule inhibitor MBRI-001 against human hepatocellular carcinoma as monotherapy or in combination with sorafenib. Cancer Chemother Pharmacol 2018. [PMID: 29532153 DOI: 10.1007/s00280-018-3547-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE MBRI-001 is a novel synthetic derivative of plinabulin. In this study, our purpose is to investigate the inhibition effects of MBRI-001 on human hepatocellular carcinoma as monotherapy or in combination with sorafenib. METHODS HCCLM3 and Bel-7402 cell lines were used for activity evaluation in vitro. The anti-proliferative activity of MBRI-001 was assessed by MTT assay. The morphological change of microtubules was determined by immunofluorescence assay. The cell cycle was measured by flow cytometer. The expression of cyclin B1 (CCNB1) was analyzed by RT-qPCR and western blotting assays. The antitumor activities in vivo were evaluated with human HCC xenograft mice model. RESULTS Our data demonstrated that MBRI-001 had better anti-proliferative activities than that of plinabulin against HCCLM3 and Bel-7402 cell lines. MBRI-001 inhibited the formation of microtubules and induced G2/M arrest with the downregulation of CCNB1. In vivo orthotopic mice model demonstrated that MBRI-001 significantly inhibited the growth of HCCLM3 with the apoptosis and necrosis observed in tumor. The combination treatment of MBRI-001 with sorafenib in subcutaneous mice model exhibited a higher antitumor inhibition rate at 72.0%, in comparison with MBRI-001 or sorafenib as monotherapy at 40.7% or 47.7%, respectively. CONCLUSION MBRI-001 had better inhibition effects on microtubules and human hepatocellular carcinoma than that of plinabulin. The combination treatment of MBRI-001 and sorafenib exhibited a higher antitumor effect, which could provide a new strategy to treat HCC in the future.
Collapse
Affiliation(s)
- Mengyan Deng
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Linna Li
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jianchun Zhao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China.,Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Shoujun Yuan
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Wenbao Li
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China. .,Innovation Center for Marine Drug Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China. .,Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China.
| |
Collapse
|
18
|
Chen JCH, Chuang HY, Hsu FT, Chen YC, Chien YC, Hwang JJ. Sorafenib pretreatment enhances radiotherapy through targeting MEK/ERK/NF-κB pathway in human hepatocellular carcinoma-bearing mouse model. Oncotarget 2018; 7:85450-85463. [PMID: 27863427 PMCID: PMC5356748 DOI: 10.18632/oncotarget.13398] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 10/28/2016] [Indexed: 12/11/2022] Open
Abstract
Patients with unresectable hepatocellular carcinoma (HCC) usually have poor prognosis because current monotherapy including surgery, chemotherapy and radiotherapy (RT) are not effective. Combination therapy may be effective to overcome this clinical problem. Here, we proposed the combination of sorafenib and RT, which have been applied in HCC treatment, could improve the treatment outcome of HCC. Our previous study showed that sorafenib could suppress the expression of NF-κB which is related to the chemo- and radio-resistance. Nevertheless, the expression of NF-κB is oscillatory and is affected by the treatments. Thus, understanding the oscillation of NF-κB expression would be beneficial for determining the optimal treatment schedule in combination therapy. Here established Huh7/NF-κB-tk-luc2/rfp cell line, in which NF-κB indicates a NF-κB promoter, was utilized to noninvasively monitor the expression of NF-κB overtime in vitro and in vivo. The results show that pretreatment of sorafenib with RT suppresses the expressions of NF-κB and its downstream proteins induced by radiation through downregulation of phosphorylated extracellular signal-regulated kinase (pERK) most significantly compared with other treatment schedules. The results were further verified with Western blotting, EMSA, and NF-κB molecular imaging. These findings suggest that pretreatment of sorafenib with RT may be the ideal treatment schedule for the treatment of HCC.
Collapse
Affiliation(s)
- John Chun-Hao Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Department of Radiation Oncology, Mackay Memorial Hospital, New Taipei City, Taiwan
| | - Hui-Yen Chuang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Fei-Ting Hsu
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Chen Chen
- Department of Radiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Chun Chien
- Department of Medical Imaging and Radiological Sciences, I-Shou University, Jiaosu Village, Kaohsiung, Taiwan
| | - Jeng-Jong Hwang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Biophotonics and Molecular Imaging Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
19
|
Dinavahi SS, Noory MA, Gowda R, Drabick JJ, Berg A, Neves RI, Robertson GP. Moving Synergistically Acting Drug Combinations to the Clinic by Comparing Sequential versus Simultaneous Drug Administrations. Mol Pharmacol 2017; 93:190-196. [PMID: 29242354 DOI: 10.1124/mol.117.110759] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/12/2017] [Indexed: 01/21/2023] Open
Abstract
Drug combinations acting synergistically to kill cancer cells have become increasingly important in melanoma as an approach to manage the recurrent resistant disease. Protein kinase B (AKT) is a major target in this disease but its inhibitors are not effective clinically, which is a major concern. Targeting AKT in combination with WEE1 (mitotic inhibitor kinase) seems to have potential to make AKT-based therapeutics effective clinically. Since agents targeting AKT and WEE1 have been tested individually in the clinic, the quickest way to move the drug combination to patients would be to combine these agents sequentially, enabling the use of existing phase I clinical trial toxicity data. Therefore, a rapid preclinical approach is needed to evaluate whether simultaneous or sequential drug treatment has maximal therapeutic efficacy, which is based on a mechanistic rationale. To develop this approach, melanoma cell lines were treated with AKT inhibitor AZD5363 [4-amino-N-[(1S)-1-(4-chlorophenyl)-3-hydroxypropyl]-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidine-4-carboxamide] and WEE1 inhibitor AZD1775 [2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((4-(4-methylpiperazin-1-yl)phenyl)amino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one] using simultaneous and sequential dosing schedules. Simultaneous treatment synergistically reduced melanoma cell survival and tumor growth. In contrast, sequential treatment was antagonistic and had a minimal tumor inhibitory effect compared with individual agents. Mechanistically, simultaneous targeting of AKT and WEE1 enhanced deregulation of the cell cycle and DNA damage repair pathways by modulating transcription factors p53 and forkhead box M1, which was not observed with sequential treatment. Thus, this study identifies a rapid approach to assess the drug combinations with a mechanistic basis for selection, which suggests that combining AKT and WEE1 inhibitors is needed for maximal efficacy.
Collapse
Affiliation(s)
- Saketh S Dinavahi
- Division of Hematology-Oncology (J.J.D.); Departments of Pharmacology (S.S.D., M.A.N., R.G., R.I.N., G.P.R.), Medicine (J.J.D.), Public Health Sciences (A.B.), Dermatology (R.I.N., G.P.R.), Surgery (R.I.N., G.P.R.), and Pathology (G.P.R.); Melanoma and Skin Cancer Center (S.S.D., M.A.N., R.G., J.J.D., A.B., R.I.N., G.P.R.); Foreman Foundation for Melanoma Research (R.G., G.P.R.); and the Melanoma Therapeutics Program (R.G., R.I.N., G.P.R.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Mohammad A Noory
- Division of Hematology-Oncology (J.J.D.); Departments of Pharmacology (S.S.D., M.A.N., R.G., R.I.N., G.P.R.), Medicine (J.J.D.), Public Health Sciences (A.B.), Dermatology (R.I.N., G.P.R.), Surgery (R.I.N., G.P.R.), and Pathology (G.P.R.); Melanoma and Skin Cancer Center (S.S.D., M.A.N., R.G., J.J.D., A.B., R.I.N., G.P.R.); Foreman Foundation for Melanoma Research (R.G., G.P.R.); and the Melanoma Therapeutics Program (R.G., R.I.N., G.P.R.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Raghavendra Gowda
- Division of Hematology-Oncology (J.J.D.); Departments of Pharmacology (S.S.D., M.A.N., R.G., R.I.N., G.P.R.), Medicine (J.J.D.), Public Health Sciences (A.B.), Dermatology (R.I.N., G.P.R.), Surgery (R.I.N., G.P.R.), and Pathology (G.P.R.); Melanoma and Skin Cancer Center (S.S.D., M.A.N., R.G., J.J.D., A.B., R.I.N., G.P.R.); Foreman Foundation for Melanoma Research (R.G., G.P.R.); and the Melanoma Therapeutics Program (R.G., R.I.N., G.P.R.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Joseph J Drabick
- Division of Hematology-Oncology (J.J.D.); Departments of Pharmacology (S.S.D., M.A.N., R.G., R.I.N., G.P.R.), Medicine (J.J.D.), Public Health Sciences (A.B.), Dermatology (R.I.N., G.P.R.), Surgery (R.I.N., G.P.R.), and Pathology (G.P.R.); Melanoma and Skin Cancer Center (S.S.D., M.A.N., R.G., J.J.D., A.B., R.I.N., G.P.R.); Foreman Foundation for Melanoma Research (R.G., G.P.R.); and the Melanoma Therapeutics Program (R.G., R.I.N., G.P.R.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Arthur Berg
- Division of Hematology-Oncology (J.J.D.); Departments of Pharmacology (S.S.D., M.A.N., R.G., R.I.N., G.P.R.), Medicine (J.J.D.), Public Health Sciences (A.B.), Dermatology (R.I.N., G.P.R.), Surgery (R.I.N., G.P.R.), and Pathology (G.P.R.); Melanoma and Skin Cancer Center (S.S.D., M.A.N., R.G., J.J.D., A.B., R.I.N., G.P.R.); Foreman Foundation for Melanoma Research (R.G., G.P.R.); and the Melanoma Therapeutics Program (R.G., R.I.N., G.P.R.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Rogerio I Neves
- Division of Hematology-Oncology (J.J.D.); Departments of Pharmacology (S.S.D., M.A.N., R.G., R.I.N., G.P.R.), Medicine (J.J.D.), Public Health Sciences (A.B.), Dermatology (R.I.N., G.P.R.), Surgery (R.I.N., G.P.R.), and Pathology (G.P.R.); Melanoma and Skin Cancer Center (S.S.D., M.A.N., R.G., J.J.D., A.B., R.I.N., G.P.R.); Foreman Foundation for Melanoma Research (R.G., G.P.R.); and the Melanoma Therapeutics Program (R.G., R.I.N., G.P.R.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Gavin P Robertson
- Division of Hematology-Oncology (J.J.D.); Departments of Pharmacology (S.S.D., M.A.N., R.G., R.I.N., G.P.R.), Medicine (J.J.D.), Public Health Sciences (A.B.), Dermatology (R.I.N., G.P.R.), Surgery (R.I.N., G.P.R.), and Pathology (G.P.R.); Melanoma and Skin Cancer Center (S.S.D., M.A.N., R.G., J.J.D., A.B., R.I.N., G.P.R.); Foreman Foundation for Melanoma Research (R.G., G.P.R.); and the Melanoma Therapeutics Program (R.G., R.I.N., G.P.R.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
20
|
Chettiar ST, Malek R, Annadanam A, Nugent KM, Kato Y, Wang H, Cades JA, Taparra K, Belcaid Z, Ballew M, Manmiller S, Proia D, Lim M, Anders RA, Herman JM, Tran PT. Ganetespib radiosensitization for liver cancer therapy. Cancer Biol Ther 2017; 17:457-66. [PMID: 26980196 PMCID: PMC4910914 DOI: 10.1080/15384047.2016.1156258] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Therapies for liver cancer particularly those including radiation are still inadequate. Inhibiting the stress response machinery is an appealing anti-cancer and radiosensitizing therapeutic strategy. Heat-shock-protein-90 (HSP90) is a molecular chaperone that is a prominent effector of the stress response machinery and is overexpressed in liver cancer cells. HSP90 client proteins include critical components of pathways implicated in liver cancer cell survival and radioresistance. The effects of a novel non-geldanamycin HSP90 inhibitor, ganetespib, combined with radiation were examined on 3 liver cancer cell lines, Hep3b, HepG2 and HUH7, using in vitro assays for clonogenic survival, apoptosis, cell cycle distribution, γH2AX foci kinetics and client protein expression in pathways important for liver cancer survival and radioresistance. We then evaluated tumor growth delay and effects of the combined ganetespib-radiation treatment on tumor cell proliferation in a HepG2 hind-flank tumor graft model. Nanomolar levels of ganetespib alone exhibited liver cancer cell anti-cancer activity in vitro as shown by decreased clonogenic survival that was associated with increased apoptotic cell death, prominent G2-M arrest and marked changes in PI3K/AKT/mTOR and RAS/MAPK client protein activity. Ganetespib caused a supra-additive radiosensitization in all liver cancer cell lines at low nanomolar doses with enhancement ratios between 1.33–1.78. These results were confirmed in vivo, where the ganetespib-radiation combination therapy produced supra-additive tumor growth delay compared with either therapy by itself in HepG2 tumor grafts. Our data suggest that combined ganetespib-radiation therapy exhibits promising activity against liver cancer cells, which should be investigated in clinical studies.
Collapse
Affiliation(s)
- Sivarajan T Chettiar
- a Department of Radiation Oncology and Molecular Radiation Sciences , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Reem Malek
- a Department of Radiation Oncology and Molecular Radiation Sciences , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Anvesh Annadanam
- a Department of Radiation Oncology and Molecular Radiation Sciences , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Katriana M Nugent
- a Department of Radiation Oncology and Molecular Radiation Sciences , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Yoshinori Kato
- b The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, Johns Hopkins University School of Medicine , Baltimore , MD , USA.,c Department of Oncology , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Hailun Wang
- a Department of Radiation Oncology and Molecular Radiation Sciences , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Jessica A Cades
- a Department of Radiation Oncology and Molecular Radiation Sciences , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Kekoa Taparra
- a Department of Radiation Oncology and Molecular Radiation Sciences , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA.,d Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Zineb Belcaid
- e Department of Neurosurgery , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Matthew Ballew
- a Department of Radiation Oncology and Molecular Radiation Sciences , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Sarah Manmiller
- a Department of Radiation Oncology and Molecular Radiation Sciences , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - David Proia
- f Synta Pharmaceuticals Corp. , Lexington , MD , USA
| | - Michael Lim
- c Department of Oncology , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA.,e Department of Neurosurgery , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Robert A Anders
- g Department of Pathology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Joseph M Herman
- a Department of Radiation Oncology and Molecular Radiation Sciences , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA.,c Department of Oncology , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Phuoc T Tran
- a Department of Radiation Oncology and Molecular Radiation Sciences , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA.,c Department of Oncology , Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore , MD , USA.,d Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine , Baltimore , MD , USA.,h Department of Urology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| |
Collapse
|
21
|
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
|
22
|
Internal Disequilibria and Phenotypic Diversification during Replication of Hepatitis C Virus in a Noncoevolving Cellular Environment. J Virol 2017; 91:JVI.02505-16. [PMID: 28275194 DOI: 10.1128/jvi.02505-16] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 02/28/2017] [Indexed: 12/14/2022] Open
Abstract
Viral quasispecies evolution upon long-term virus replication in a noncoevolving cellular environment raises relevant general issues, such as the attainment of population equilibrium, compliance with the molecular-clock hypothesis, or stability of the phenotypic profile. Here, we evaluate the adaptation, mutant spectrum dynamics, and phenotypic diversification of hepatitis C virus (HCV) in the course of 200 passages in human hepatoma cells in an experimental design that precluded coevolution of the cells with the virus. Adaptation to the cells was evidenced by increase in progeny production. The rate of accumulation of mutations in the genomic consensus sequence deviated slightly from linearity, and mutant spectrum analyses revealed a complex dynamic of mutational waves, which was sustained beyond passage 100. The virus underwent several phenotypic changes, some of which impacted the virus-host relationship, such as enhanced cell killing, a shift toward higher virion density, and increased shutoff of host cell protein synthesis. Fluctuations in progeny production and failure to reach population equilibrium at the genomic level suggest internal instabilities that anticipate an unpredictable HCV evolution in the complex liver environment.IMPORTANCE Long-term virus evolution in an unperturbed cellular environment can reveal features of virus evolution that cannot be explained by comparing natural viral isolates. In the present study, we investigate genetic and phenotypic changes that occur upon prolonged passage of hepatitis C virus (HCV) in human hepatoma cells in an experimental design in which host cell evolutionary change is prevented. Despite replication in a noncoevolving cellular environment, the virus exhibited internal population disequilibria that did not decline with increased adaptation to the host cells. The diversification of phenotypic traits suggests that disequilibria inherent to viral populations may provide a selective advantage to viruses that can be fully exploited in changing environments.
Collapse
|
23
|
Yan Y, Wang L, He J, Liu P, Lv X, Zhang Y, Xu X, Zhang L, Zhang Y. Synergy with interferon-lambda 3 and sorafenib suppresses hepatocellular carcinoma proliferation. Biomed Pharmacother 2017; 88:395-402. [PMID: 28122304 DOI: 10.1016/j.biopha.2017.01.077] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common and fatal malignancy of the liver. Sorafenib is a small molecule multikinase inhibitor that acts against different cancer cell lines and is used for the treatment of HCC. However, some advanced HCC patients fail to respond to sorafenib, and those who do lack a meaningful clinical benefit. Interferon-lambda 3 (IFN-λ3) is a type III interferon with antiviral, antiproliferative, and immunomodulatory functions. Here, we evaluated the use of IFN-λ3 as an adjuvant treatment with sorafenib in HCC. In the present study, CCK-8 and colony formation assay results showed that treatment with a combination of IFN-λ3 and sorafenib suppresses the viability of HepG2 and SMMC7721 liver cancer cell lines more than treatment with either alone. In addition, flow cytometry results confirmed that treatment with a combination of IFN-λ3 and sorafenib promotes the loss of mitochondrial membrane potential and induces the production of ROS more than treatment with either alone. Furthermore, using a subcutaneous SMMC7721 tumor model, treatment with a combination of IFN-λ3 and sorafenib significantly reduced the tumor growth/volume and induced apoptosis compared to treatment with sorafenib alone. These results show that combined treatment with IFN-λ3 and sorafenib facilitates a synergistic effect on suppressing HCC cancer growth and promoting cell apoptosis in vitro and in vivo. Thus, IFN-λ3 in combination with sorafenib might prove to be a useful adjunctive strategy for the clinical treatment of HCC.
Collapse
Affiliation(s)
- Yuke Yan
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Hepato-Biliary-Pancreatic Institute, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Gansu Provincial-Level Key Laboratory of Digestive System Tumors, Lanzhou 730030, PR China
| | - Liang Wang
- Hepato-Biliary-Pancreatic Institute, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Gansu Provincial-Level Key Laboratory of Digestive System Tumors, Lanzhou 730030, PR China; Division of Liver Disease, Lanzhou University Second Hospital, Lanzhou 730030, PR China
| | - Jingjing He
- Hepato-Biliary-Pancreatic Institute, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Gansu Provincial-Level Key Laboratory of Digestive System Tumors, Lanzhou 730030, PR China; Division of Liver Disease, Lanzhou University Second Hospital, Lanzhou 730030, PR China
| | - Pengcheng Liu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Hepato-Biliary-Pancreatic Institute, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Gansu Provincial-Level Key Laboratory of Digestive System Tumors, Lanzhou 730030, PR China
| | - Xi Lv
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Hepato-Biliary-Pancreatic Institute, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Gansu Provincial-Level Key Laboratory of Digestive System Tumors, Lanzhou 730030, PR China
| | - Yawu Zhang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Hepato-Biliary-Pancreatic Institute, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Gansu Provincial-Level Key Laboratory of Digestive System Tumors, Lanzhou 730030, PR China
| | - Xiaodong Xu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Hepato-Biliary-Pancreatic Institute, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Gansu Provincial-Level Key Laboratory of Digestive System Tumors, Lanzhou 730030, PR China
| | - Lingyi Zhang
- Hepato-Biliary-Pancreatic Institute, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Gansu Provincial-Level Key Laboratory of Digestive System Tumors, Lanzhou 730030, PR China; Division of Liver Disease, Lanzhou University Second Hospital, Lanzhou 730030, PR China
| | - Youcheng Zhang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Hepato-Biliary-Pancreatic Institute, Lanzhou University Second Hospital, Lanzhou 730030, PR China; Gansu Provincial-Level Key Laboratory of Digestive System Tumors, Lanzhou 730030, PR China.
| |
Collapse
|
24
|
Abstract
Hepatocellular cancer (HCC) is a leading cause of cancer death worldwide, and most patients who are diagnosed with HCC are ineligible for curative local therapy. The targeted agent sorafenib provides modest survival benefits in the setting of advanced disease. Novel systemic treatment options for HCC are sorely needed. In this review, we identify and categorize the drugs and targets that are in various phases of testing for use against HCC. We also focus on the potential for combining these agents with radiotherapy. This would help identify directions for future study that are likely to yield positive findings and improve outcomes for patients with HCC.
Collapse
Affiliation(s)
- Nitin Ohri
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY
| | - Andreas Kaubisch
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY
| | - Madhur Garg
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY
| | - Chandan Guha
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
25
|
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: 37] [Impact Index Per Article: 4.1] [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
|
26
|
Yamada T, Abei M, Danjoh I, Shirota R, Yamashita T, Hyodo I, Nakamura Y. Identification of a unique hepatocellular carcinoma line, Li-7, with CD13(+) cancer stem cells hierarchy and population change upon its differentiation during culture and effects of sorafenib. BMC Cancer 2015; 15:260. [PMID: 25885470 PMCID: PMC4396571 DOI: 10.1186/s12885-015-1297-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 03/31/2015] [Indexed: 12/26/2022] Open
Abstract
Backgrounds Cancer stem cell (CSC) research has highlighted the necessity of developing drugs targeting CSCs. We investigated a hepatocellular carcinoma (HCC) cell line that not only has CSC hierarchy but also shows phenotypic changes (population changes) upon differentiation of CSC during culture and can be used for screening drugs targeting CSC. Methods Based on a hypothesis that the CSC proportion should decrease upon its differentiation into progenitors (population change), we tested HCC cell lines (HuH-7, Li-7, PLC/PRF/5, HLF, HLE) before and after 2 months culture for several markers (CD13, EpCAM, CD133, CD44, CD90, CD24, CD166). Tumorigenicity was tested using nude mice. To evaluate the CSC hierarchy, we investigated reconstructivity, proliferation, ALDH activity, spheroid formation, chemosensitivity and microarray analysis of the cell populations sorted by FACS. Results Only Li-7 cells showed a population change during culture: the proportion of CD13 positive cells decreased, while that of CD166 positive cells increased. The high tumorigenicity of the Li-7 was lost after the population change. CD13(+)/CD166(−) cells showed slow growth and reconstructed the bulk Li-7 populations composed of CD13(+)/CD166(−), CD13(−)/CD166(−) and CD13(−)/CD166(+) fractions, whereas CD13(−)/CD166(+) cells showed rapid growth but could not reproduce any other population. CD13(+)/CD166(−) cells showed high ALDH activity, spheroid forming ability and resistance to 5-fluorouracil. Microarray analysis demonstrated higher expression of stemness-related genes in CD166(−) than CD166(+) fraction. These results indicated a hierarchy in Li-7 cells, in which CD13(+)/CD166(−) and CD13(−)/CD166(+) cells serve as slow growing CSCs and rapid growing progenitors, respectively. Sorafenib selectively targeted the CD166(−) fraction, including CD13(+) CSCs, which exhibited higher mRNA expression for FGF3 and FGF4, candidate biomarkers for sorafenib. 5-fluorouracil followed by sorafenib inhibited the growth of bulk Li-7 cells more effectively than the reverse sequence or either alone. Conclusions We identified a unique HCC line, Li-7, which not only shows heterogeneity for a CD13(+) CSC hierarchy, but also undergoes a “population change” upon CSC differentiation. Sorafenib targeted the CSC in vitro, supporting the use of this model for screening drugs targeting the CSC. This type of “heterogeneous, unstable” cell line may prove more useful in the CSC era than conventional “homogeneous, stable” cell lines. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1297-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Takeshi Yamada
- Division of Gastroenterology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8575, Japan. .,Cell Engineering Division, RIKEN BioResource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan.
| | - Masato Abei
- Division of Gastroenterology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Inaho Danjoh
- Functional Evaluation of Genomic Polymorphisms, Tohoku Medical Megabank Organization, Sendai, Japan.
| | - Ryoko Shirota
- Cell Engineering Division, RIKEN BioResource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan.
| | - Taro Yamashita
- Department of Gastroenterology, Kanazawa University Hospital, 13-1, Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan.
| | - Ichinosuke Hyodo
- Division of Gastroenterology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan.
| |
Collapse
|
27
|
Kondo Y, Kimura O, Shimosegawa T. Radiation therapy has been shown to be adaptable for various stages of hepatocellular carcinoma. World J Gastroenterol 2015; 21:94-101. [PMID: 25574082 PMCID: PMC4284364 DOI: 10.3748/wjg.v21.i1.94] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/12/2014] [Accepted: 06/26/2014] [Indexed: 02/06/2023] Open
Abstract
In addition to surgical procedures, radiofrequency ablation is commonly used for the treatment of hepatocellular carcinomas (HCCs) of limited size and number. Transcatheter arterial chemoembolization (TACE), using iodized poppy seed oil, Lipiodol and anticancer drugs, has been actively performed for the treatment of unresectable HCC, particularly in Asian countries. Recently, Sorafenib become available for advanced HCCs when the liver is still sufficiently functional. Sorafenib is an oral multikinase inhibitor with antiproliferative and antiangiogenic effects. However, the effect of sorafenib seems to be inadequate to control the progression of HCC. Radiation therapy (RT) for HCC has a potential role across all stages of HCC. However, RT is generally not considered an option in HCC consensus documents or national guidelines, primarily because of insufficient supporting evidence. However, the method of RT has much improved because of advances in technology. Moreover, combined treatment of RT plus other treatments (TACE, sorafenib and chemotherapy etc.) has become one of the alternative therapies for HCC. Therefore, we should understand the various kinds of RT available for HCC. In this review, we focus on various kinds of external beam radiation therapy.
Collapse
|
28
|
Hsieh CH, Lin SC, Shueng PW, Kuo DY. Recall radiation dermatitis by sorafenib following stereotactic body radiation therapy. Onco Targets Ther 2014; 7:1111-4. [PMID: 24971021 PMCID: PMC4069149 DOI: 10.2147/ott.s64706] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We report on a 63-year-old man with a history of hepatitis B virus–related hepatocellular carcinoma with a thrombus extending into the inferior vena cava, who received image-guided stereotactic body radiation therapy (SBRT) with helical tomotherapy, followed by sorafenib. A total tumor dose of 48 Gy was delivered by 6 fractions within 2 weeks. The tumor responded dramatically, and the patient tolerated the courses well. Ten days after SBRT, sorafenib (200 mg), at 1.5 tablets twice a day, was prescribed. One week later, grade 2 recall radiation dermatitis subsequently developed in the previous SBRT off-target area. SBRT followed by sorafenib for the treatment of a portal vein thrombosis provided effective results, but the potential risk of enhanced adverse effects between radiation and sorafenib should be considered with caution, especially under a SBRT scheme.
Collapse
Affiliation(s)
- Chen-Hsi Hsieh
- Division of Radiation Oncology, Department of Radiology, New Taipei City, Taiwan ; Department of Medicine, National Yang-Ming University, Taipei, Taiwan ; Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Chiang Lin
- Division of Medical Oncology and Hematology, Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Pei-Wei Shueng
- Division of Radiation Oncology, Department of Radiology, New Taipei City, Taiwan ; Department of Radiation Oncology, National Defense Medical Center, Taipei, Taiwan ; Oriental Institute of Technology, New Taipei City, Taiwan
| | - Deng-Yu Kuo
- Division of Radiation Oncology, Department of Radiology, New Taipei City, Taiwan
| |
Collapse
|
29
|
Chen SW, Lin LC, Kuo YC, Liang JA, Kuo CC, Chiou JF. Phase 2 study of combined sorafenib and radiation therapy in patients with advanced hepatocellular carcinoma. Int J Radiat Oncol Biol Phys 2014; 88:1041-7. [PMID: 24661657 DOI: 10.1016/j.ijrobp.2014.01.017] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/04/2014] [Accepted: 01/13/2014] [Indexed: 12/11/2022]
Abstract
PURPOSE This phase 2 study evaluated the efficacy of radiation therapy (RT) with concurrent and sequential sorafenib therapy in patients with unresectable hepatocellular carcinoma (HCC). METHODS AND MATERIALS Forty patients with unresectable HCC unfit for transarterial chemoembolization were treated with RT with concurrent and sequential sorafenib. Sorafenib was administered from the commencement of RT at a dose of 400 mg twice daily and continued to clinical or radiologic progression, unacceptable adverse events, or death. All patients had underlying Child-Pugh A cirrhosis. The maximal tumor diameter ranged from 3.0 cm to 15.5 cm. Coexisting portal vein thrombosis was found in 24 patients and was irradiated simultaneously. The cumulative RT dose ranged from 40 Gy to 60 Gy (median, 50 Gy). Image studies were done 1 month after RT and then every 3 months thereafter. RESULTS Thirty-three (83%) completed the allocated RT. During RT, the incidence of hand-foot skin reactions ≥ grade 2 and diarrhea were 37.5% and 25%, respectively, and 35% of patients had hepatic toxicities grade ≥2. Twenty-two (55.0%) patients achieved complete or partial remission at the initial assessment, and 18 (45%) had stable or progressive disease. The 2-year overall survival and infield progression-free survival (IFPS) were 32% and 39%, respectively. A Cancer of the Liver Italian Program (CLIP) score ≥2 was associated with an inferior outcome in overall survival. Six patients (15%) developed treatment-related hepatic toxicity grade ≥3 during the sequential phase, and 3 of them were fatal. CONCLUSIONS When RT and sorafenib therapy were combined in patients with unresectable HCC, the initial complete or partial response rate was 55% with a 2-year IFPS of 39%. A CLIP score ≥2 was associated with an inferior outcome in overall survival. Hepatic toxicities are a major determinant of the safety; the combination should be used with caution and needs further investigation.
Collapse
Affiliation(s)
- Shang-Wen Chen
- Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan; School of Medicine, Taipei Medical University, Taipei, Taiwan; School of Medicine, China Medical University, Taichung, Taiwan.
| | - Li-Ching Lin
- Department of Radiation Oncology, Chi-Mei Hospital, Tainan, Taiwan; School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Cheng Kuo
- Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan; Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Ji-An Liang
- Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan; School of Medicine, China Medical University, Taichung, Taiwan
| | - Chia-Chun Kuo
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Jeng-Fong Chiou
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan; School of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
30
|
Fujiki M, Aucejo F, Choi M, Kim R. Neo-adjuvant therapy for hepatocellular carcinoma before liver transplantation: Where do we stand? World J Gastroenterol 2014; 20:5308-5319. [PMID: 24833861 PMCID: PMC4017046 DOI: 10.3748/wjg.v20.i18.5308] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 02/08/2014] [Accepted: 02/20/2014] [Indexed: 02/06/2023] Open
Abstract
Liver transplantation (LT) for hepatocellular carcinoma (HCC) within Milan criteria is a widely accepted optimal therapy. Neo-adjuvant therapy before transplantation has been used as a bridging therapy to prevent dropout during the waiting period and as a down-staging method for the patient with intermediate HCC to qualify for liver transplantation. Transarterial chemoembolization and radiofrequency ablation are the most commonly used method for locoregional therapy. The data associated with newer modalities including drug-eluting beads, radioembolization with Y90, stereotactic radiation therapy and sorafenib will be discussed as a tool for converting advanced HCC to LT candidates. The concept “ablate and wait” has gained the popularity where mandated observation period after neo-adjuvant therapy allows for tumor biology to become apparent, thus has been recommended after down-staging. The role of neo-adjuvant therapy with conjunction of “ablate and wait” in living donor liver transplantation for intermediate stage HCC is also discussed in the paper.
Collapse
|
31
|
Huang CY, Tai WT, Hsieh CY, Hsu WM, Lai YJ, Chen LJ, Shiau CW, Chen KF. A sorafenib derivative and novel SHP-1 agonist, SC-59, acts synergistically with radiotherapy in hepatocellular carcinoma cells through inhibition of STAT3. Cancer Lett 2014; 349:136-43. [PMID: 24735751 DOI: 10.1016/j.canlet.2014.04.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 03/31/2014] [Accepted: 04/07/2014] [Indexed: 12/17/2022]
Abstract
Radiotherapy shows limited benefit as treatment for hepatocellular carcinoma (HCC). In this study, we aimed to overcome the radioresistance of HCC by using a novel sorafenib derivative, SC-59 that targets SHP-1-related signaling. HCC cell lines (SK-Hep1, Hep3B, and Huh7) were treated with sorafenib, SC-59, radiation, sorafenib plus radiation, or SC-59 plus radiation, and then apoptosis, colony formation, signal transduction and the phosphatase activity were analyzed. The synergistic effect of radiotherapy and SC-59 was analyzed using a combination index (CI) approach. In vivo efficacy was determined in a Huh7-bearing subcutaneous model. Mice were treated with radiation (5 Gy, one fraction per day) for 4 days, SC-59 (10mg/kg/day) for 24 days, or a combination. Tumor samples were further analyzed for p-STAT3 and SHP-1 activity. SC-59 displayed a better synergistic effect when used in combination with radiotherapy than sorafenib in HCC cell lines. SC-59 downregulated p-STAT3 and its downstream targets and increased SHP-1 phosphatase activity. Both ectopic STAT3 and inhibition of SHP-1 abolished SC-59-induced radiosensitization. Moreover, SC-59 significantly synergized radiotherapy in a Huh7 xenograft model by targeting SHP-1/STAT3 signaling. The novel sorafenib derivative, SC-59, acting as a SHP-1 agonist, displays a better synergistic effect when used in combination with radiotherapy than sorafenib for the treatment of HCC. Further clinical investigation is warranted.
Collapse
Affiliation(s)
- Chao-Yuan Huang
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Department of Radiological Technology, Yuanpei University, Hsinchu, Taiwan
| | - Wei-Tien Tai
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chi-Ying Hsieh
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Wan-Mai Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Ying-Jiun Lai
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Ju Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Wai Shiau
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan.
| | - Kuen-Feng Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
32
|
Chiorean EG, Schneider BP, Akisik FM, Perkins SM, Anderson S, Johnson CS, DeWitt J, Helft P, Clark R, Johnston EL, Spittler AJ, Deluca J, Bu G, Shahda S, Loehrer PJ, Sandrasegaran K, Cardenes HR. Phase 1 pharmacogenetic and pharmacodynamic study of sorafenib with concurrent radiation therapy and gemcitabine in locally advanced unresectable pancreatic cancer. Int J Radiat Oncol Biol Phys 2014; 89:284-91. [PMID: 24726286 DOI: 10.1016/j.ijrobp.2014.02.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 12/19/2013] [Accepted: 02/14/2014] [Indexed: 12/21/2022]
Abstract
PURPOSE To define the safety, efficacy, and pharmacogenetic and pharmacodynamic effects of sorafenib with gemcitabine-based chemoradiotherapy (CRT) in locally advanced pancreatic cancer. METHODS AND MATERIALS Patients received gemcitabine 1000 mg/m(2) intravenously weekly × 3 every 4 weeks per cycle for 1 cycle before CRT and continued for up to 4 cycles after CRT. Weekly gemcitabine 600 mg/m(2) intravenously was given during concurrent intensity modulated radiation therapy of 50 Gy to gross tumor volume in 25 fractions. Sorafenib was dosed orally 400 mg twice daily until progression, except during CRT when it was escalated from 200 mg to 400 mg daily, and 400 mg twice daily. The maximum tolerated dose cohort was expanded to 15 patients. Correlative studies included dynamic contrast-enhanced MRI and angiogenesis genes polymorphisms (VEGF-A and VEGF-R2 single nucleotide polymorphisms). RESULTS Twenty-seven patients were enrolled. No dose-limiting toxicity occurred during induction gemcitabine/sorafenib followed by concurrent CRT. The most common grade 3/4 toxicities were fatigue, hematologic, and gastrointestinal. The maximum tolerated dose was sorafenib 400 mg twice daily. The median progression-free survival and overall survival for 25 evaluable patients were 10.6 and 12.6 months, respectively. The median overall survival for patients with VEGF-A -2578 AA, -1498 CC, and -1154 AA versus alternate genotypes was 21.6 versus 14.7 months. Dynamic contrast-enhanced MRI demonstrated higher baseline K(trans) in responding patients. CONCLUSIONS Concurrent sorafenib with CRT had modest clinical activity with increased gastrointestinal toxicity in localized unresectable pancreatic cancer. Select VEGF-A/VEGF-R2 genotypes were associated with favorable survival.
Collapse
Affiliation(s)
- E Gabriela Chiorean
- Department of Medicine, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana; Department of Medicine, University of Washington, Seattle, Washington.
| | - Bryan P Schneider
- Department of Medicine, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Fatih M Akisik
- Department of Radiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Susan M Perkins
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Stephen Anderson
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Cynthia S Johnson
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana
| | - John DeWitt
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Paul Helft
- Department of Medicine, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Romnee Clark
- Department of Medicine, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Erica L Johnston
- Department of Medicine, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - A John Spittler
- Department of Medicine, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Jill Deluca
- Department of Radiation Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Guixue Bu
- Department of Radiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Safi Shahda
- Department of Medicine, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Patrick J Loehrer
- Department of Medicine, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Kumar Sandrasegaran
- Department of Radiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Higinia R Cardenes
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|