1
|
Zhou Y, Zhang Y, Bao J, Chen J, Song W. Low Temperature Plasma Suppresses Lung Cancer Cells Growth via VEGF/VEGFR2/RAS/ERK Axis. Molecules 2022; 27:5934. [PMID: 36144670 PMCID: PMC9502791 DOI: 10.3390/molecules27185934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Low temperature plasma (LTP) is a promising cancer therapy in clinical practice. In this study, dielectric barrier discharge plasma with helium gas was used to generate LTP. Significant increases in extracellular and intracellular reactive species were found in lung cancer cells (CALU-1 and SPC-A1) after LTP treatments. Cells viability and apoptosis assays demonstrated that LTP inhibited cells viability and induced cells death, respectively. Moreover, Western blotting revealed that the growth of CALU-1 cells was suppressed by LTP via the VEGF/VEGFR2/RAS/ERK axis for the first time. The results showed that LTP-induced ROS and RNS could inhibit the growth of lung cancer cells via VEGF/VEGFR2/RAS/ERK axis. These findings advance our understanding of the inhibitory mechanism of LTP on lung cancer and will facilitate its clinical application.
Collapse
Affiliation(s)
- Yuanyuan Zhou
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Yan Zhang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Jie Bao
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
| | - Jinwu Chen
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- School of Life Science, Hefei Normal University, Hefei 230061, China
| | - Wencheng Song
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
- Collaborative Innovation Center of Radiation Medicine, Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences, Soochow University, Suzhou 215123, China
| |
Collapse
|
2
|
Vendramini E, Bomben R, Pozzo F, Bittolo T, Tissino E, Gattei V, Zucchetto A. KRAS and RAS-MAPK Pathway Deregulation in Mature B Cell Lymphoproliferative Disorders. Cancers (Basel) 2022; 14:666. [PMID: 35158933 PMCID: PMC8833570 DOI: 10.3390/cancers14030666] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 02/04/2023] Open
Abstract
KRAS mutations account for the most frequent mutations in human cancers, and are generally correlated with disease aggressiveness, poor prognosis, and poor response to therapies. KRAS is required for adult hematopoiesis and plays a key role in B cell development and mature B cell proliferation and survival, proved to be critical for B cell receptor-induced ERK pathway activation. In mature B cell neoplasms, commonly seen in adults, KRAS and RAS-MAPK pathway aberrations occur in a relevant fraction of patients, reaching high recurrence in some specific subtypes like multiple myeloma and hairy cell leukemia. As inhibitors targeting the RAS-MAPK pathway are being developed and improved, it is of outmost importance to precisely identify all subgroups of patients that could potentially benefit from their use. Herein, we review the role of KRAS and RAS-MAPK signaling in malignant hematopoiesis, focusing on mature B cell lymphoproliferative disorders. We discuss KRAS and RAS-MAPK pathway aberrations describing type, incidence, mutual exclusion with other genetic abnormalities, and association with prognosis. We review the current therapeutic strategies applied in mature B cell neoplasms to counteract RAS-MAPK signaling in pre-clinical and clinical studies, including most promising combination therapies. We finally present an overview of genetically engineered mouse models bearing KRAS and RAS-MAPK pathway aberrations in the hematopoietic compartment, which are valuable tools in the understanding of cancer biology and etiology.
Collapse
Affiliation(s)
- Elena Vendramini
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (R.B.); (F.P.); (T.B.); (E.T.); (V.G.); (A.Z.)
| | | | | | | | | | | | | |
Collapse
|
3
|
Kurokawa R, Inui S, Tanishima T, Nakaya M, Kurokawa M, Ishida M, Gonoi W, Amemiya S, Nakai Y, Ishigaki K, Tateishi R, Koike K, Abe O. Incidence and computed tomography findings of lenvatinib-induced pancreatobiliary inflammation: A single-center, retrospective study. Medicine (Baltimore) 2021; 100:e27182. [PMID: 34477177 PMCID: PMC8415931 DOI: 10.1097/md.0000000000027182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/23/2021] [Indexed: 01/05/2023] Open
Abstract
In this single-center retrospective study, we intended to evaluate the frequencies and characteristics of computed tomography findings of pancreatobiliary inflammation (PBI) in patients treated with lenvatinib and the relationship of these findings with treatment-planning changes.We included 78 patients (mean ± standard deviation, 69.8 ± 9.4 years, range: 39-84 years, 62 men) with hepatocellular carcinoma (n = 62) or thyroid carcinoma (n = 16) who received lenvatinib (June 2016-September 2020). Two radiologists interpreted the posttreatment computed tomography images and assessed the radiological findings of PBI (symptomatic pancreatitis, cholecystitis, or cholangitis). The PBI effect on treatment was statistically evaluated.PBI (pancreatitis, n = 1; cholecystitis, n = 7; and cholangitis, n = 2) was diagnosed in 11.5% (9/78) of the patients at a median of 35 days after treatment initiation; 6 of 9 patients discontinued treatment because of PBI. Three cases of cholecystitis and 1 of cholangitis were accompanied by gallstones, while the other 5 were acalculous. The treatment duration was significantly shorter in patients with PBI than in those without (median: 44 days vs. 201 days, P = .02). Overall, 9 of 69 patients without PBI showed asymptomatic gallbladder subserosal edema.Lenvatinib-induced PBI developed in 11.5% of patients, leading to a significantly shorter treatment duration. Approximately 55.6% of the PBI cases were acalculous. The recognition of this phenomenon would aid physicians during treatment planning in the future.
Collapse
Affiliation(s)
- Ryo Kurokawa
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shohei Inui
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoya Tanishima
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Moto Nakaya
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mariko Kurokawa
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masanori Ishida
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Wataru Gonoi
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shiori Amemiya
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yousuke Nakai
- Department of Endoscopy and Endoscopic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazunaga Ishigaki
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryosuke Tateishi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Osamu Abe
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Signaling Pathway Mediating Myeloma Cell Growth and Survival. Cancers (Basel) 2021; 13:cancers13020216. [PMID: 33435632 PMCID: PMC7827005 DOI: 10.3390/cancers13020216] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The bone marrow (BM) microenvironment plays a crucial role in pathogenesis of multiple myeloma (MM), and delineation of the intracellular signaling pathways activated in the BM microenvironment in MM cells is essential to develop novel therapeutic strategies to improve patient outcome. Abstract The multiple myeloma (MM) bone marrow (BM) microenvironment consists of different types of accessory cells. Both soluble factors (i.e., cytokines) secreted from these cells and adhesion of MM cells to these cells play crucial roles in activation of intracellular signaling pathways mediating MM cell growth, survival, migration, and drug resistance. Importantly, there is crosstalk between the signaling pathways, increasing the complexity of signal transduction networks in MM cells in the BM microenvironment, highlighting the requirement for combination treatment strategies to blocking multiple signaling pathways.
Collapse
|
5
|
Ma X, Li L, Zhang L, Fu X, Li X, Wang X, Wu J, Sun Z, Zhang X, Feng X, Chang Y, Zhou Z, Nan F, Zhang J, Li Z, Zhang M. Apatinib in Patients with Relapsed or Refractory Diffuse Large B Cell Lymphoma: A Phase II, Open-Label, Single-Arm, Prospective Study. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:275-284. [PMID: 32158186 PMCID: PMC6986930 DOI: 10.2147/dddt.s227477] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/17/2019] [Indexed: 12/13/2022]
Abstract
Purpose Treatment options for relapsed or refractory diffuse large B-cell lymphoma (RR DLBCL) represent an unmet medical need. Apatinib is a new oral tyrosine kinase inhibitor mainly targeting vascular endothelial growth factor receptor-2 (VEGFR-2) to inhibit tumour angiogenesis. In the present study, we evaluated the efficacy and safety of apatinib for patients with RR DLBCL. Patients and Methods In this phase II, open-label, single-arm, prospective study, we enrolled patients aged 14–70 years with treatment failure of at least two chemotherapeutic regimens using Simon’s two-stage design. All patients were administered apatinib at an initial dose of 500 mg on a 4-week cycle at home and visited the outpatient clinic every two cycles to evaluate efficacy and to record adverse events. We considered objective response rate (ORR) as the primary end point, and progression-free survival (PFS), and overall survival (OS) plus duration of response (DoR) as the secondary end point. (This trial was registered at ClinicalTrials.gov, identifier: NCT03376958.). Results From January 2017 to February 2019, we screened 35 patients and enrolled 32 eligible patients. At the cutoff point (April 2019), we noted 2 (6.3%) complete responses, 12 (37.5%) partial responses, and 9 (28.1%) stable diseases, attributing to an ORR of 43.8% and a disease control rate of 71.9%. The median PFS and OS were 6.9 (95% confidence interval [CI], 5.8–7.9) and 7.9 months (95% CI, 7.0–8.7), respectively. The median DoR was 5.0 months (95% CI, 3.5–6.5) for patients who achieved PR. The most common grade 3–4 adverse events (AE) were hypertension (12.6%), hand–foot syndrome (9.4%), and leucopenia (6.3%). No apatinib-related deaths were noted. Conclusion Home administration of apatinib shows promising efficacy and manageable AEs in patients with RR DLBCL.
Collapse
Affiliation(s)
- Xinran Ma
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Ling Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Lei Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Xiaorui Fu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Xin Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Xinhua Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Jingjing Wu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Zhenchang Sun
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Xiaoyan Feng
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Yu Chang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Zhiyuan Zhou
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Feifei Nan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Jieming Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
6
|
Rapamycin and fingolimod modulate Treg/Th17 cells in experimental autoimmune encephalomyelitis by regulating the Akt-mTOR and MAPK/ERK pathways. J Neuroimmunol 2018; 324:26-34. [DOI: 10.1016/j.jneuroim.2018.08.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/24/2018] [Accepted: 08/24/2018] [Indexed: 12/31/2022]
|
7
|
McDonald JT, Kritharis A, Beheshti A, Pilichowska M, Burgess K, Ricks-Santi L, McNiel E, London CA, Ravi D, Evens AM. Comparative oncology DNA sequencing of canine T cell lymphoma via human hotspot panel. Oncotarget 2018; 9:22693-22702. [PMID: 29854308 PMCID: PMC5978258 DOI: 10.18632/oncotarget.25209] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/08/2018] [Indexed: 12/31/2022] Open
Abstract
T-cell lymphoma (TCL) is an uncommon and aggressive form of human cancer. Lymphoma is the most common hematopoietic tumor in canines (companion animals), with TCL representing approximately 30% of diagnoses. Collectively, the canine is an appealing model for cancer research given the spontaneous occurrence of cancer, intact immune system, and phytogenetic proximity to humans. We sought to establish mutational congruence of the canine with known human TCL mutations in order to identify potential actionable oncogenic pathways. Following pathologic confirmation, DNA was sequenced in 16 canine TCL (cTCL) cases using a custom Human Cancer Hotspot Panel of 68 genes commonly mutated in human TCL. Sequencing identified 4,527,638 total reads with average length of 229 bases containing 346 unique variants and 1,474 total variants; each sample had an average of 92 variants. Among these, there were 258 germline and 32 somatic variants. Among the 32 somatic variants there were 8 missense variants, 1 splice junction variant and the remaining were intron or synonymous variants. A frequency of 4 somatic mutations per sample were noted with >7 mutations detected in MET, KDR, STK11 and BRAF. Expression of these associated proteins were also detected via Western blot analyses. In addition, Sanger sequencing confirmed three variants of high quality (MYC, MET, and TP53 missense mutation). Taken together, the mutational spectrum and protein analyses showed mutations in signaling pathways similar to human TCL and also identified novel mutations that may serve as drug targets as well as potential biomarkers.
Collapse
Affiliation(s)
| | - Athena Kritharis
- Division of Blood Disorders, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | | | | | - Kristine Burgess
- Cummings School of Veterinary Medicine, Tufts University, Boston, MA, USA
| | | | - Elizabeth McNiel
- Cummings School of Veterinary Medicine, Tufts University, Boston, MA, USA
| | - Cheryl A London
- Cummings School of Veterinary Medicine, Tufts University, Boston, MA, USA
| | - Dashnamoorthy Ravi
- Division of Blood Disorders, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Andrew M Evens
- Division of Blood Disorders, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
8
|
Kießling MK, Nicolay JP, Schlör T, Klemke CD, Süss D, Krammer PH, Gülow K. NRAS mutations in cutaneous T cell lymphoma (CTCL) sensitize tumors towards treatment with the multikinase inhibitor Sorafenib. Oncotarget 2018; 8:45687-45697. [PMID: 28537899 PMCID: PMC5542218 DOI: 10.18632/oncotarget.17669] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/24/2017] [Indexed: 01/26/2023] Open
Abstract
Therapy of cutaneous T cell lymphoma (CTCL) is complicated by a distinct resistance of the malignant T cells towards apoptosis that can be caused by NRAS mutations in late-stage patients. These mutations correlate with decreased overall survival, but sensitize the respective CTCL cells towards MEK-inhibition-induced apoptosis which represents a promising novel therapeutic target in CTCL. Here, we show that the multi-kinase inhibitor Sorafenib induces apoptosis in NRAS-mutated CTCL cells. CTCL cell lines and to a minor extent primary T cells from Sézary patients without NRAS mutations are also affected by Sorafenib-induced apoptosis suggesting a sensitizing role of NRAS mutations for Sorafenib-induced apoptosis. When combining Sorafenib with the established CTCL medication Vorinostat we detected an increase in cell death sensitivity in CTCL cells. The combination treatment acted synergistically in apoptosis induction in both non-mutant and mutant CTCL cells. Mechanistically, this synergistic apoptosis induction by Sorafenib and Vorinostat is based on the downregulation of the anti-apoptotic protein Mcl-1, but not of other Bcl-2 family members. Taken together, these findings suggest that Sorafenib in combination with Vorinostat represents a novel therapeutic approach for the treatment of CTCL patients.
Collapse
Affiliation(s)
- Michael K Kießling
- German Cancer Research Center, 69120 Heidelberg, Germany.,Current address: Department of Gastroenterology, University Hospital of Zürich, 8091 Zürich, Switzerland
| | - Jan P Nicolay
- German Cancer Research Center, 69120 Heidelberg, Germany.,Department of Dermatology, Venerology and Allergology, University Medical Center Mannheim, Ruprecht Karls University of Heidelberg, 68167 Mannheim, Germany
| | - Tabea Schlör
- Department of Dermatology, Venerology and Allergology, University Medical Center Mannheim, Ruprecht Karls University of Heidelberg, 68167 Mannheim, Germany
| | - Claus-Detlev Klemke
- Department of Dermatology, Venerology and Allergology, University Medical Center Mannheim, Ruprecht Karls University of Heidelberg, 68167 Mannheim, Germany.,Current address: Department of Dermatology, Venerology and Allergology, General Hospital Karlsruhe, 76187 Karlsruhe, Germany
| | - Dorothee Süss
- German Cancer Research Center, 69120 Heidelberg, Germany
| | | | - Karsten Gülow
- German Cancer Research Center, 69120 Heidelberg, Germany
| |
Collapse
|
9
|
Prieto-Domínguez N, Ordóñez R, Fernández A, García-Palomo A, Muntané J, González-Gallego J, Mauriz JL. Modulation of Autophagy by Sorafenib: Effects on Treatment Response. Front Pharmacol 2016; 7:151. [PMID: 27375485 PMCID: PMC4896953 DOI: 10.3389/fphar.2016.00151] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/26/2016] [Indexed: 12/13/2022] Open
Abstract
The multikinase inhibitor sorafenib is, at present, the only drug approved for the treatment of hepatocellular carcinoma (HCC), one of the most lethal types of cancer worldwide. However, the increase in the number of sorafenib tumor resistant cells reduces efficiency. A better knowledge of the intracellular mechanism of the drug leading to reduced cell survival could help to improve the benefits of sorafenib therapy. Autophagy is a bulk cellular degradation process activated in a broad range of stress situations, which allows cells to degrade misfolded proteins or dysfunctional organelles. This cellular route can induce survival or death, depending on cell status and media signals. Sorafenib, alone or in combination with other drugs is able to induce autophagy, but cell response to the drug depends on the complex integrative crosstalk of different intracellular signals. In cancerous cells, autophagy can be regulated by different cellular pathways (Akt-related mammalian target of rapamycin (mTOR) inhibition, 5′ AMP-activated protein kinase (AMPK) induction, dissociation of B-cell lymphoma 2 (Bcl-2) family proteins from Beclin-1), or effects of some miRNAs. Inhibition of mTOR signaling by sorafenib and diminished interaction between Beclin-1 and myeloid cell leukemia 1 (Mcl-1) have been related to induction of autophagy in HCC. Furthermore, changes in some miRNAs, such as miR-30α, are able to modulate autophagy and modify sensitivity in sorafenib-resistant cells. However, although AMPK phosphorylation by sorafenib seems to play a role in the antiproliferative action of the drug, it does not relate with modulation of autophagy. In this review, we present an updated overview of the effects of sorafenib on autophagy and its related activation pathways, analyzing in detail the involvement of autophagy on sorafenib sensitivity and resistance.
Collapse
Affiliation(s)
- Nestor Prieto-Domínguez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain; Institute of Biomedicine (IBIOMED), University of LeónLeón, Spain
| | - Raquel Ordóñez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain; Institute of Biomedicine (IBIOMED), University of LeónLeón, Spain
| | - Anna Fernández
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain; Institute of Biomedicine (IBIOMED), University of LeónLeón, Spain
| | - Andres García-Palomo
- Service of Clinical Oncology, Complejo Asistencial Universitario de León (Hospital of León) León, Spain
| | - Jordi Muntané
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain; Department of General Surgery"Virgen del Rocío"-"Virgen Macarena" University Hospital/IBiS/CSIC/Universidad de Sevilla, Spain
| | - Javier González-Gallego
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain; Institute of Biomedicine (IBIOMED), University of LeónLeón, Spain
| | - José L Mauriz
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain; Institute of Biomedicine (IBIOMED), University of LeónLeón, Spain
| |
Collapse
|
10
|
Witzig TE, Reeder C, Han JJ, LaPlant B, Stenson M, Tun HW, Macon W, Ansell SM, Habermann TM, Inwards DJ, Micallef IN, Johnston PB, Porrata LF, Colgan JP, Markovic S, Nowakowski GS, Gupta M. The mTORC1 inhibitor everolimus has antitumor activity in vitro and produces tumor responses in patients with relapsed T-cell lymphoma. Blood 2015; 126:328-35. [PMID: 25921059 PMCID: PMC4504947 DOI: 10.1182/blood-2015-02-629543] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/20/2015] [Indexed: 12/17/2022] Open
Abstract
Everolimus is an oral agent that targets the mammalian target of rapamycin (mTOR) pathway. This study investigated mTOR pathway activation in T-cell lymphoma (TCL) cell lines and assessed antitumor activity in patients with relapsed/refractory TCL in a phase 2 trial. The mTOR pathway was activated in all 6 TCL cell lines tested and everolimus strongly inhibited malignant T-cell proliferation with minimal cytotoxic effects. Everolimus completely inhibited phosphorylation of ribosomal S6, a raptor/mTOR complex 1 (mTORC1) target, without a compensatory activation of the rictor/mTORC2 target Akt (S475). In the clinical trial, 16 patients with relapsed TCL were enrolled and received everolimus 10 mg by mouth daily. Seven patients (44%) had cutaneous (all mycosis fungoides); 4 (25%) had peripheral T cell not otherwise specified; 2 (13%) had anaplastic large cell; and 1 each had extranodal natural killer/T cell, angioimmunoblastic, and precursor T-lymphoblastic leukemia/lymphoma types. The overall response rate was 44% (7/16; 95% confidence interval [CI]: 20% to 70%). The median progression-free survival was 4.1 months (95% CI, 1.5-6.5) and the median overall survival was 10.2 months (95% CI, 2.6-44.3). The median duration of response for the 7 responders was 8.5 months (95% CI, 1.0 to not reached). These studies indicate that everolimus has antitumor activity and provide proof-of-concept that targeting the mTORC1 pathway in TCL is clinically relevant. This trial was registered at www.clinicaltrials.gov as #NCT00436618.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Apoptosis/drug effects
- Blotting, Western
- Cell Proliferation/drug effects
- Cytokines/blood
- Everolimus
- Female
- Flow Cytometry
- Humans
- Immunosuppressive Agents/pharmacology
- In Vitro Techniques
- Lymphoma, T-Cell/drug therapy
- Lymphoma, T-Cell/metabolism
- Lymphoma, T-Cell/mortality
- Lymphoma, T-Cell/pathology
- Male
- Mechanistic Target of Rapamycin Complex 1
- Mechanistic Target of Rapamycin Complex 2
- Middle Aged
- Multiprotein Complexes/antagonists & inhibitors
- Multiprotein Complexes/metabolism
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/mortality
- Neoplasm Recurrence, Local/pathology
- Phosphorylation
- Prognosis
- Sirolimus/analogs & derivatives
- Sirolimus/pharmacology
- Survival Rate
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Thomas E Witzig
- Division of Hematology, Department of Medicine, Mayo Clinic Rochester, Rochester, MN
| | - Craig Reeder
- Division of Hematology, Department of Medicine, Mayo Clinic Scottsdale, Scottsdale, AZ
| | - Jing Jing Han
- Division of Hematology, Department of Medicine, Mayo Clinic Rochester, Rochester, MN
| | - Betsy LaPlant
- Division of Biomedical Statistics and Bioinformatics, Department of Health Sciences Research, Mayo Clinic Rochester, Rochester, MN
| | - Mary Stenson
- Division of Hematology, Department of Medicine, Mayo Clinic Rochester, Rochester, MN
| | - Han W Tun
- Division of Hematology, Department of Medicine, Mayo Clinic Jacksonville, Jacksonville, FL; and
| | - William Macon
- Division of Hematopathology, Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, MN
| | - Stephen M Ansell
- Division of Hematology, Department of Medicine, Mayo Clinic Rochester, Rochester, MN
| | - Thomas M Habermann
- Division of Hematology, Department of Medicine, Mayo Clinic Rochester, Rochester, MN
| | - David J Inwards
- Division of Hematology, Department of Medicine, Mayo Clinic Rochester, Rochester, MN
| | - Ivana N Micallef
- Division of Hematology, Department of Medicine, Mayo Clinic Rochester, Rochester, MN
| | - Patrick B Johnston
- Division of Hematology, Department of Medicine, Mayo Clinic Rochester, Rochester, MN
| | - Luis F Porrata
- Division of Hematology, Department of Medicine, Mayo Clinic Rochester, Rochester, MN
| | - Joseph P Colgan
- Division of Hematology, Department of Medicine, Mayo Clinic Rochester, Rochester, MN
| | - Svetomir Markovic
- Division of Hematology, Department of Medicine, Mayo Clinic Rochester, Rochester, MN
| | - Grzegorz S Nowakowski
- Division of Hematology, Department of Medicine, Mayo Clinic Rochester, Rochester, MN
| | - Mamta Gupta
- Division of Hematology, Department of Medicine, Mayo Clinic Rochester, Rochester, MN
| |
Collapse
|
11
|
Bojarczuk K, Bobrowicz M, Dwojak M, Miazek N, Zapala P, Bunes A, Siernicka M, Rozanska M, Winiarska M. B-cell receptor signaling in the pathogenesis of lymphoid malignancies. Blood Cells Mol Dis 2015; 55:255-65. [PMID: 26227856 DOI: 10.1016/j.bcmd.2015.06.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 06/21/2015] [Indexed: 11/17/2022]
Abstract
B-cell receptor (BCR) signaling pathway plays a central role in B-lymphocyte development and initiation of humoral immunity. Recently, BCR signaling pathway has been shown as a major driver in the pathogenesis of B-cell malignancies. As a result, a vast array of BCR-associated kinases has emerged as rational therapeutic targets changing treatment paradigms in B cell malignancies. Based on high efficacy in early-stage clinical trials, there is rapid clinical development of inhibitors targeting BCR signaling pathway. Here, we describe the essential components of BCR signaling, their function in normal and pathogenic signaling and molecular effects of their inhibition in vitro and in vivo.
Collapse
Affiliation(s)
- Kamil Bojarczuk
- Department of Immunology, Center for Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; Postgraduate School of Molecular Medicine, Medical University of Warsaw, Zwirki I Wigury 61, 02-091 Warsaw, Poland
| | - Malgorzata Bobrowicz
- Department of Immunology, Center for Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; Postgraduate School of Molecular Medicine, Medical University of Warsaw, Zwirki I Wigury 61, 02-091 Warsaw, Poland
| | - Michal Dwojak
- Department of Immunology, Center for Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; Postgraduate School of Molecular Medicine, Medical University of Warsaw, Zwirki I Wigury 61, 02-091 Warsaw, Poland
| | - Nina Miazek
- Department of Immunology, Center for Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Piotr Zapala
- Department of Immunology, Center for Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Anders Bunes
- Department of Immunology, Center for Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Marta Siernicka
- Department of Immunology, Center for Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; Postgraduate School of Molecular Medicine, Medical University of Warsaw, Zwirki I Wigury 61, 02-091 Warsaw, Poland
| | - Maria Rozanska
- Department of Immunology, Center for Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Magdalena Winiarska
- Department of Immunology, Center for Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland.
| |
Collapse
|
12
|
Sie M, den Dunnen WFA, Lourens HJ, Meeuwsen-de Boer TGJ, Scherpen FJG, Zomerman WW, Kampen KR, Hoving EW, de Bont ESJM. Growth-factor-driven rescue to receptor tyrosine kinase (RTK) inhibitors through Akt and Erk phosphorylation in pediatric low grade astrocytoma and ependymoma. PLoS One 2015; 10:e0122555. [PMID: 25799134 PMCID: PMC4370756 DOI: 10.1371/journal.pone.0122555] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 02/23/2015] [Indexed: 01/20/2023] Open
Abstract
Up to now, several clinical studies have been started investigating the relevance of receptor tyrosine kinase (RTK) inhibitors upon progression free survival in various pediatric brain tumors. However, single targeted kinase inhibition failed, possibly due to tumor resistance mechanisms. The present study will extend our previous observations that vascular endothelial growth factor receptor (VEGFR)-2, platelet derived growth factor receptor (PDGFR)β, Src, the epidermal growth factor receptor (ErbB) family, and hepatocyte growth factor receptor (HGFR/cMet) are potentially drugable targets in pediatric low grade astrocytoma and ependymoma with investigations concerning growth-factor-driven rescue. This was investigated in pediatric low grade astrocytoma and ependymoma cell lines treated with receptor tyrosine kinase (RTK) inhibitors e.g. sorafenib, dasatinib, canertinib and crizotinib. Flow cytometry analyses showed high percentage of cells expressing VEGFR-1, fibroblast growth factor receptor (FGFR)-1, ErbB1/EGFR, HGFR and recepteur d’origine nantais (RON) (respectively 52-77%, 34-51%, 63-90%, 83-98%, 65-95%). Their respective inhibitors induced decrease of cell viability, measured with WST-1 cell viability assays. At least this was partially due to increased apoptotic levels measured by Annexin V/Propidium Iodide apoptosis assays. EGF, HGF and FGF, which are normally expressed in brain (tumor) tissue, showed to be effective rescue inducing growth factors resulting in increased cell survival especially during treatment with dasatinib (complete rescue) or sorafenib (partial rescue). Growth-factor-driven rescue was less prominent when canertinib or crizotinib were used. Rescue was underscored by significantly activating downstream Akt and/or Erk phosphorylation and increased tumor cell migration. Combination treatment showed to be able to overcome the growth-factor-driven rescue. In conclusion, our study highlights the extensive importance of environmentally present growth factors in developing tumor escape towards RTK inhibitors in pediatric low grade astrocytoma and ependymoma. It is of great interest to anticipate upon these results for the design of new therapeutic trials with RTK inhibitors in these pediatric brain tumors.
Collapse
Affiliation(s)
- Mariska Sie
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Wilfred F. A. den Dunnen
- Department of Pathology and Medical Biology, Pathology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Harm Jan Lourens
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Tiny G. J. Meeuwsen-de Boer
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Frank J. G. Scherpen
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Walderik W. Zomerman
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kim R. Kampen
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Eelco W. Hoving
- Department of Neurosurgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Eveline S. J. M. de Bont
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- * E-mail:
| |
Collapse
|
13
|
Feng K, Wang C, Zhou H, Yang J, Dong L, Zhou K, Liu X, Song Y. [Effect of ERK1/2 inhibitor AZD8330 on human Burkitt's lymphoma cell line Raji cells and its mechanism]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2015; 36:148-52. [PMID: 25778893 PMCID: PMC7342153 DOI: 10.3760/cma.j.issn.0253-2727.2015.02.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
目的 探讨细胞外信号调节激酶1/2(ERK1/2)抑制剂AZD8330对Burkitt淋巴瘤细胞株Raji细胞的作用及其机制。 方法 Raji细胞用不同浓度的AZD8330进行处理;采用CCK-8检测细胞存活率;流式细胞术检测细胞凋亡情况;实时定量PCR法检测Bcl-2、Bcl-xl、caspase-3和血管内皮生长因子(VEGF)mRNA表达;Western blot法检测Bcl-2、Bcl-xl、caspase-3、磷酸化(p)-ERK1/2蛋白表达。 结果 1.00 µmol/L的AZD8330处理24、48和72 h后细胞存活率分别为(62.09±0.86)%、(50.06±1.33)%和(39.13±2.34)%,差异有统计学意义(P值均<0.05);0.10、1.00、10.00 µmol/L的AZD8330分别处理Raji细胞24、48和72 h,Raji细胞发生凋亡,凋亡率呈时间和剂量依赖性,差异有统计学意义(P值均<0.05);随着浓度增加和时间延长,Bcl-2、Bcl-xl、VEGF mRNA表达降低,caspase-3 mRNA表达升高,差异有统计学意义(P值均<0.05);同时,Bcl-2、Bcl-xl、p-ERK1/2蛋白表达明显受抑制,而caspase-3蛋白表达增强。 结论 AZD8330可能通过抑制ERK1/2通路相关基因和蛋白的表达而诱导Burkitt淋巴瘤Raji细胞凋亡,抑制其增殖。
Collapse
Affiliation(s)
- Ke Feng
- Department of Hematology,the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Chao Wang
- Department of Hematology,the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Hu Zhou
- Department of Hematology,the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Jingyi Yang
- Department of Hematology,the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Lihua Dong
- Department of Hematology,the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Keshu Zhou
- Department of Hematology,the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Xinjian Liu
- Department of Hematology,the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Yongping Song
- Department of Hematology,the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| |
Collapse
|
14
|
Managing Hodgkin lymphoma relapsing after autologous hematopoietic cell transplantation: a not-so-good cancer after all! Bone Marrow Transplant 2014; 49:599-606. [PMID: 24442246 DOI: 10.1038/bmt.2013.226] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 11/27/2013] [Indexed: 01/02/2023]
Abstract
Hodgkin lymphoma (HL) relapsing after an autologous hematopoietic cell transplant (HCT) poses a therapeutic challenge. In this setting, salvage chemotherapy (for example, gemcitabine-based, ifosfamide-containing and others) or immunotherapy (for example, brentuximab vedotin) is essential as a bridging-cytoreduction strategy to an allogeneic HCT. Myeloablative allogeneic hematopoietic cell transplantation in relapsed HL is associated with high rates of non-relapse mortality. In carefully selected patients with chemosensitive disease, allografting following lower-intensity conditioning regimens can provide durable disease control rates of about 25-35%. Promising early results with haploidentical and umbilical cord transplantation are noteworthy and are expanding this procedure to patients for whom HLA-matched related or unrelated donors are not available. Unfortunately, a significant number of HL patients relapsing after an autologous HCT are not candidates for allografting because of the presence of resistant disease, donor unavailability or comorbidities. Brentuximab vedotin is approved for HL relapsing after a prior autograft. Rituximab and bendamustine are also active in this setting, albeit with short durations of remission. Histone deacetylase inhibitors (for example, panobinostat, mocetinostat), mTOR inhibitors (for example, everolimus) and immunomodulatory agents (lenalidomide) have shown activity in phase II trials, but currently are not approved for this indication. Second autologous HCT are rarely performed but this approach should not be considered standard practice at this time. The need for effective agents for post autograft failures of HL largely remains unmet. Continuous efforts to ensure early referral of such patients for allogeneic HCT or investigational therapies are the key to improving outcomes of this not-so-good lymphoma.
Collapse
|
15
|
Zhang YN, Wu XY, Zhong N, Deng J, Zhang L, Chen W, Li X, Zhong CJ. Stimulatory effects of sorafenib on human non‑small cell lung cancer cells in vitro by regulating MAPK/ERK activation. Mol Med Rep 2013; 9:365-9. [PMID: 24213303 DOI: 10.3892/mmr.2013.1782] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 11/05/2013] [Indexed: 11/06/2022] Open
Abstract
Sorafenib is an inhibitor of a number of intracellular signaling kinases with antiproliferative, anti‑angiogenic and pro‑apoptotic effects in tumor cells. Sorafenib has been used in the therapy of advanced renal cell carcinoma. In the present study, using two human non‑small cell lung cancer (NSCLC)cell lines, A549 and NCI‑H1975, the effects of sorafenib on proliferation, apoptosis and intracellular signaling were systematically characterized. The results revealed that at a low concentration (5 µM) and early time point (6 h), sorafenib is capable of significantly stimulating proliferation of A549 cells, but not NCI‑H1975 cells. In addition, the comparison of the two cell lines revealed different cell cycle redistribution and apoptotic susceptibility to sorafenib at this concentration and time point. Western blot analysis revealed that sorafenib upregulated the expression of cyclin D1 and cyclin‑dependent kinase 2 and downregulated the expression of BAX at this specific point. Furthermore, sorafenib was confirmed to regulate the expression of cyclin D1 and apoptosis‑associated proteins through the regulation of extracellular signal‑regulated kinase 1/2 phosphorylation in A549 cells. These findings suggest that, although sorafenib has the potential for use in the treatment of renal cell carcinoma, this compound may also activate NSCLC cells at a specific time point.
Collapse
Affiliation(s)
- Ya-Nian Zhang
- Department of Thoracic Surgery, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Du S, Wang S, Wu Q, Hu J, Li T. Decorin inhibits angiogenic potential of choroid-retinal endothelial cells by downregulating hypoxia-induced Met, Rac1, HIF-1α and VEGF expression in cocultured retinal pigment epithelial cells. Exp Eye Res 2013; 116:151-60. [PMID: 24016866 DOI: 10.1016/j.exer.2013.08.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 08/17/2013] [Accepted: 08/27/2013] [Indexed: 01/10/2023]
Abstract
Choroidal neovascularization (CNV) is one of the most common causes of severe vision loss. Decorin, a multiple receptor tyrosine kinase inhibitor, has been recently shown to play an important regulatory role in angiogenic response. This study aims to investigate whether the overexpression of decorin in retinal pigment epithelial (RPE) cells under hypoxia alters the in vitro angiogenic ability of cocultured choroid-retinal endothelial cells and to explore the possible mechanisms involved. Human RPE cells (ARPE-19) were subjected to hypoxia with or without decorin pretreatment, and RNA interference technique was used to knock down the Met gene in ARPE-19 cells. Cell viability was determined using the Cell Counting Kit-8 assay. Expression of Met, Rac1 and hypoxia-inducible factor-1 alpha (HIF-1α) was evaluated by western blot and quantitative real-time polymerase chain reaction (qRT-PCR). Vascular endothelial growth factor (VEGF) expression was evaluated by enzyme-linked immunosorbent assay (ELISA) and qRT-PCR. We then constructed a recombinant lentiviral vector carrying the decorin gene to transduce ARPE-19 cells. The overexpression of decorin in transduced RPE cells was confirmed by qRT-PCR and western blot. The transduced RPE cells were then cocultured with rhesus macaque choroid-retinal endothelial cells (RF/6A) in a transwell coculture system to observe the effects of decorin overexpression in ARPE-19 cells on the proliferation, migration and tube formation of RF/6A cells. In response to hypoxia, the VEGF concentrations in the culture supernatants increased greatly at 24 and 48 h, and this effect was inhibited significantly and nearly equally in the presence of 50-200 nM decorin. Decorin pretreatment before hypoxia exposure effectively reduced the hypoxia-induced expression of Met, Rac1, HIF-1α and VEGF in ARPE-19 cells. Transfection of small interfering RNA against Met to ARPE-19 cells also resulted in significant downregulation of Rac1, HIF-1α and VEGF under hypoxia, and this effect was similar to that noted with decorin pretreatment alone or with their combination. Results from the coculture system showed that the overexpression of decorin in ARPE-19 cells significantly inhibited the proliferation, migration and tube formation of RF/6A cells. These results indicate that Met pathway activation plays an important role in the upregulation of VEGF in RPE cells under hypoxia. Decorin may interfere with angiogenesis by downregulating hypoxia-induced Met, Rac1, HIF-1α and VEGF expression in RPE cells, which suggests a potential strategy for the inhibition of CNV.
Collapse
Affiliation(s)
- Shanshan Du
- Department of Ophthalmology, The Sixth People's Hospital, Shanghai Jiaotong University, 600 Yishan Road, Shanghai 200233, China
| | | | | | | | | |
Collapse
|
17
|
Waqar SN, Gopalan PK, Williams K, Devarakonda S, Govindan R. A phase I trial of sunitinib and rapamycin in patients with advanced non-small cell lung cancer. Chemotherapy 2013; 59:8-13. [PMID: 23635552 DOI: 10.1159/000348584] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 02/03/2013] [Indexed: 11/19/2022]
Abstract
BACKGROUND Sunitinib is an oral multitargeted tyrosine kinase inhibitor, with single-agent activity in non-small cell lung cancer (NSCLC). Resistance to tyrosine kinase inhibitor therapy is mediated by the mammalian target of rapamycin (mTOR) pathway, and may be reversed by using mTOR inhibitors. METHODS We performed a phase I study evaluating the combination of sunitinib and rapamycin in patients with advanced NSCLC. RESULTS Nineteen patients were enrolled in the study. The dose-limiting toxicities included infection, pneumonia, diarrhea/dehydration and treatment delay due to thrombocytopenia in 1 patient each. Sunitinib 25 mg orally daily and rapamycin 2 mg orally daily with 4 weeks on and 2 weeks off therapy were determined to be the maximum tolerated dose. No objective responses were noted, and 6 patients had stable disease as a best response. CONCLUSION The combination of sunitinib and rapamycin is well-tolerated and warrants further investigation in the phase II setting.
Collapse
Affiliation(s)
- Saiama N Waqar
- Division of Medical Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
18
|
Sorafenib inhibits lymphoma xenografts by targeting MAPK/ERK and AKT pathways in tumor and vascular cells. PLoS One 2013; 8:e61603. [PMID: 23620775 PMCID: PMC3631141 DOI: 10.1371/journal.pone.0061603] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 03/11/2013] [Indexed: 12/22/2022] Open
Abstract
The anti-lymphoma activity and mechanism(s) of action of the multikinase inhibitor sorafenib were investigated using a panel of lymphoma cell lines, including SU-DHL-4V, Granta-519, HD-MyZ, and KMS-11 cell lines. In vitro, sorafenib significantly decreased cell proliferation and phosphorylation levels of MAPK and PI3K/Akt pathways while increased apoptotic cell death. In vivo, sorafenib treatment resulted in a cytostatic rather than cytotoxic effect on tumor cell growth associated with a limited inhibition of tumor volumes. However, sorafenib induced an average 50% reduction of tumor vessel density and a 2-fold increase of necrotic areas. Upon sorafenib treatment, endothelial and tumor cells from SU-DHL-4V, Granta-519, and KMS-11 nodules showed a potent inhibition of either phospho-ERK or phospho-AKT, whereas a concomitant inhibition of phospho-ERK and phospho-AKT was only observed in HD-MyZ nodules. In conclusion, sorafenib affects the growth of lymphoid cell lines by triggering antiangiogenic mechanism(s) and directly targeting tumor cells.
Collapse
|
19
|
Synergistic interactions between sorafenib and everolimus in pancreatic cancer xenografts in mice. Cancer Chemother Pharmacol 2013; 71:1231-40. [PMID: 23455452 DOI: 10.1007/s00280-013-2117-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 02/05/2013] [Indexed: 12/22/2022]
Abstract
PURPOSE Molecular targeting of cellular signaling pathways is a promising approach in cancer therapy, but often fails to achieve sustained benefit because of the activation of collateral cancer cell survival and proliferation pathways. We tested the hypothesis that a combination of targeted agents that inhibit compensatory pathways would be more effective than single agents in controlling pancreatic cancer cell growth. We investigated whether everolimus, an mTOR inhibitor, and sorafenib, a multi-kinase inhibitor, would together inhibit growth of low-passage, patient-derived pancreatic cancer xenografts in mice more efficaciously than either agent alone. METHODS Tumor volume progression was measured following treatment with both drugs as single agents, in combination, and at multiple doses. Pharmacokinetics in tumors and other tissues was also assessed. Pharmacodynamic interactions were evaluated quantitatively. RESULTS A 5-week regimen of daily oral doses of 10 mg/kg sorafenib and 0.5 mg/kg everolimus, alone and in combination, did not achieve significant tumor growth inhibition. Higher doses (20 mg/kg of sorafenib and 1 mg/kg of everolimus) inhibited tumor growth significantly when given alone and caused complete inhibition of growth when given in combination. Tumor volume progression was described by a linear growth model, and drug effects were described by Hill-type inhibition. Using population modeling approaches, dual-interaction parameter estimates indicated a highly synergistic pharmacodynamic interaction between the two drugs. CONCLUSIONS The results indicate that combinations of mTOR and multi-kinase inhibitors may offer greater efficacy in pancreatic cancer than either drug alone. Drug effects upon tumor stromal elements may contribute to the enhanced anti-tumor efficacy.
Collapse
|
20
|
Holz MS, Janning A, Renné C, Gattenlöhner S, Spieker T, Bräuninger A. Induction of endoplasmic reticulum stress by sorafenib and activation of NF-κB by lestaurtinib as a novel resistance mechanism in Hodgkin lymphoma cell lines. Mol Cancer Ther 2012; 12:173-83. [PMID: 23243060 DOI: 10.1158/1535-7163.mct-12-0532] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hodgkin-Reed/Sternberg (HRS) cells of classical Hodgkin lymphoma show aberrant expression and activation of several receptor tyrosine kinases (RTK) in the majority of cases. Therefore, we tested whether tyrosine kinase inhibitors (TKI) already in clinical use or late stages of clinical trials have antiproliferative effects on HRS cell lines and evaluated the targets, affected signaling pathways, and mechanisms of cell death and resistance. Sorafenib and lestaurtinib had antiproliferative effects on HRS cell lines at concentrations achievable in patients. Sorafenib inhibited platelet-derived growth factor receptor (PDGFR) α, TRKA and RON, caused decreases in total and phosphorylated amounts of several signaling molecules, and provoked caspase-3-independent cell death, most likely due to endoplasmic reticulum stress as indicated by upregulation of GADD34 and GADD153 and phosphorylation of PERK. Lestaurtinib inhibited TRKA, PDGFRα, RON, and JAK2 and had only a cytostatic effect. Besides deactivation, lestaurtinib also caused activation of signaling pathways. It caused increases in CD30L and TRAIL expression, and CD30L/CD30 signaling likely led to the observed concomitant activation of extracellular signal-regulated kinase 1/2 and the alternative NF-κB pathway. These data disclose the possible use of sorafenib for the treatment of Hodgkin lymphoma and highlight NF-κB activation as a potential novel mechanism of resistance toward TKIs.
Collapse
Affiliation(s)
- Meike Stefanie Holz
- Gerhard-Domagk-Institute for Pathology, Westfälische Wilhelms-University, Münster, Germany
| | | | | | | | | | | |
Collapse
|
21
|
Barton S, Hawkes EA, Wotherspoon A, Cunningham D. Are we ready to stratify treatment for diffuse large B-cell lymphoma using molecular hallmarks? Oncologist 2012; 17:1562-73. [PMID: 23086691 PMCID: PMC3528389 DOI: 10.1634/theoncologist.2012-0218] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 09/04/2012] [Indexed: 01/01/2023] Open
Abstract
The division of the heterogeneous entity of diffuse large B-cell lymphoma (DLBCL) into the ontogenic phenotypes of germinal center B-cell-like (GCB) and activated B-cell-like (ABC) is optimally determined by gene expression profiling (GEP), although simpler immunohistochemistry (IHC) algorithms are alternatively being used. The cell-of-origin (COO) classification assists in prognostication and may be predictive of response to therapy. Mounting data suggests that IHC methods of classifying COO may be inaccurate. GEP categorization of COO is superior in defining prognostically and biologically distinct DLBCL subtypes, but current barriers to its widescale use include inaccessibility, cost, and lack of methodological standardization and prospective validation. The poorer prognosis of ABC-DLBCL is frequently associated with constitutive activity in the NF-κB pathway and aberrations in upstream or downstream regulators of this pathway. The molecular mechanisms underlying lymphomagenesis in GCB-DLBCL are arguably less well defined, but C-REL amplification and mutations in BCL-2 and EZH2 are common. New technologies, such as next-generation sequencing, are rapidly revealing novel pathogenic genetic aberrations, and DLBCL treatment strategies are increasingly being designed focusing on distinctive pathogenic drivers within ontogenic phenotypes. This review examines emerging molecular targets and novel therapeutic agents in DLBCL, and discusses whether stratifying therapy for DLBCL using molecular features is merited by current preclinical and clinical evidence.
Collapse
Affiliation(s)
| | | | - Andrew Wotherspoon
- Department of Histopathology, Royal Marsden Hospital, London, United Kingdom
| | | |
Collapse
|
22
|
Pawaskar DK, Straubinger RM, Fetterly GJ, Ma WW, Jusko WJ. Interactions of everolimus and sorafenib in pancreatic cancer cells. AAPS JOURNAL 2012; 15:78-84. [PMID: 23054975 DOI: 10.1208/s12248-012-9417-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 09/23/2012] [Indexed: 01/01/2023]
Abstract
Everolimus targets the mammalian target of rapamycin, a kinase that promotes cell growth and proliferation in pancreatic cancer. Sorafenib inhibits the Raf-mitogen-activated protein kinase, vascular endothelial growth factor, and platelet-derived growth factor pathways, thus inhibiting cell growth and angiogenesis. Combinations of these two agents are under evaluation for therapy of several cancers. This study examined the effects of everolimus and sorafenib on proliferation of the pancreatic cancer cell lines MiaPaCa-2 and Panc-1. Cell growth inhibition was evaluated in vitro for a range of concentrations of the drugs alone and in combination. Maximum inhibition capacity (I (max)) and potency (IC(50)) were determined. The data were analyzed to characterize drug interactions using two mathematical analysis techniques. The Ariens noncompetitive interaction model and Earp model were modified to accommodate alterations in the inhibition parameters of one drug in the presence of another. Sorafenib alone inhibited growth of both cell lines completely (I (max) = 1), with an IC(50) of 5-8 μM. Maximal inhibition by everolimus alone was only 40% (I (max) = 0.4) in both cell lines, with an IC(50) of 5 nM. Slight antagonistic interaction occurred between the drugs; both analytic methods estimated the interaction term Ψ as greater than 1 for both cell lines. The in vitro data for two pancreatic cancer cell lines suggest that a combination of these two drugs would be no more efficacious than the individual drugs alone, consistent with the drug interaction analysis that indicated slight antagonism for growth inhibition.
Collapse
Affiliation(s)
- Dipti K Pawaskar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, 404 Kapoor Hall, Buffalo, NY 14214, USA
| | | | | | | | | |
Collapse
|
23
|
Paesler J, Gehrke I, Poll-Wolbeck SJ, Kreuzer KA. Targeting the vascular endothelial growth factor in hematologic malignancies. Eur J Haematol 2012; 89:373-84. [DOI: 10.1111/ejh.12009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2012] [Indexed: 12/16/2022]
Affiliation(s)
- Julian Paesler
- Department I of Internal Medicine I; University at Cologne; Cologne; Germany
| | - Iris Gehrke
- Department I of Internal Medicine I; University at Cologne; Cologne; Germany
| | | | - Karl-Anton Kreuzer
- Department I of Internal Medicine I; University at Cologne; Cologne; Germany
| |
Collapse
|