51
|
Abstract
Prostate cancer at advanced stages including metastatic and castration-resistant cancer remains incurable due to the lack of effective therapies. The CAMK2N1 gene, cloned and characterized as an inhibitor of CaMKII (calcium/calmodulin-dependent protein kinase II), has been shown to affect tumorigenesis and tumor growth. However, it is still unknown whether CAMK2N1 plays a role in prostate cancer development. We first examined the protein and mRNA levels of CAMK2N1 and observed a significant decrease in human prostate cancers comparing to normal prostate tissues. Re-expression of CAMK2N1 in prostate cancer cells reduced cellular proliferation, arrested cells in G0/G1 phases, and induced apoptotic cell death accompanied by down-regulation of IGF-1, ErbB2, and VEGF downstream kinases PI3K/AKT, as well as the MEK/ERK-mediated signaling pathways. Conversely, knockdown of CAMK2N1 had a significant opposite effects on these phenotypes. Our analyses suggest that CAMK2N1 plays a tumor suppressive role in prostate cancer cells. Reduced CAMK2N1 expression correlates to human prostate cancer progression and predicts poor clinical outcome, indicating that CAMK2N1 may serve as a biomarker. The inhibition of tumor growth by expressing CAMK2N1 established a role of CAMK2N1 as a therapeutic target.
Collapse
|
52
|
Huang HL, Peng CY, Lai MJ, Chen CH, Lee HY, Wang JC, Liou JP, Pan SL, Teng CM. Novel oral histone deacetylase inhibitor, MPT0E028, displays potent growth-inhibitory activity against human B-cell lymphoma in vitro and in vivo. Oncotarget 2015; 6:4976-91. [PMID: 25669976 PMCID: PMC4467128 DOI: 10.18632/oncotarget.3213] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/26/2014] [Indexed: 12/16/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitor has been a promising therapeutic option in cancer therapy due to its ability to induce growth arrest, differentiation, and apoptosis. In this study, we demonstrated that MPT0E028, a novel HDAC inhibitor, reduces the viability of B-cell lymphomas by inducing apoptosis and shows a more potent HDAC inhibitory effect compared to SAHA, the first HDAC inhibitor approved by the FDA. In addition to HDACs inhibition, MPT0E028 also possesses potent direct Akt targeting ability as measured by the kinome diversity screening assay. Also, MPT0E028 reduces Akt phosphorylation in B-cell lymphoma with an IC50 value lower than SAHA. Transient transfection assay revealed that both targeting HDACs and Akt contribute to the apoptosis induced by MPT0E028, with both mechanisms functioning independently. Microarray analysis also shows that MPT0E028 may regulate many oncogenes expression (e.g., TP53, MYC, STAT family). Furthermore, in vivo animal model experiments demonstrated that MPT0E028 (50-200 mg/kg, po, qd) prolongs the survival rate of mice bearing human B-cell lymphoma Ramos cells and inhibits tumor growth in BJAB xenograft model. In summary, MPT0E028 possesses strong in vitro and in vivo activity against malignant cells, representing a potential therapeutic approach for cancer therapy.
Collapse
Affiliation(s)
- Han-Li Huang
- 1 Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chieh-Yu Peng
- 2 Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
- 3 School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Mei-Jung Lai
- 4 Center for Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun-Han Chen
- 1 Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsueh-Yun Lee
- 5 School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jing-Chi Wang
- 6 The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Jing-Ping Liou
- 5 School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Shiow-Lin Pan
- 6 The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Che-Ming Teng
- 1 Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
53
|
Kebenko M, Drenckhan A, Gros SJ, Jücker M, Grabinski N, Ewald F, Grottke A, Schultze A, Izbicki JR, Bokemeyer C, Wellbrock J, Fiedler W. ErbB2 signaling activates the Hedgehog pathway via PI3K-Akt in human esophageal adenocarcinoma: identification of novel targets for concerted therapy concepts. Cell Signal 2014; 27:373-81. [PMID: 25435423 DOI: 10.1016/j.cellsig.2014.11.022] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/21/2014] [Indexed: 01/12/2023]
Abstract
The Hedgehog pathway plays an important role in the pathogenesis of several tumor types, including esophageal cancer. In our study, we show an expression of the ligand Indian hedgehog (Ihh) and its downstream mediator Gli-1 in primary resected adenocarcinoma tissue by immunohistochemistry and quantitative PCR in fifty percent of the cases, while matching healthy esophagus mucosa was negative for both proteins. Moreover, a functionally important regulation of Gli-1 by ErbB2-PI3K-mTORC signaling as well as a Gli-1-dependent regulation of Ihh in the ErbB2 amplified esophageal adenocarcinoma cell line OE19 was observed. Treatment of OE19 cells with the Her2 antibody trastuzumab, the PI3K-mTORC1 inhibitor NVP BEZ235 (BEZ235) or the knockdown of Akt1 resulted in a downregulation of Gli-1 and Ihh as well as in a reduction of viable OE19 cells in vitro. Interestingly, the Hedgehog receptor Smo, which acts upstream of Gli-1, was not expressed in OE19 cells and in the majority of primary human esophageal adenocarcinoma, suggesting a non-canonical upregulation of Gli-1 expression by the ErbB2-PI3K axis. To translate our findings into a therapeutic concept, we targeted ErbB2-PI3K-mTORC1 by trastuzumab and BEZ235, combining both compounds with the Gli-1/2 inhibitor GANT61. The triple combination led to significantly stronger reduction of tumor cell viability than cisplatinum or each biological alone. Therefore, concomitant blockage of the ErbB2-PI3K pathway and the Hedgehog downstream mediator Gli-1 may provide a new therapeutic strategy for esophageal cancer.
Collapse
Affiliation(s)
- Maxim Kebenko
- Hubertus Wald Tumorzentrum, Department of Oncology-Hematology, Bone Marrow Transplantation and Pneumology, University Cancer Center, Hamburg, Germany
| | - Astrid Drenckhan
- Department of General, Visceral and Thoracic Surgery, Hamburg, Germany
| | - Stephanie J Gros
- Department of General, Visceral and Thoracic Surgery, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Eppendorf, Hamburg, Germany
| | - Nicole Grabinski
- Institute of Biochemistry and Signal Transduction, University Medical Center Eppendorf, Hamburg, Germany
| | - Florian Ewald
- Institute of Biochemistry and Signal Transduction, University Medical Center Eppendorf, Hamburg, Germany
| | - Astrid Grottke
- Institute of Biochemistry and Signal Transduction, University Medical Center Eppendorf, Hamburg, Germany
| | - Alexander Schultze
- Hubertus Wald Tumorzentrum, Department of Oncology-Hematology, Bone Marrow Transplantation and Pneumology, University Cancer Center, Hamburg, Germany
| | - Jakob R Izbicki
- Department of General, Visceral and Thoracic Surgery, Hamburg, Germany
| | - Carsten Bokemeyer
- Hubertus Wald Tumorzentrum, Department of Oncology-Hematology, Bone Marrow Transplantation and Pneumology, University Cancer Center, Hamburg, Germany
| | - Jasmin Wellbrock
- Hubertus Wald Tumorzentrum, Department of Oncology-Hematology, Bone Marrow Transplantation and Pneumology, University Cancer Center, Hamburg, Germany
| | - Walter Fiedler
- Hubertus Wald Tumorzentrum, Department of Oncology-Hematology, Bone Marrow Transplantation and Pneumology, University Cancer Center, Hamburg, Germany.
| |
Collapse
|
54
|
Qin Y, Zhao X, Fang Y. PP242 synergizes with suberoylanilide hydroxamic acid to inhibit growth of ovarian cancer cells. Int J Gynecol Cancer 2014; 24:1373-80. [PMID: 25188886 DOI: 10.1097/igc.0000000000000238] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVES Overexpression of histone deacetylases and activation of the phosphatidylinositol 3-kinase/mammalian target of rapamycin pathway are common aberrations in ovarian cancer. For this reason, simultaneous inhibition of such targets is a rational therapeutic strategy to treat patients with ovarian cancer. This study aimed to investigate the biological effect of the histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), in combination with the dual mTOR complex 1 and mTOR complex 2 inhibitor, PP242, against ovarian cancer cells. MATERIALS AND METHODS The effects of SAHA and PP242 on the growth of SKOV3 and A2780 cells were examined using Cell Counting Kit-8. The apoptosis was analyzed through flow cytometry, and the expression of apoptosis-related proteins was investigated through Western blotting. Induction of autophagy was determined through fluorescence microscopy using a stably transfected green fluorescent protein/microtubule-associated protein light chain 3 construct to visualize autophagosome formation. The expression of autophagy-related proteins was determined through Western blot analysis. The effect of SAHA and PP242 on the growth of ovarian cancer was also examined in an orthotopic ovarian cancer model. RESULTS The combination of SAHA and PP242 significantly inhibited cell proliferation and synergistically increased apoptosis and autophagy compared with each agent alone in vitro. In vivo, this combination exhibited greater inhibition on tumor growth than monotreatments did and it significantly prolonged the survival time of the mice. CONCLUSIONS These results suggest that the combination of SAHA and PP242 may lead to a novel strategy in treating patients with ovarian cancer.
Collapse
Affiliation(s)
- Yu Qin
- *Cancer Biology Research Center; and †Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | | | | |
Collapse
|
55
|
Toren P, Zoubeidi A. Targeting the PI3K/Akt pathway in prostate cancer: challenges and opportunities (review). Int J Oncol 2014; 45:1793-801. [PMID: 25120209 DOI: 10.3892/ijo.2014.2601] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 07/04/2014] [Indexed: 11/06/2022] Open
Abstract
The PI3K/Akt pathway is an actively pursued therapeutic target in oncology. In prostate cancer, the activation of this pathway appears to be characteristic of many aggressive prostate cancers. Further, activation of the PI3K/Akt pathway is more frequently observed as prostate cancer progresses toward a resistant, metastatic disease. Signalling from this pathway activates numerous survival, growth, metabolic and metastatic functions characteristic of aggressive cancer. Biomarkers of this pathway have correlated activation of this pathway to high grade disease and higher risk of disease progression. Therefore there is significant interest in developing effective strategies to target this pathway in prostate cancer. In this review, we discuss the pre-clinical and clinical data relevant to targeting of the PI3K/Akt pathway in prostate cancer. In particular, we review the rationale and relevance of co-targeting approaches against the PI3K/Akt pathway. It is anticipated that through an improved understanding of the biology of the PI3K/Akt pathway in prostate cancer, relevant biomarkers and rationale combination therapies will optimize targeting of this pathway to improve outcomes among patients with aggressive prostate cancer.
Collapse
Affiliation(s)
- Paul Toren
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia V6H 3Z6, Canada
| | - Amina Zoubeidi
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia V6H 3Z6, Canada
| |
Collapse
|
56
|
Rahmani M, Aust MM, Benson EC, Wallace L, Friedberg J, Grant S. PI3K/mTOR inhibition markedly potentiates HDAC inhibitor activity in NHL cells through BIM- and MCL-1-dependent mechanisms in vitro and in vivo. Clin Cancer Res 2014; 20:4849-60. [PMID: 25070836 DOI: 10.1158/1078-0432.ccr-14-0034] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
PURPOSE The aim of this study is to explore the efficacy and define mechanisms of action of coadministration of the PI3K/mTOR inhibitor BEZ235 and pan-HDAC inhibitor panobinostat in diffuse large B-cell lymphoma (DLBCL) cells. EXPERIMENTAL DESIGN Various DLBCL cells were exposed to panobinostat and BEZ235 alone or together after which apoptosis and signaling/survival pathway perturbations were monitored by flow cytometry and Western blot analysis. Genetic strategies defined the functional significance of such changes, and xenograft mouse models were used to assess tumor growth and animal survival. RESULTS Panobinostat and BEZ235 interacted synergistically in ABC-, GC-, and double-hit DLBCL cells and MCL cells but not in normal CD34(+) cells. Synergism was associated with pronounced AKT dephosphorylation, GSK3 dephosphorylation/activation, Mcl-1 downregulation, Bim upregulation, increased Bcl-2/Bcl-xL binding, diminished Bax/Bak binding to Bcl-2/Bcl-xL/Mcl-1, increased γH2A.X phosphorylation and histone H3/H4 acetylation, and abrogation of p21(CIP1) induction. BEZ235/panobinostat lethality was not susceptible to stromal/microenvironmental forms of resistance. Genetic strategies confirmed significant functional roles for AKT inactivation, Mcl-1 downregulation, Bim upregulation, and Bax/Bak in synergism. Finally, coadministration of BEZ235 with panobinostat in immunocompromised mice bearing SU-DHL4-derived tumors significantly reduced tumor growth in association with similar signaling changes observed in vitro, and combined treatment increased animal survival compared with single agents. CONCLUSIONS BEZ235/panobinostat exhibits potent anti-DLBCL activity, including in poor-prognosis ABC- and double-hit subtypes, but not in normal CD34(+) cells. Synergism is most likely multifactorial, involving AKT inactivation/GSK3 activation, Bim upregulation, Mcl-1 downregulation, enhanced DNA damage, and is operative in vivo. Combined PI3K/mTOR and HDAC inhibition warrants further attention in DLBCL.
Collapse
Affiliation(s)
- Mohamed Rahmani
- Department of Medicine, Virginia Commonwealth University, Richmond, Virginia.
| | - Mandy Mayo Aust
- Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Elisa C Benson
- Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - LaShanale Wallace
- Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Jonathan Friedberg
- James T. Wilmot Cancer Center, University of Rochester, Rochester, New York
| | | |
Collapse
|
57
|
Castel P, Toska E, Zumsteg ZS, Carmona FJ, Elkabets M, Bosch A, Scaltriti M. Rationale-based therapeutic combinations with PI3K inhibitors in cancer treatment. Mol Cell Oncol 2014; 1:e963447. [PMID: 27308344 PMCID: PMC4904898 DOI: 10.4161/23723548.2014.963447] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 08/06/2014] [Accepted: 08/12/2014] [Indexed: 02/07/2023]
Abstract
The PI3K/AKT/mTOR signaling is important for cell proliferation, survival, and metabolism. Hyperactivation of this pathway is one of the most common signaling abnormalities observed in cancer and a substantial effort has recently been made to develop molecules targeting this signaling cascade. However, it is becoming evident that PI3K inhibitors used as single agents do not elicit dramatic or durable responses. Given the numerous mechanisms mediating intrinsic and acquired resistance to these agents, hypothesis-based combinatorial strategies are probably needed to fully exploit their antitumor activity. In the first part of this review, we briefly dissect the PI3K/AKT/mTOR axis and list the most advanced compounds targeting different nodes of this cascade. The second part focuses on what we believe to be the most promising rationale-based therapeutic combinations with PI3K/AKT/mTOR inhibitors in solid tumors, with special emphasis on breast cancer.
Collapse
Affiliation(s)
- Pau Castel
- Human Oncology & Pathogenesis Program (HOPP); Memorial Sloan Kettering Cancer Center; New York, NY USA
| | - Eneda Toska
- Human Oncology & Pathogenesis Program (HOPP); Memorial Sloan Kettering Cancer Center; New York, NY USA
| | - Zachary S Zumsteg
- Human Oncology & Pathogenesis Program (HOPP); Memorial Sloan Kettering Cancer Center; New York, NY USA
| | - F Javier Carmona
- Human Oncology & Pathogenesis Program (HOPP); Memorial Sloan Kettering Cancer Center; New York, NY USA
| | - Moshe Elkabets
- Human Oncology & Pathogenesis Program (HOPP); Memorial Sloan Kettering Cancer Center; New York, NY USA
| | - Ana Bosch
- Human Oncology & Pathogenesis Program (HOPP); Memorial Sloan Kettering Cancer Center; New York, NY USA
| | - Maurizio Scaltriti
- Human Oncology & Pathogenesis Program (HOPP); Memorial Sloan Kettering Cancer Center; New York, NY USA
| |
Collapse
|
58
|
Juengel E, Nowaz S, Makarevi J, Natsheh I, Werner I, Nelson K, Reiter M, Tsaur I, Mani J, Harder S, Bartsch G, Haferkamp A, Blaheta RA. HDAC-inhibition counteracts everolimus resistance in renal cell carcinoma in vitro by diminishing cdk2 and cyclin A. Mol Cancer 2014; 13:152. [PMID: 24935000 PMCID: PMC4073177 DOI: 10.1186/1476-4598-13-152] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 06/03/2014] [Indexed: 02/06/2023] Open
Abstract
Background Targeted therapies have improved therapeutic options of treating renal cell carcinoma (RCC). However, drug response is temporary due to resistance development. Methods Functional and molecular changes in RCC Caki-1 cells, after acquired resistance to the mammalian target of rapamycin (mTOR)-inhibitor everolimus (Cakires), were investigated with and without additional application of the histone deacetylase (HDAC)-inhibitor valproic acid (VPA). Cell growth was evaluated by MTT assay, cell cycle progression and apoptosis by flow cytometry. Target molecules of everolimus and VPA, apoptotic and cell cycle regulating proteins were investigated by western blotting. siRNA blockade was performed to evaluate the functional relevance of the proteins. Results Everolimus resistance was accompanied by significant increases in the percentage of G2/M-phase cells and in the IC50. Akt and p70S6K, targets of everolimus, were activated in Cakires compared to drug sensitive cells. The most prominent change in Cakires cells was an increase in the cell cycle activating proteins cdk2 and cyclin A. Knock-down of cdk2 and cyclin A caused significant growth inhibition in the Cakires cells. The HDAC-inhibitor, VPA, counteracted everolimus resistance in Cakires, evidenced by a significant decrease in tumor growth and cdk2/cyclin A. Conclusion It is concluded that non-response to everolimus is characterized by increased cdk2/cyclin A, driving RCC cells into the G2/M-phase. VPA hinders everolimus non-response by diminishing cdk2/cyclin A. Therefore, treatment with HDAC-inhibitors might be an option for patients with advanced renal cell carcinoma and acquired everolimus resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Roman A Blaheta
- Department of Urology, Goethe-University, Interdisciplinary Science Building, Building 25A, Room 404, Theodor-Stern-Kai 7, Frankfurt / Main D-60590, Germany.
| |
Collapse
|
59
|
Makarević J, Tawanaie N, Juengel E, Reiter M, Mani J, Tsaur I, Bartsch G, Haferkamp A, Blaheta RA. Cross-communication between histone H3 and H4 acetylation and Akt-mTOR signalling in prostate cancer cells. J Cell Mol Med 2014; 18:1460-6. [PMID: 24779401 PMCID: PMC4124028 DOI: 10.1111/jcmm.12299] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 03/18/2014] [Indexed: 12/26/2022] Open
Abstract
Molecular tumour targeting has significantly improved anti-cancer protocols. Still, the addition of molecular targeting to the treatment regime has not led to a curative breakthrough. Combined mammalian target of Rapamycin (mTOR) and histone deacetylase (HDAC) inhibition has been shown not only to enhance anti-tumour potential, but also to prevent resistance development seen under mono-drug therapy. This investigation was designed to evaluate whether cross-communication exists between mTOR signalling and epigenetic events regulated by HDAC. DU-145 prostate cancer cells were treated with insulin-like growth factor (IGF) to activate the Akt-mTOR cascade or with the HDAC-inhibitor valproic acid (VPA) to induce histone H3 and H4 acetylation (aH3, aH4). Subsequently, mTOR, Rictor, Raptor, p70s6k, Akt (all: total and phosphorylated), H3 and H4 (total and acetylated) were analysed by western blotting. Both techniques revealed a link between mTOR and the epigenetic machinery. IGF activated mTOR, Rictor, Raptor, p70s6k and Akt, but also enhanced aH3 and aH4. Inversely, IGFr blockade and knock-down blocked the Akt-mTOR axis, but simultaneously diminished aH3 and aH4. VPA treatment up-regulated histone acetylation, but also activated mTOR-Akt signalling. HDAC1 and 2 knock-down revealed that the interaction with the mTOR system is initiated by histone H3 acetylation. HDAC-mTOR communication, therefore, is apparent whereby tumour-promoting (Akt/mTORhigh, aH3/aH4low) and tumour-suppressing signals (Akt/mTORlow, aH3/aH4high) are activated in parallel. Combined use of an HDAC- and mTOR inhibitor might then diminish pro-tumour effects triggered by the HDAC- (Akt/mTORhigh) or mTOR inhibitor (aH3/aH4low) alone.
Collapse
Affiliation(s)
- Jasmina Makarević
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|