151
|
Kobayashi Y, Masuda K, Banno K, Kobayashi N, Umene K, Nogami Y, Tsuji K, Ueki A, Nomura H, Sato K, Tominaga E, Shimizu T, Saya H, Aoki D. Glycan profiling of gestational choriocarcinoma using a lectin microarray. Oncol Rep 2014; 31:1121-6. [PMID: 24424471 DOI: 10.3892/or.2014.2979] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 09/09/2013] [Indexed: 11/05/2022] Open
Abstract
Glycosylation is an important post-translational modification, in which attachment of glycans to proteins has effects on biological functions and carcinogenesis. Analysis of human chorionic gonadotropin, a glycoprotein hormone produced by placental trophoblasts and trophoblastic tumors, has contributed to the diagnosis and treatment of trophoblastic disease, resulting in reduced incidence and mortality. However, alterations of the glycan structure itself in choriocarcinoma have not been characterized. We established a new choriocarcinoma cell line, induced choriocarcinoma cell-1 (iC3-1), which mimics the clinical pathohistology in vivo, to examine the tumorigenesis and pathogenesis of choriocarcinoma. In this study, the alterations of glycan structures in the development of choriocarcinoma were examined by performance of comprehensive glycan profiling in clinical samples and in iC3-1 cells using a conventional microarray and the recently introduced lectin microarray. Microarray comparison showed significant upregulation of several characteristic glycogenes in the iC3-1 cells as compared to the parental HTR8/SVneo cells. The lectin array showed increased α-2-6-sialic acid, Galβ1-4GlcNAc, GlcNAcβ1-3GalNAc, and decreased α-1-6 core fucose, high mannose, GalNacβ1-4Gal, GALNAc (Tn antigen) and Galβ1-3Gal in choriocarcinoma tissue compared to normal villi. This is the first report of a lectin array analysis in choriocarcinoma and provides useful information for understanding of the disease.
Collapse
Affiliation(s)
- Yusuke Kobayashi
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Kenta Masuda
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Kouji Banno
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Nana Kobayashi
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Kiyoko Umene
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Yuya Nogami
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Kosuke Tsuji
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Arisa Ueki
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Hiroyuki Nomura
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Kenji Sato
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Eiichiro Tominaga
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Takatsune Shimizu
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| |
Collapse
|
152
|
Tumor-necrosis-factor-related apoptosis-inducing ligand (TRAIL). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 818:167-80. [PMID: 25001536 DOI: 10.1007/978-1-4471-6458-6_8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The concept to exploit death receptors for cancer therapy is very attractive, since these cell surface receptors have a direct connection to the intracellular cell death machinery. Among the death receptor superfamily, the tumor-necrosis-factor-related apoptosis-inducing ligand (TRAIL) receptor/ligand system is of special interest. TRAIL receptor agonists have recently entered the stage of clinical evaluation for the treatment of human cancers. Further insights into the regulatory mechanisms of TRAIL signaling will help to better understand the determinants of TRAIL sensitivity versus resistance of human cancers.
Collapse
|
153
|
von Pawel J, Harvey JH, Spigel DR, Dediu M, Reck M, Cebotaru CL, Humphreys RC, Gribbin MJ, Fox NL, Camidge DR. Phase II trial of mapatumumab, a fully human agonist monoclonal antibody to tumor necrosis factor-related apoptosis-inducing ligand receptor 1 (TRAIL-R1), in combination with paclitaxel and carboplatin in patients with advanced non-small-cell lung cancer. Clin Lung Cancer 2013; 15:188-196.e2. [PMID: 24560012 DOI: 10.1016/j.cllc.2013.12.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 12/16/2013] [Accepted: 12/23/2013] [Indexed: 10/25/2022]
Abstract
BACKGROUND This phase II study examined the efficacy of mapatumumab in combination with paclitaxel and carboplatin in patients with non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS Patients with stage IIIB or stage IV advanced primary NSCLC were randomly assigned (1:1:1) to receive up to 6 courses of standard-dose paclitaxel and carboplatin or a combination of paclitaxel, carboplatin, and mapatumumab (10 mg/kg or 30 mg/kg). Primary efficacy end points were overall response rate and median progression-free survival (PFS). Secondary efficacy end points included disease control rate, overall survival (OS), time to response, and duration of response. Exploratory studies included evaluation of historical biopsy materials for TRAIL-R1 expression by immunohistochemical analysis and serum levels of M30, a marker of apoptosis, before and after the first 2 doses of mapatumumab. Safety parameters, including adverse events (AEs), laboratory tests, and immunogenicity, were assessed. RESULTS The majority of patients had stage IV disease (79%) and an Eastern Cooperative Oncology Group (ECOG) performance status of 0 (58%); baseline characteristics were similar across treatment arms. No improvements in response or disease control rates, PFS, or OS were gained from the addition of mapatumumab. Adverse events in the mapatumumab arms were generally consistent with toxicities seen in the carboplatin and paclitaxel control arm. Levels of M30 were highly variable, and consistent patterns were not seen across treatment arms. CONCLUSION This study showed no clinical benefit from adding mapatumumab to carboplatin and paclitaxel in unselected patients with NSCLC. The combination was generally well tolerated. The possibility of subgroups sensitive to mapatumumab is discussed.
Collapse
Affiliation(s)
- Joachim von Pawel
- Department of Oncology, Asklepios-Fachkliniken München-Gauting, Munich, Germany.
| | | | - David R Spigel
- Tennessee Oncology, Sarah Cannon Research Institute, Nashville, TN
| | - Mircea Dediu
- Department of Medical Oncology, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", Bucharest, Romania
| | - Martin Reck
- Department of Thoracic Oncology, Hospital Grosshansdorf, Grosshansdorf, Germany
| | - Cristina L Cebotaru
- Department of Radiotherapy I-Medical Oncology, Prof Dr Ion Chircuta Institute of Oncology, Cluj, Romania
| | | | | | | | - D Ross Camidge
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO
| |
Collapse
|
154
|
Holland PM. Death receptor agonist therapies for cancer, which is the right TRAIL? Cytokine Growth Factor Rev 2013; 25:185-93. [PMID: 24418173 DOI: 10.1016/j.cytogfr.2013.12.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/15/2013] [Indexed: 01/28/2023]
Abstract
The activation of cell-surface death receptors represents an attractive therapeutic strategy to promote apoptosis of tumor cells. Several investigational therapeutics that target this extrinsic pathway, including recombinant human Apo2L/TRAIL and monoclonal agonist antibodies directed against death receptors-4 (DR4) or -5 (DR5), have been evaluated in the clinic. Although Phase 1/1b studies provided encouraging preliminary results, findings from randomized Phase 2 studies failed to demonstrate significant clinical benefit. This has raised multiple questions as to why pre-clinical data were not predictive of clinical response. Results from clinical studies and insight into why current agents have failed to yield robust responses are discussed. In addition, new strategies for the development of next generation death receptor agonists are reviewed.
Collapse
Affiliation(s)
- Pamela M Holland
- Therapeutic Innovation Unit, Amgen Inc., 360 Binney Street, Cambridge, MA 02142, United States.
| |
Collapse
|
155
|
Reck M, Krzakowski M, Chmielowska E, Sebastian M, Hadler D, Fox T, Wang Q, Greenberg J, Beckman RA, von Pawel J. A randomized, double-blind, placebo-controlled phase 2 study of tigatuzumab (CS-1008) in combination with carboplatin/paclitaxel in patients with chemotherapy-naïve metastatic/unresectable non-small cell lung cancer. Lung Cancer 2013; 82:441-8. [DOI: 10.1016/j.lungcan.2013.09.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 09/18/2013] [Accepted: 09/23/2013] [Indexed: 01/21/2023]
|
156
|
Tatsuta T, Hosono M, Takahashi K, Omoto T, Kariya Y, Sugawara S, Hakomori S, Nitta K. Sialic acid-binding lectin (leczyme) induces apoptosis to malignant mesothelioma and exerts synergistic antitumor effects with TRAIL. Int J Oncol 2013; 44:377-84. [PMID: 24297392 PMCID: PMC3898873 DOI: 10.3892/ijo.2013.2192] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/17/2013] [Indexed: 11/06/2022] Open
Abstract
Malignant mesothelioma is a highly aggressive tumor with poor prognosis. An effective drug for treatment of malignant mesothelioma is greatly needed. Sialic acid-binding lectin (SBL) isolated from oocytes of Rana catesbeiana is a multifunctional protein which has lectin activity, ribonuclease activity and antitumor activity, so it could be developed as a new type of anticancer drug. The validity of SBL for treatment of malignant mesothelioma was assessed using three malignant mesotheliomas and a non-malignant mesothlial cell line. Effectiveness of combinatorial treatment of SBL and tumor necrosis factor-related apoptosis inducing ligand (TRAIL) was also elucidated and characterized. SBL induced tumor-selective cytotoxicity that was attributed to induction of apoptosis. Combinatorial treatment of SBL and TRAIL showed synergistic apoptosis-inducing effect. Additional experiments revealed that Bid was the mediating molecule for the synergistic effect in SBL and TRAIL. These results suggested that SBL could be a promising candidate for the therapeutics for malignant mesothelioma. Furthermore, the combinatorial treatment of SBL and TRAIL could be an effective regimen against malignant mesothelioma.
Collapse
Affiliation(s)
- Takeo Tatsuta
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Aoba-ku, Sendai 981-8558, Japan
| | - Masahiro Hosono
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Aoba-ku, Sendai 981-8558, Japan
| | - Kohta Takahashi
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Aoba-ku, Sendai 981-8558, Japan
| | - Takashi Omoto
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Aoba-ku, Sendai 981-8558, Japan
| | - Yukiko Kariya
- Fukushima Medical University, Fukushima 960-1295, Japan
| | - Shigeki Sugawara
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Aoba-ku, Sendai 981-8558, Japan
| | - Senitiroh Hakomori
- Division of Biomembrane Research, Pacific Northwest Research Institute, WA 98122, USA
| | - Kazuo Nitta
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Aoba-ku, Sendai 981-8558, Japan
| |
Collapse
|
157
|
Koyama M, Sowa Y, Horinaka M, Goda AE, Fujiwara J, Sakai T. Peroxisome proliferator-activated receptor γ ligand troglitazone and TRAIL synergistically induce apoptosis. Oncol Rep 2013; 31:947-54. [PMID: 24276615 DOI: 10.3892/or.2013.2868] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 10/25/2013] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is known to cause apoptosis in several types of malignant tumor cells through its interaction with the death domain-containing receptor, death receptor 5 (DR5). In the present study, we showed that co-treatment with troglitazone (TGZ), a synthetic ligand of peroxisome proliferator-activated receptor γ (PPARγ), and TRAIL synergistically induced apoptosis through DR5 upregulation in human colon cancer DLD-1 cells. TGZ elevated DR5 expression at the promoter level through the CCAAT/enhancer-binding protein homologous protein (CHOP) binding site. These results suggest that combined treatment with TGZ and TRAIL may be promising as a new therapy against malignant tumors.
Collapse
Affiliation(s)
- Makoto Koyama
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yoshihiro Sowa
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Mano Horinaka
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Ahmed E Goda
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Jun Fujiwara
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Toshiyuki Sakai
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
158
|
Quast SA, Berger A, Plötz M, Eberle J. Sensitization of melanoma cells for TRAIL-induced apoptosis by activation of mitochondrial pathways via Bax. Eur J Cell Biol 2013; 93:42-8. [PMID: 24361324 DOI: 10.1016/j.ejcb.2013.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/15/2013] [Accepted: 11/15/2013] [Indexed: 12/13/2022] Open
Abstract
The death ligand TRAIL (TNF-related apoptosis-inducing ligand) represents a promising therapeutic strategy for metastatic melanoma, however prevalent and inducible resistance limits its applicability and therapeutic use. Recent work has revealed that combinations with survival pathway inhibitors could efficiently sensitize melanoma cells for TRAIL. Here, a particular role was attributed to the activation of Bax, which is regulated by phosphorylation. Thus, TRAIL resistance in melanoma is explained by three major steps, namely high levels of antiapoptotic Bcl-2 proteins, high levels of inhibitor of apoptosis proteins (cIAPs) and suppressed Bax activity. Importantly, Bid was activated in response to TRAIL alone also in resistant cells to antagonize Bcl-2, and Bax was activated in response to pathway inhibitors. However, only in combinations, mitochondrial apoptosis pathways were opened to result in release of Smac/DIABLO, which functions as antagonist of cIAPs. Opening the caspase cascade by Smac then allowed efficient induction of apoptosis. Thus, direct or indirect targeting of Bax represents a suitable strategy to overcome TRAIL resistance in melanoma and may allow the establishment of TRAIL-based therapeutic approaches.
Collapse
Affiliation(s)
- Sandra-Annika Quast
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | - Anja Berger
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | - Michael Plötz
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | - Jürgen Eberle
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany.
| |
Collapse
|
159
|
De Luca A, D'Alessio A, Gallo M, Maiello MR, Bode AM, Normanno N. Src and CXCR4 are involved in the invasiveness of breast cancer cells with acquired resistance to lapatinib. Cell Cycle 2013; 13:148-56. [PMID: 24200972 DOI: 10.4161/cc.26899] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Lapatinib is a dual EGFR and ErbB-2 tyrosine kinase inhibitor that has significantly improved the clinical outcome of ErbB-2-overexpressing breast cancer patients. However, patients inexorably develop mechanisms of resistance that limit the efficacy of the drug. In order to identify potential targets for therapeutic intervention in lapatinib-resistant patients, we isolated, from ErbB-2-overexpressing SK-Br-3 breast cancer cells, the SK-Br-3 Lap-R-resistant subclone, which is able to routinely grow in 1 µM lapatinib. Resistant cells have a more aggressive phenotype compared with parental cells, as they show a higher ability to invade through a matrigel-coated membrane. Lapatinib-resistant cells have an increased Src kinase activity and persistent levels of activation of ERK1/2 and AKT compared with parental cells. Treatment with the Src inhibitor saracatinib in combination with lapatinib reduces AKT and ERK1/2 phosphorylation and restores the sensitivity of resistant cells to lapatinib. SK-Br-3 Lap-R cells also show levels of expression of CXCR4 that are higher compared with parental cells and are not affected by Src inhibition. Treatment with saracatinib or a specific CXCR4 antibody reduces the invasive ability of SK-Br-3 Lap-R cells, with the two drugs showing cooperative effects. Finally, blockade of Src signaling significantly increases TRAIL-induced cell death in SK-Br-3 Lap-R cells. Taken together, our results demonstrate that breast cancer cells with acquired resistance to lapatinib have a more aggressive phenotype compared with their parental counterpart, and that Src signaling and CXCR4 play an important role in this phenomenon, thus representing potential targets for therapeutic intervention in lapatinib-resistant breast cancer patients.
Collapse
Affiliation(s)
- Antonella De Luca
- Cell Biology and Biotherapy Unit; Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"; IRCCS; Naples, Italy
| | - Amelia D'Alessio
- Cell Biology and Biotherapy Unit; Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"; IRCCS; Naples, Italy
| | - Marianna Gallo
- Cell Biology and Biotherapy Unit; Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"; IRCCS; Naples, Italy
| | - Monica R Maiello
- Cell Biology and Biotherapy Unit; Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"; IRCCS; Naples, Italy
| | - Ann M Bode
- The Hormel Institute; University of Minnesota; Austin, MN USA
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit; Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"; IRCCS; Naples, Italy
| |
Collapse
|
160
|
Meijer A, Kruyt FAE, van der Zee AGJ, Hollema H, Le P, ten Hoor KA, Groothuis GMM, Quax WJ, de Vries EGE, de Jong S. Nutlin-3 preferentially sensitises wild-type p53-expressing cancer cells to DR5-selective TRAIL over rhTRAIL. Br J Cancer 2013; 109:2685-95. [PMID: 24136147 PMCID: PMC3833221 DOI: 10.1038/bjc.2013.636] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 09/20/2013] [Accepted: 09/24/2013] [Indexed: 12/13/2022] Open
Abstract
Background: Tumour cell-selective activation of apoptosis by recombinant human TNF-related apoptosis-inducing ligand (rhTRAIL) is enhanced through co-activation of p53 by chemotherapeutic drugs. The novel anticancer agent nutlin-3 provides a promising alternative for p53 activation by disrupting the interaction between p53 and its negative feedback regulator MDM2. Methods: We examined whether nutlin-3 enhances apoptosis induction by rhTRAIL and the DR5-selective TRAIL variant D269H/E195R in wild-type p53-expressing ovarian, colon and lung cancer cell lines and in an ex vivo model of human ovarian cancer. Results: Nutlin-3 enhanced p53, p21, MDM2 and DR5 surface expression. Although nutlin-3 did not induce apoptosis, it preferentially enhanced D269H/E195R-induced apoptosis over rhTRAIL. Combination treatment potentiated the cleavage of caspases 8, 9, 3 and PARP. P53 and MDM2 siRNA experiments showed that this enhanced apoptotic effect was mediated by wild-type p53. Indeed, nutlin-3 did not enhance rhTRAIL-induced apoptosis in OVCAR-3 cells harbouring mutant p53. Addition of the chemotherapeutic drug cisplatin to the combination further increased p53 and DR5 levels and rhTRAIL- and D269H/E195R-induced apoptosis. As a proof of concept, we show that the combination of D269H/E195R, nutlin-3 and cisplatin induced massive apoptosis in ex vivo tissue slices of primary human ovarian cancers. Conclusion: Nutlin-3 is a potent enhancer of D269H/E195R-induced apoptosis in wild-type p53-expressing cancer cells. Addition of DNA-damaging agents such as cisplatin further enhances DR5-mediated apoptosis.
Collapse
Affiliation(s)
- A Meijer
- Department of Medical Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
161
|
ROS-dependent phosphorylation of Bax by wortmannin sensitizes melanoma cells for TRAIL-induced apoptosis. Cell Death Dis 2013; 4:e839. [PMID: 24113173 PMCID: PMC3824654 DOI: 10.1038/cddis.2013.344] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 07/26/2013] [Accepted: 08/09/2013] [Indexed: 02/08/2023]
Abstract
The pathways of reactive oxygen species (ROS)-mediated apoptosis induction, of Bax activation and the sensitization of tumor cells for TRAIL (TNF-related apoptosis-inducing ligand)-induced apoptosis are still largely elusive. Here, sensitization of melanoma cells for TRAIL by the PI3-kinase inhibitor wortmannin correlated to the activation of mitochondrial apoptosis pathways. Apoptosis was dependent on Bax and abrogated by Bcl-2 overexpression. The synergistic enhancement was explained by Bax activation through wortmannin, which tightly correlated to the characteristic Bax phosphorylation patterns. Thus, wortmannin resulted in early reduction of the Bax-inactivating phosphorylation at serine-184, whereas the Bax-activating phosphorylation at threonine-167 was enhanced. Proving the responsibility of the pathway, comparable effects were obtained with an Akt inhibitor (MK-2206); while suppressed phosphorylation of serine-184 may be attributed to reduced Akt activity itself, the causes of enhanced threonine-167 phosphorylation were addressed here. Characteristically, production of ROS was seen early in response to wortmannin and MK-2206. Providing the link between ROS and Bax, we show that abrogated ROS production by α-tocopherol or by NADPH oxidase 4 (NOX4) siRNA suppressed apoptosis and Bax activation. This correlated with reduced Bax phosphorylation at threonine-167. The data unraveled a mechanism by which NOX4-dependent ROS production controls apoptosis via Bax phosphorylation. The pathway may be considered for proapoptotic, anticancer strategies.
Collapse
|
162
|
Gieffers C, Kluge M, Merz C, Sykora J, Thiemann M, Schaal R, Fischer C, Branschädel M, Abhari BA, Hohenberger P, Fulda S, Fricke H, Hill O. APG350 induces superior clustering of TRAIL receptors and shows therapeutic antitumor efficacy independent of cross-linking via Fcγ receptors. Mol Cancer Ther 2013; 12:2735-47. [PMID: 24101228 DOI: 10.1158/1535-7163.mct-13-0323] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer cells can be specifically driven into apoptosis by activating Death-receptor-4 (DR4; TRAIL-R1) and/or Death-receptor-5 (DR5; TRAIL-R2). Albeit showing promising preclinical efficacy, first-generation protein therapeutics addressing this pathway, especially agonistic anti-DR4/DR5-monoclonal antibodies, have not been clinically successful to date. Due to their bivalent binding mode, effective apoptosis induction by agonistic TRAIL-R antibodies is achieved only upon additional events leading to antibody-multimer formation. The binding of these multimers to their target subsequently leads to effective receptor-clustering on cancer cells. The research results presented here report on a new class of TRAIL-receptor agonists overcoming this intrinsic limitation observed for antibodies in general. The main feature of these agonists is a TRAIL-mimic consisting of three TRAIL-protomer subsequences combined in one polypeptide chain, termed the single-chain TRAIL-receptor-binding domain (scTRAIL-RBD). In the active compounds, two scTRAIL-RBDs with three receptor binding sites each are brought molecularly in close proximity resulting in a fusion protein with a hexavalent binding mode. In the case of APG350-the prototype of this engineering concept-this is achieved by fusing the Fc-part of a human immunoglobulin G1 (IgG1)-mutein C-terminally to the scTRAIL-RBD polypeptide, thereby creating six receptor binding sites per drug molecule. In vitro, APG350 is a potent inducer of apoptosis on human tumor cell lines and primary tumor cells. In vivo, treatment of mice bearing Colo205-xenograft tumors with APG350 showed a dose-dependent antitumor efficacy. By dedicated muteins, we confirmed that the observed in vivo efficacy of the hexavalent scTRAIL-RBD fusion proteins is-in contrast to agonistic antibodies-independent of FcγR-based cross-linking events.
Collapse
Affiliation(s)
- Christian Gieffers
- Corresponding Author: Oliver Hill, Apogenix GmbH, Im Neuenheimer Feld 584, Heidelberg 69120, Germany. Phone: 49-6221-58608-18; Fax: 49-6221-58608-10; E-Mail:
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Seifert O, Plappert A, Fellermeier S, Siegemund M, Pfizenmaier K, Kontermann RE. Tetravalent antibody-scTRAIL fusion proteins with improved properties. Mol Cancer Ther 2013; 13:101-11. [PMID: 24092811 DOI: 10.1158/1535-7163.mct-13-0396] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We applied the immunoglobulin E (IgE) heavy-chain domain 2 (EHD2) as the covalently linked homodimerization module to generate antibody-scTRAIL fusion proteins. By fusing a humanized single-chain fragment variable (scFv) directed against EGFR to the N-terminus of the EHD2 and a single-chain derivative of TRAIL (scTRAIL) to the C-terminus of the EHD2, we produced a dimeric, tetravalent fusion protein. The fusion protein retained its binding activity for EGFR and TRAIL receptors. In vitro, the targeted antibody-scTRAIL fusion protein exhibited an approximately 8- to 18-fold increased cytotoxic activity compared with the untargeted EHD2-scTRAIL fusion protein. This resulted in increased antitumor activity in a subcutaneous Colo205 xenograft tumor murine model. In summary, the scFv-EHD2-scTRAIL fusion protein combines target cell selectivity with an increased TRAIL activity leading to improved antitumor activities.
Collapse
Affiliation(s)
- Oliver Seifert
- Corresponding Author: Roland E. Kontermann, Institut für Zellbiologie und Immunologie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany.
| | | | | | | | | | | |
Collapse
|
164
|
Fuchs CS, Fakih M, Schwartzberg L, Cohn AL, Yee L, Dreisbach L, Kozloff MF, Hei YJ, Galimi F, Pan Y, Haddad V, Hsu CP, Sabin A, Saltz L. TRAIL receptor agonist conatumumab with modified FOLFOX6 plus bevacizumab for first-line treatment of metastatic colorectal cancer: A randomized phase 1b/2 trial. Cancer 2013; 119:4290-8. [PMID: 24122767 DOI: 10.1002/cncr.28353] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 06/24/2013] [Accepted: 07/15/2013] [Indexed: 01/26/2023]
Abstract
BACKGROUND In patients with previously untreated metastatic colorectal cancer (mCRC), we conducted a phase 1b/randomized phase 2 trial to define the safety, tolerability, and efficacy of mFOLFOX6 plus bevacizumab (mFOLFOX6/bev) with conatumumab, an investigational, fully human monoclonal IgG1 antibody that specifically activates death receptor 5 (DR5). METHODS Twelve patients were enrolled in a phase 1b open-label dose-escalation trial of conatumumab with mFOLFOX6/bev; thereafter, 190 patients were randomized 1:1:1 to receive mFOLFOX6/bev in combination with 2 mg/kg conatumumab, 10 mg/kg conatumumab, or placebo. Therapy cycles were repeated every 2 weeks until disease progression or the occurrence of unacceptable toxicity. RESULTS In phase 1b, conatumumab with mFOLFOX6/bev was tolerated without apparent added toxicity over mFOLFOX6/bev alone. In phase 2, conatumumab with mFOLFOX6/bev did not confer a benefit in progression-free survival when compared with placebo with mFOLFOX6/bev. Toxicity was similar in all treatment arms. Following treatment, similar increases in circulating caspase-3 levels were observed in all arms. CONCLUSIONS Conatumumab with mFOLFOX6/bev did not offer improved efficacy over the same chemotherapy with placebo in first-line treatment of patients with mCRC. These data do not support further development of conatumumab in advanced CRC.
Collapse
|
165
|
RAF inhibition overcomes resistance to TRAIL-induced apoptosis in melanoma cells. J Invest Dermatol 2013; 134:430-440. [PMID: 23955071 DOI: 10.1038/jid.2013.347] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 05/14/2013] [Accepted: 05/26/2013] [Indexed: 12/29/2022]
Abstract
Mutated BRAF represents a critical oncogene in melanoma, and selective inhibitors have been approved for melanoma therapy. However, the molecular consequences of RAF inhibition in melanoma cells remained largely elusive. Here, we investigated the effects of the pan-RAF inhibitor L-779,450, which inhibited cell proliferation both in BRAF-mutated and wild-type melanoma cell lines. It furthermore enhanced apoptosis in combination with the death ligand tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and overcame TRAIL resistance in melanoma cells. Enhanced apoptosis coincided with activation of mitochondrial pathways, seen by loss of mitochondrial membrane potential and release of cytochrome c, Smac (second mitochondria-derived activator of caspases), and apoptosis-inducing factor (AIF). Subsequently, caspase-9 and -3 were activated. Apoptosis induction by L-779,450/TRAIL was prevented by Bcl-2 overexpression and was dependent on Bax. Thus, activation of Bax by L-779,450 alone was demonstrated by Bax conformational changes, whereas Bak was not activated. Furthermore, the BH3-only protein Bim was upregulated in response to L-779,450. The significant roles of Smac, Bax, and Bim in this setting were proven by small interfering RNA (siRNA)-mediated knockdown experiments. L-779,450 also resulted in morphological changes indicating autophagy confirmed by the autophagy marker light chain 3-II (LC3-II). The pro-apoptotic effects of L-779,450 may explain the antitumor effects of RAF inhibition and may be considered when evaluating RAF inhibitors for melanoma therapy.
Collapse
|
166
|
Micheau O, Shirley S, Dufour F. Death receptors as targets in cancer. Br J Pharmacol 2013; 169:1723-44. [PMID: 23638798 PMCID: PMC3753832 DOI: 10.1111/bph.12238] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 04/25/2013] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Anti-tumour therapies based on the use pro-apoptotic receptor agonists, including TNF-related apoptosis-inducing ligand (TRAIL) or monoclonal antibodies targeting TRAIL-R1 or TRAIL-R2, have been disappointing so far, despite clear evidence of clinical activity and lack of adverse events for the vast majority of these compounds, whether combined or not with conventional or targeted anti-cancer therapies. This brief review aims at discussing the possible reasons for the lack of apparent success of these therapeutic approaches and at providing hints in order to rationally design optimal protocols based on our current understanding of TRAIL signalling regulation or resistance for future clinical trials. LINKED ARTICLES This article is part of a themed section on Emerging Therapeutic Aspects in Oncology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.169.issue-8.
Collapse
|
167
|
A randomized phase 2 study of paclitaxel and carboplatin with or without conatumumab for first-line treatment of advanced non-small-cell lung cancer. J Thorac Oncol 2013; 8:329-37. [PMID: 23370314 DOI: 10.1097/jto.0b013e31827ce554] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION This study evaluated the efficacy, safety, and pharmacokinetics of conatumumab combined with paclitaxel-carboplatin (PC) as first-line treatment for advanced non-small-cell lung cancer (NSCLC). METHODS Patients (aged >18 years) with previously untreated advanced or recurrent NSCLC were randomized 1:1:1 (stratified by Eastern Cooperative Oncology Group performance status and disease stage) to receive up to six 3-week cycles of PC combined with conatumumab (arm 1, 3 mg/kg; arm 2, 15 mg/kg) or placebo (arm 3) every 3 weeks. The primary endpoint was progression-free survival (PFS). This study is registered with ClinicalTrials.gov (NCT00534027). RESULTS Between August 8, 2007 and April 9, 2009, 172 patients were randomized (arm 1, n = 57; arm 2, n = 56; arm 3, n = 59). Median PFS was 5.4 months (95% confidence interval [CI] 4.1-6.3) in arm 1 (hazard ratio [HR] 0.84 [95% CI 0.57-1.24]; p = 0.41), 4.8 months (95% CI 3.2-6.5) in arm 2 (HR 0.93 [0.64-1.35]; p = 0.57), and 5.5 months (95% CI 4.3-5.7) in arm 3. There was an interaction between tumor histology and the effect of conatumumab on PFS (squamous HR 0.47 [0.23-0.94]; nonsquamous HR 1.08 [0.74-1.57]; interaction p = 0.039).The most common grade of three or more adverse events were neutropenia, anemia, and thrombocytopenia. There was no evidence of pharmacokinetic interactions between conatumumab and PC. Of 158 patients assessable for FCGR3A polymorphisms, conatumumab treatment was associated with a trend toward longer overall survival (HR 0.72 [0.43-1.23]) among V-allele carriers (V/V or F/V; n = 54) but not among F-allele homozygotes (n = 34; HR 1.37 [0.66-2.86]). CONCLUSION Although well tolerated, the addition of conatumumab to PC did not improve outcomes in unselected patients with previously untreated advanced NSCLC.
Collapse
|
168
|
Farhat FS, Houhou W. Targeted therapies in non-small cell lung carcinoma: what have we achieved so far? Ther Adv Med Oncol 2013; 5:249-70. [PMID: 23858333 PMCID: PMC3707340 DOI: 10.1177/1758834013492001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The search for innovative therapeutic agents in non-small cell lung cancer (NSCLC) has witnessed a swift evolution. The number of targeted drugs that can improve patient outcomes with an acceptable safety profile is steadily increasing. In this review, we highlight current drugs that have already been approved or are under evaluation for the treatment of patients with NSCLC, either in monotherapy or combined therapy for both the first- and second-line settings. Experience with drugs targeting the vascular endothelial growth factor and its receptor, as well as the epidermal growth factor receptor is summarized. Moreover, we provide an overview of more novel targets in NSCLC and initial experience with the respective therapeutic agents.
Collapse
Affiliation(s)
- Fadi S Farhat
- Hematology-Oncology Division (Head), Hammoud Hospital University Medical Center, Ghassan Hammoud Street, 652, Saida, Lebanon
| | | |
Collapse
|
169
|
Cohn AL, Tabernero J, Maurel J, Nowara E, Sastre J, Chuah BYS, Kopp MV, Sakaeva DD, Mitchell EP, Dubey S, Suzuki S, Hei YJ, Galimi F, McCaffery I, Pan Y, Loberg R, Cottrell S, Choo SP. A randomized, placebo-controlled phase 2 study of ganitumab or conatumumab in combination with FOLFIRI for second-line treatment of mutant KRAS metastatic colorectal cancer. Ann Oncol 2013; 24:1777-1785. [PMID: 23510984 DOI: 10.1093/annonc/mdt057] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Targeted agents presently available for mutant KRAS metastatic colorectal cancer (mCRC) are bevacizumab and aflibercept. We evaluated the efficacy and safety of conatumumab (an agonistic monoclonal antibody against human death receptor 5) and ganitumab (a monoclonal antibody against the type 1 insulin-like growth factor receptor) combined with standard FOLFIRI chemotherapy as a second-line treatment in patients with mutant KRAS mCRC. PATIENTS AND METHODS Patients with mutant KRAS metastatic adenocarcinoma of the colon or rectum refractory to fluoropyrimidine- and oxaliplatin-based chemotherapy were randomized 1 : 1 : 1 to receive intravenous FOLFIRI plus conatumumab 10 mg/kg (Arm A), ganitumab 12 mg/kg (Arm B), or placebo (Arm C) Q2W. The primary end point was progression-free survival (PFS). RESULTS In total, 155 patients were randomized. Median PFS in Arms A, B, and C was 6.5 months (HR, 0.69; P = 0.147), 4.5 months (HR, 1.01; P = 0.998), and 4.6 months, respectively; median overall survival was 12.3 months (HR, 0.89; P = 0.650), 12.4 months (HR, 1.27; P = 0.357), and 12.0 months; and objective response rate was 14%, 8%, and 2%. The most common grade ≥3 adverse events in Arms A/B/C included neutropenia (30%/25%/18%) and diarrhea (18%/2%/10%). CONCLUSIONS Conatumumab, but not ganitumab, plus FOLFIRI was associated with a trend toward improved PFS. Both combinations had acceptable toxicity.
Collapse
Affiliation(s)
- A L Cohn
- Rocky Mountain Cancer Center, Denver, USA.
| | - J Tabernero
- Medical Oncology Department, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona
| | - J Maurel
- Medical Oncology Department, Hospital Clinic de Barcelona, Barcelona, Spain
| | - E Nowara
- Maria Skodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - J Sastre
- Hospital Clinico San Carlos, Servicio de Oncologíca Medíca, Madrid, and Instituto Carlos III, Spanish Ministry of Science and Innovation, Madrid, Spain
| | - B Y S Chuah
- Department of Internal Medicine, National University Hospital, Singapore, Singapore
| | - M V Kopp
- Samara Regional Oncology Dispensary, Samara
| | - D D Sakaeva
- Clinical Oncology Dispensary of the Republic of Bashkortostan, Ufa, Russia
| | - E P Mitchell
- Department of Medical Oncology, Thomas Jefferson University Hospital, Philadelphia
| | - S Dubey
- Amgen Inc., South San Francisco
| | | | | | | | | | | | | | | | - S-P Choo
- Medical Oncology, National Cancer Centre Singapore, Singapore
| |
Collapse
|
170
|
Targeting of the tumor necrosis factor receptor superfamily for cancer immunotherapy. ISRN ONCOLOGY 2013; 2013:371854. [PMID: 23840967 PMCID: PMC3693168 DOI: 10.1155/2013/371854] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 05/11/2013] [Indexed: 12/17/2022]
Abstract
The tumor necrosis factor (TNF) ligand and cognate TNF receptor superfamilies constitute an important regulatory axis that is pivotal for immune homeostasis and correct execution of immune responses. TNF ligands and receptors are involved in diverse biological processes ranging from the selective induction of cell death in potentially dangerous and superfluous cells to providing costimulatory signals that help mount an effective immune response. This diverse and important regulatory role in immunity has sparked great interest in the development of TNFL/TNFR-targeted cancer immunotherapeutics. In this review, I will discuss the biology of the most prominent proapoptotic and co-stimulatory TNF ligands and review their current status in cancer immunotherapy.
Collapse
|
171
|
Swers JS, Grinberg L, Wang L, Feng H, Lekstrom K, Carrasco R, Xiao Z, Inigo I, Leow CC, Wu H, Tice DA, Baca M. Multivalent scaffold proteins as superagonists of TRAIL receptor 2-induced apoptosis. Mol Cancer Ther 2013; 12:1235-44. [PMID: 23645592 DOI: 10.1158/1535-7163.mct-12-1107] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Activation of TNF-related apoptosis-inducing ligand receptor 2 (TRAILR2) can induce apoptosis in a variety of human cancer cell lines and xenografts, while lacking toxicity in normal cells. The natural ligand and agonistic antibodies show antitumor activity in preclinical models of cancer, and this had led to significant excitement in the clinical potential of these agents. Unfortunately, this optimism has been tempered by trial data that, thus far, are not showing clear signs of efficacy in cancer patients. The reasons for discrepant preclinical and clinical observations are not understood, but one possibility is that the current TRAILR2 agonists lack sufficient potency to achieve a meaningful response in patients. Toward addressing that possibility, we have developed multivalent forms of a new binding scaffold (Tn3) that are superagonists of TRAILR2 and can induce apoptosis in tumor cell lines at subpicomolar concentrations. The monomer Tn3 unit was a fibronectin type III domain engineered for high-affinity TRAILR2 binding. Multivalent presentation of this basic unit induced cell death in TRAILR2-expressing cell lines. Optimization of binding affinity, molecular format, and valency contributed to cumulative enhancements of agonistic activity. An optimized multivalent agonist consisting of 8 tandem Tn3 repeats was highly potent in triggering cell death in TRAIL-sensitive cell lines and was 1 to 2 orders of magnitude more potent than TRAIL. Enhanced potency was also observed in vivo in a tumor xenograft setting. The TRAILR2 superagonists described here have the potential for superior clinical activity in settings insensitive to the current therapeutic agonists that target this pathway.
Collapse
Affiliation(s)
- Jeffery S Swers
- Department of Antibody Discovery and Protein Engineering, MedImmune, LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
den Hollander MW, Gietema JA, de Jong S, Walenkamp AM, Reyners AK, Oldenhuis CN, de Vries EG. Translating TRAIL-receptor targeting agents to the clinic. Cancer Lett 2013; 332:194-201. [DOI: 10.1016/j.canlet.2012.04.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 04/11/2012] [Accepted: 04/15/2012] [Indexed: 12/21/2022]
|
173
|
Wang W, He W, Wang L, Zhang G, Gao B. Pentamerisation of a scFv directed against TRAIL receptor 2 increases its antitumour efficacy. Immunol Cell Biol 2013; 91:360-7. [DOI: 10.1038/icb.2013.10] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Wei Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Institute of Microbiology, Chinese Academy of SciencesBeijingPRC
- School of Life Sciences, University of Science and Technology of ChinaHefeiPRC
| | - Wen He
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Institute of Microbiology, Chinese Academy of SciencesBeijingPRC
- Hebei Key Laboratory of Medical Biotechnology, Biochemistry Teaching and Research Office, Hebei Medical UniversityShijiazhuangPRC
- Graduate University of Chinese Academy of SciencesBeijingPRC
| | - Lei Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Institute of Microbiology, Chinese Academy of SciencesBeijingPRC
- Graduate University of Chinese Academy of SciencesBeijingPRC
| | - Ge Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Institute of Microbiology, Chinese Academy of SciencesBeijingPRC
- Graduate University of Chinese Academy of SciencesBeijingPRC
| | - Bin Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Institute of Microbiology, Chinese Academy of SciencesBeijingPRC
- School of Life Sciences, University of Science and Technology of ChinaHefeiPRC
- Graduate University of Chinese Academy of SciencesBeijingPRC
| |
Collapse
|
174
|
Pennarun B, Kleibeuker JH, Boersma-van Ek W, Kruyt FAE, Hollema H, de Vries EGE, de Jong S. Targeting FLIP and Mcl-1 using a combination of aspirin and sorafenib sensitizes colon cancer cells to TRAIL. J Pathol 2013; 229:410-21. [PMID: 23132258 DOI: 10.1002/path.4138] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 10/15/2012] [Accepted: 10/19/2012] [Indexed: 12/16/2022]
Abstract
The multikinase inhibitor sorafenib is highly effective against certain types of cancer in the clinic and prevents colon cancer cell proliferation in vitro. Non-steroidal anti-inflammatory drugs, such as acetylsalicylic acid (aspirin), have shown activity against colon cancer cells. The aims of this study were to determine whether the combination of aspirin with sorafenib has enhanced anti-proliferative effects and increases recombinant human tumour necrosis factor-related apoptosis-inducing ligand (rhTRAIL)-induced apoptosis in the human SW948, Lovo, Colo205, Colo320, Caco-2 and HCT116 colon cancer cell lines. In four cell lines, aspirin strongly stimulated the anti-proliferative effects of sorafenib (∼four-fold enhancement) by inducing cell cycle arrest. Furthermore, combining low doses of aspirin (≤ 5 mm) and sorafenib (≤ 2.5 µm) greatly sensitized TRAIL-sensitive and TRAIL-resistant colon cancer cells to rhTRAIL, much more potently than either drug combined with rhTRAIL. The increase in rhTRAIL sensitivity was due to inhibition of FLIP and Mcl-1 protein expression following aspirin and sorafenib co-treatment, as confirmed by knock-down studies. Next, the clinical relevance of targeting FLIP and Mcl-1 in colon cancer was examined. Using immunohistochemistry, we found that Mcl-1 expression was significantly increased in colon adenoma and carcinoma patient material compared to healthy colonic epithelium, similar to the enhanced FLIP expression we recently observed in colon cancer. These results underscore the potential of combining low doses of aspirin with sorafenib to inhibit proliferation and target the anti-apoptotic proteins FLIP and Mcl-1 in colon cancer cells.
Collapse
Affiliation(s)
- Bodvael Pennarun
- Department of Medical Oncology, University of Groningen, University Medical Centre Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
175
|
Dimberg LY, Anderson CK, Camidge R, Behbakht K, Thorburn A, Ford HL. On the TRAIL to successful cancer therapy? Predicting and counteracting resistance against TRAIL-based therapeutics. Oncogene 2013; 32:1341-50. [PMID: 22580613 PMCID: PMC4502956 DOI: 10.1038/onc.2012.164] [Citation(s) in RCA: 220] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 03/19/2012] [Accepted: 03/21/2012] [Indexed: 12/11/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and agonistic antibodies against TRAIL death receptors (DR) kill tumor cells while causing virtually no damage to normal cells. Several novel drugs targeting TRAIL receptors are currently in clinical trials. However, TRAIL resistance is a common obstacle in TRAIL-based therapy and limits the efficiency of these drugs. In this review article we discuss different mechanisms of TRAIL resistance, and how they can be predicted and therapeutically circumvented. In addition, we provide a brief overview of all TRAIL-based clinical trials conducted so far. It is apparent that although the effects of TRAIL therapy are disappointingly modest overall, a small subset of patients responds very well to TRAIL. We argue that the true potential of targeting TRAIL DRs in cancer can only be reached when we find efficient ways to select for those patients that are most likely to benefit from the treatment. To achieve this, it is crucial to identify biomarkers that can help us predict TRAIL sensitivity.
Collapse
Affiliation(s)
- L Y Dimberg
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | | | | | | | | | | |
Collapse
|
176
|
Kaminskyy VO, Surova OV, Piskunova T, Zborovskaya IB, Tchevkina EM, Andera L, Zhivotovsky B. Upregulation of c-FLIP-short in response to TRAIL promotes survival of NSCLC cells, which could be suppressed by inhibition of Ca2+/calmodulin signaling. Cell Death Dis 2013; 4:e522. [PMID: 23470529 PMCID: PMC3613829 DOI: 10.1038/cddis.2013.51] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) is a promising cytokine for killing tumor cells. However, a number of studies have demonstrated that different cancer cells resist TRAIL treatment and, moreover, TRAIL can promote invasion and metastasis in resistant cells. Here we report that TRAIL rapidly activates caspase-8 in a panel of non-small-cell lung carcinomas (NSCLCs). Adenocarcinomas derived from the lung in addition to high caspase-8 expression are characterized by increased expression of DR4 compared with adjacent non-neoplastic tissues. Blocking DR4 or lowering caspase-8 expression significantly reduced apoptosis in NSCLC cell lines, indicating the importance of DR4 and signifying that higher levels of caspase-8 in lung adenocarcinomas make them more susceptible to TRAIL treatment. Despite rapid and robust initial responsiveness to TRAIL, surviving cells quickly acquired resistance to the additional TRAIL treatment. The expression of cellular-FLIP-short (c-FLIPS) was significantly increased in surviving cells. Such upregulation of c-FLIPS was rapidly reduced and TRAIL sensitivity was restored by treatment with cycloheximide. Silencing of c-FLIPS, but not c-FLIP-long (c-FLIPL), resulted in a remarkable increase in apoptosis and significant reduction of clonogenic survival. Furthermore, chelation of intracellular Ca2+ or inhibition of calmodulin caused a rapid proteasomal degradation of c-FLIPS, a significant increase of the two-step processing of procaspase-8, and reduced clonogenicity in response to TRAIL. Thus, our results revealed that the upregulation of DR4 and caspase-8 expression in NSCLC cells make them more susceptible to TRAIL. However, these cells could survive TRAIL treatment via upregulation of c-FLIPS, and it is suggested that blocking c-FLIPS expression by inhibition of Ca2+/calmodulin signaling significantly overcomes the acquired resistance of NSCLC cells to TRAIL.
Collapse
Affiliation(s)
- V O Kaminskyy
- Institute of Environmental Medicine, Division of Toxicology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
177
|
Gallmeier E, Bader DC, Kriegl L, Berezowska S, Seeliger H, Göke B, Kirchner T, Bruns C, De Toni EN. Loss of TRAIL-receptors is a recurrent feature in pancreatic cancer and determines the prognosis of patients with no nodal metastasis after surgery. PLoS One 2013; 8:e56760. [PMID: 23460812 PMCID: PMC3584093 DOI: 10.1371/journal.pone.0056760] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 01/14/2013] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Agonistic antibodies targeting TRAIL-receptors 1 and 2 (TRAIL-R1 and TRAIL-R2) are being developed as a novel therapeutic approach in cancer therapy including pancreatic cancer. However, the cellular distribution of these receptors in primary pancreatic cancer samples has not been sufficiently investigated and no study has yet addressed the issue of their prognostic significance in this tumor entity. AIMS AND METHODS Applying tissue microarray (TMA) analysis, we performed an immunohistochemical assessment of TRAIL-receptors in surgical samples from 84 consecutive patients affected by pancreatic adenocarcinoma and in 26 additional selected specimens from patients with no lymph nodes metastasis at the time of surgery. The prognostic significance of membrane staining and staining intensity for TRAIL-receptors was evaluated. RESULTS The fraction of pancreatic cancer samples with positive membrane staining for TRAIL-R1 and TRAIL-R2 was lower than that of cells from surrounding non-tumor tissues (TRAIL-R1: p<0.001, TRAIL-R2: p = 0.006). In addition, subgroup analyses showed that loss of membrane staining for TRAIL-R2 was associated with poorer prognosis in patients without nodal metastases (multivariate Cox regression analysis, Hazard Ratio: 0.44 [95% confidence interval: 0.22-0.87]; p = 0.019). In contrast, analysis of decoy receptors TRAIL-R3 and -R4 in tumor samples showed an exclusively cytoplasmatic staining pattern and no prognostic relevance. CONCLUSION This is a first report on the prognostic significance of TRAIL-receptors expression in pancreatic cancer showing that TRAIL-R2 might represent a prognostic marker for patients with early stage disease. In addition, our data suggest that loss of membrane-bound TRAIL-receptors could represent a molecular mechanism for therapeutic failure upon administration of TRAIL-receptors-targeting antibodies in pancreatic cancer. This hypothesis should be evaluated in future clinical trials.
Collapse
Affiliation(s)
- Eike Gallmeier
- Department of Medicine 2, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Dominik C. Bader
- Department of Medicine 2, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Lydia Kriegl
- Institute of Pathology, University of Munich, Munich, Germany
| | | | - Hendrik Seeliger
- Department of Surgery, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Burkhard Göke
- Department of Medicine 2, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Thomas Kirchner
- Institute of Pathology, University of Munich, Munich, Germany
| | - Christiane Bruns
- Department of Surgery, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Enrico N. De Toni
- Department of Medicine 2, University Hospital Grosshadern, University of Munich, Munich, Germany
| |
Collapse
|
178
|
张 涛, 袁 帅, 王 子, 张 茜, 赵 盼, 单 莉. [Bevacizumab combined with chemotherapy for advanced non-small cell lung cancer: a meta-analysis]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2013; 16:82-90. [PMID: 23425900 PMCID: PMC6000394 DOI: 10.3779/j.issn.1009-3419.2013.02.05] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 02/06/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND OBJECTIVE Bevacizumab is a recombinant, humanised, monoclonal antibody against the vascular endothelial growth factor receptor (VEGFR), the aim of this meta-analysis is to evaluate the clinical efficacy and safety of bevacizumab combined with chemotherapy for non-small cell lung cancer (NSCLC). METHODS The CBM, CNKI, VIP, WanFang, The Cochrane Library, PubMed, Ovid, EMBASE and SCI etc were retrieved by computer, the randomized controlled trials (randomized control trial, RCT) of bevacizumab combined with chemotherapy in the treatment of NSCLC were collected by us. The outcomes included overall survival (OS), progression-free survival (PFS), response rate (RR), toxicities and treatment related mortality. Relative risk (RR) and hazard ratios (HR) were used for the meta-analysis and were expressed with 95% confidence intervals (95%CI), and the software Stata 12.0 was used for meta-analyses. RESULTS Six trials and 2,338 advanced NSCLC patients were included. Meta analysis indicated that compared with chemotherapy alone, the bevacizumab (7.5 mg/kg or 15 mg/kg) combination with chemotherapy could increase overall survival rate (RR=1.68, P<0.01, 95%CI: 1.31-2.15; RR= 1.79, P<0.01, 95%CI: 1.53-2.08), and could decrease the progression of the disease risk (HR=0.75, P<0.01, 95%CI: 0.61-0.89; HR=0.69, P<0.01, 95%CI: 0.62-0.77) and the risk of disease death (HR=0.94, P<0.01, 95%CI: 0.77-1.10; HR=0.87, P<0.01, 95%CI: 0.78-0.97). The high dose of bevacizumab (15 mg/kg) combination with chemotherapy could increase the treatment related mortality (RR=1.88, P=0.010, 95%CI: 1.16-3.05) and the rates of other toxic reaction. CONCLUSIONS Whether first-line or second-line treatment, the addition of bevacizumab to chemotherapy in patients with advanced NSCLC prolongs RR, OS and PFS.
Collapse
Affiliation(s)
- 涛 张
- 830011 乌鲁木齐,新疆医科大学附属肿瘤医院内一科Department of Medical Oncology, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830011, China
| | - 帅飞 袁
- 830011 乌鲁木齐,新疆医科大学附属肿瘤医院内一科Department of Medical Oncology, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830011, China
| | - 子平 王
- 100021 北京,中国医学科学院北京协和医学院肿瘤医院内科Department of Medical Oncology, Cancer Hospital & Institute, Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - 茜 张
- 830011 乌鲁木齐,新疆医科大学附属肿瘤医院研究生管理科Department of Graduate, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830011, China
| | - 盼盼 赵
- 830011 乌鲁木齐,新疆医科大学附属肿瘤医院内一科Department of Medical Oncology, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830011, China
| | - 莉 单
- 830011 乌鲁木齐,新疆医科大学附属肿瘤医院内一科Department of Medical Oncology, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830011, China
| |
Collapse
|
179
|
Abstract
Inhibitors of tumour necrosis factor (TNF) are among the most successful protein-based drugs (biologics) and have proven to be clinically efficacious at reducing inflammation associated with several autoimmune diseases. As a result, attention is focusing on the therapeutic potential of additional members of the TNF superfamily of structurally related cytokines. Many of these TNF-related cytokines or their cognate receptors are now in preclinical or clinical development as possible targets for modulating inflammatory diseases and cancer as well as other indications. This Review focuses on the biologics that are currently in clinical trials for immune-related diseases and other syndromes, discusses the successes and failures to date as well as the expanding therapeutic potential of modulating the activity of this superfamily of molecules.
Collapse
Affiliation(s)
- Michael Croft
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA.
| | | | | |
Collapse
|
180
|
Monma H, Harashima N, Inao T, Okano S, Tajima Y, Harada M. The HSP70 and autophagy inhibitor pifithrin-μ enhances the antitumor effects of TRAIL on human pancreatic cancer. Mol Cancer Ther 2013; 12:341-51. [PMID: 23371857 DOI: 10.1158/1535-7163.mct-12-0954] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
TRAIL and agonistic death receptor-specific antibodies can induce apoptosis in cancer cells with little cytotoxicity to normal cells. To improve TRAIL-induced antitumor effects, we tested its effectiveness in combination with pifithrin (PFT)-μ, which has the potential to inhibit HSP70 function and autophagy, both of which participate in TRAIL resistance in cancer cells. Among the four human pancreatic cancer cell lines tested, MiaPaca-2, Panc-1, and BxPC-3 cells showed varying sensitivities to TRAIL. In MiaPaca-2 and Panc-1 cells, knockdown of HSP70 or beclin-1, the latter an autophagy-related molecule, by RNA interference augmented TRAIL-induced antitumor effects, decreasing cell viability, and increasing apoptosis. On the basis of these findings, we next determined whether the TRAIL-induced antitumor effects could be augmented by its combination with PFT-μ. The combination of TRAIL plus PFT-μ significantly decreased the viability and colony-forming ability of MiaPaca-2 and Panc-1 cells compared with cells treated with either agent alone. When applied alone, PFT-μ increased Annexin V(+) cells in both caspase-dependent and -independent manners. It also promoted TRAIL-induced apoptosis and arrested cancer cell growth. Furthermore, PFT-μ antagonized TRAIL-associated NF-κB activation in cancer cells. In a xenograft mouse model, combination therapy significantly inhibited MiaPaca-2 tumor growth compared with treatment with either agent alone. The results of this study suggest protective roles for HSP70 and autophagy in TRAIL resistance in pancreatic cancer cells and suggest that PFT-μ is a promising agent for use in therapies intended to enhance the antitumor effects of TRAIL.
Collapse
Affiliation(s)
- Hiroyuki Monma
- Department of Immunology, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan
| | | | | | | | | | | |
Collapse
|
181
|
Berger A, Quast SA, Plötz M, Kammermeier A, Eberle J. Sensitization of melanoma cells for TRAIL-induced apoptosis by BMS-345541 correlates with altered phosphorylation and activation of Bax. Cell Death Dis 2013; 4:e477. [PMID: 23348591 PMCID: PMC3563986 DOI: 10.1038/cddis.2012.198] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 11/27/2012] [Accepted: 11/28/2012] [Indexed: 12/12/2022]
Abstract
Resistance to TRAIL (TNF-related apoptosis-inducing ligand)- induced apoptosis limits its therapeutic use. Different strategies of TRAIL sensitization and a dependency on Bax have been reported, but common principles of TRAIL resistance and the way of Bax activation remained poorly understood. Applying a melanoma model of TRAIL-sensitive and -resistant cell lines, efficient sensitization for TRAIL-induced apoptosis is demonstrated by the kinase inhibitor BMS-345541 (N-(1,8-dimethylimidazo(1,2-a)quinoxalin-4-yl)-1,2-ethanediamine hydrochloride), which targets IκB (inhibitor of κB proteins) kinase β (IKKβ). This effect was completely abrogated by Bax knockout as well as by Bcl-2 overexpression, in accordance with a Bax dependency. Early loss of the mitochondrial membrane potential, release of cytochrome c and Smac (second mitochondria-derived activator of caspases) clearly indicated the activation of mitochondrial apoptosis pathways. Of note, BMS-345541 alone resulted in an early Bax activation, seen by conformational changes and by Bax translocation. The synergistic effects can be explained by Bid activation through TRAIL, which inhibits Bcl-2, and the activation of Bax through BMS-345541. The critical roles of XIAP (X-chromosome-linked inhibitor of apoptosis protein), Smac and Bid were clearly proven by overexpression and siRNA knockdown, respectively. The way of Bax activation by BMS-345541 was unraveled by establishing new assays for Bax activation. These showed reduction of the inactivating Bax phosphorylation at serine-184, while the activating Bax phosphorylation at threonine-167 was enhanced. Thus, modulation of Bax phosphorylation appeared as tightly related to TRAIL sensitivity/resistance in melanoma cells, and therapeutic strategies may be considered.
Collapse
Affiliation(s)
- A Berger
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | - S-A Quast
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | - M Plötz
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | - A Kammermeier
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | - J Eberle
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| |
Collapse
|
182
|
Long JS, Ryan KM. New frontiers in promoting tumour cell death: targeting apoptosis, necroptosis and autophagy. Oncogene 2012; 31:5045-60. [PMID: 22310284 DOI: 10.1038/onc.2012.7] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 12/24/2011] [Accepted: 12/24/2011] [Indexed: 12/12/2022]
Abstract
Cancer is a multifaceted disease comprising a combination of genetic, metabolic and signalling aberrations, which severely disrupt the normal homeostasis of cell growth and death. Many oncogenic events while promoting tumour development also increase the sensitivity of cells to cell death stimuli including chemotherapeutic drugs. As a result, tumour cells often acquire the ability to evade death by inactivating cell death pathways that normally function to eliminate damaged and harmful cells. The impairment of cell death function is also often the reason for the development of chemotherapeutic resistance encountered during treatment. It is therefore necessary to achieve a comprehensive understanding of existing cell death pathways and the relevant regulatory components involved, with the intention of identifying new strategies to kill cancer cells. This review provides an insightful overview of the common forms of cell death signalling pathways, the interactions between these pathways and the ways in which these pathways are deregulated in cancer. We also discuss the emerging therapies targeted at activating or restoring cell death pathways to induce tumour cell death, which are currently being tested in clinical trials.
Collapse
Affiliation(s)
- J S Long
- Tumour Cell Death Laboratory, Beatson Institute for Cancer Research, Glasgow, UK
| | | |
Collapse
|
183
|
Antibody–cytokine fusion proteins. Arch Biochem Biophys 2012; 526:194-205. [DOI: 10.1016/j.abb.2012.03.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 02/28/2012] [Accepted: 03/01/2012] [Indexed: 01/01/2023]
|
184
|
Bellail AC, Hao C. The roadmap of TRAIL apoptotic pathway-targeted cancer therapies: What is next? Expert Rev Anticancer Ther 2012; 12:547-9. [PMID: 22594889 DOI: 10.1586/era.12.33] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
185
|
Quast SA, Berger A, Buttstädt N, Friebel K, Schönherr R, Eberle J. General Sensitization of melanoma cells for TRAIL-induced apoptosis by the potassium channel inhibitor TRAM-34 depends on release of SMAC. PLoS One 2012; 7:e39290. [PMID: 22723988 PMCID: PMC3377761 DOI: 10.1371/journal.pone.0039290] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 05/20/2012] [Indexed: 11/18/2022] Open
Abstract
The death ligand TRAIL represents a promising therapeutic strategy for metastatic melanoma, however prevalent and inducible resistance limit its applicability. A new approach is presented here for sensitization to TRAIL. It is based on inhibition of the membrane potassium channel KCa3.1 (IK1), which serves fundamental cellular functions related to membrane potential. The selective inhibitor TRAM-34 did not induce apoptosis by itself but synergistically enhanced TRAIL sensitivity and overrode TRAIL resistance in a large panel of melanoma cell lines. Expression of IK1 was also found in mitochondria, and its inhibition resulted in mitochondrial membrane hyperpolarization and an early activation of Bax. The combination of TRAM-34 and TRAIL resulted in massive release of mitochondrial factors, cytochrome c, AIF and SMAC/DIABLO. Bax knockdown and Bcl-2 overexpression abolished apoptosis. Overexpression of XIAP diminished apoptosis by two-fold, and SMAC knockdown almost completely abolished apoptosis. These data uncover the existence of a rheostat in melanoma cells, consisting of inhibitor of apoptosis proteins and SMAC, which regulates TRAIL sensitivity. Thus, a new strategy is described based on mitochondrial membrane channels, which correspond to Bax activation. As both TRAIL and IK1 inhibitors had shown only minor side effects in clinical trials, a clinical application of this combination is conceivable.
Collapse
Affiliation(s)
- Sandra-Annika Quast
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
- Institute for Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
| | - Anja Berger
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | - Nicole Buttstädt
- Department of Biophysics, Center of Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | - Kristin Friebel
- Department of Biophysics, Center of Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | - Roland Schönherr
- Department of Biophysics, Center of Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | - Jürgen Eberle
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
- * E-mail:
| |
Collapse
|
186
|
Bellail AC, Hao C. TRAIL apoptotic pathway-targeted therapies for NSCLC. Transl Lung Cancer Res 2012; 1:155-7. [PMID: 25806174 DOI: 10.3978/j.issn.2218-6751.2012.02.02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 03/04/2012] [Indexed: 11/14/2022]
Affiliation(s)
- Anita C Bellail
- Department of Pathology & Laboratory Medicine, Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road NE, Atlanta, GA 30322, USA
| | - Chunhai Hao
- Department of Pathology & Laboratory Medicine, Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road NE, Atlanta, GA 30322, USA
| |
Collapse
|
187
|
Ocker M, Höpfner M. Apoptosis-modulating drugs for improved cancer therapy. ACTA ACUST UNITED AC 2012; 48:111-20. [PMID: 22538523 DOI: 10.1159/000336875] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 01/09/2012] [Indexed: 12/16/2022]
Abstract
Resistance to cell death induction has been recognized as a hallmark of cancer. Increasing understanding of the underlying molecular events regulating different cell death mechanisms like apoptosis, endoplasmic reticulum stress, autophagy, necroptosis and others has opened new possibilities for targeted interference with these pathways. While conventional chemotherapeutic agents usually inhibit cell cycle progression, DNA replication or mitosis execution, novel agents like small molecule kinase inhibitors also target survival-related kinases and signaling pathways and contribute to overcome resistance to chemotherapy and apoptosis. Additionally, antibodies targeting cellular death receptors have been described to specifically target tumor cells only. This review briefly highlights the pathways involved in (apoptotic) cell death and summarizes the current state of development of specific modulators of cell death and how they can help to improve the tolerability of chemotherapy regimens and increase survival rates in patients with advanced cancer diseases.
Collapse
Affiliation(s)
- M Ocker
- Institute for Surgical Research, Philipps University Marburg, Marburg, Germany.
| | | |
Collapse
|
188
|
Su Y, Yang WB, Li S, Ye ZJ, Shi HZ, Zhou Q. Effect of angiogenesis inhibitor bevacizumab on survival in patients with cancer: a meta-analysis of the published literature. PLoS One 2012; 7:e35629. [PMID: 22539986 PMCID: PMC3335091 DOI: 10.1371/journal.pone.0035629] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 03/19/2012] [Indexed: 02/07/2023] Open
Abstract
Bevacizumab is a recombinant humanized monoclonal antibody against vascular endothelial growth factor which has been used in conjunction with other anti-cancer agents in the treatment of patients with many cancers. It remains controversial whether bevacizumab can prolong survival in cancer patients. This meta-analysis was therefore performed to evaluate effect of bevacizumab on survival in cancer patients. PubMed, EMBASE, and Web of Science databases were searched for English-language studies of randomized controlled trials comparing bevacizumab with control therapy published through February 8, 2012. Progression-free survival, overall survival, and one-year survival rate were analyzed using random- or fixed-effects model. Thirty one assessable randomized controlled trials were identified. A significant improvement in progression-free survival in cancer patients was attributable to bevacizumab compared with control therapy (hazard ratio, 0.72; 95% confidence interval, 0.68 to 0.76; p<0.001). Overall survival was also significantly longer in patients were treated with bevacizumab (hazard ratio, 0.87; 95% confidence interval, 0.83 to 0.91; p<0.001). The significant benefit in one-year survival rate was further seen in cancer patients receiving bevacizumab (odds ratio, 1.30; 95% confidence interval, 1.20 to 1.41; p<0.001). Current evidences showed that bevacizumab prolong progression-free survival and overall survival, and increase one-year survival rate in cancer patients as compared with control therapy.
Collapse
Affiliation(s)
| | | | | | | | - Huan-Zhong Shi
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiong Zhou
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
189
|
Dean E, Greystoke A, Ranson M, Dive C. Biomarkers of cell death applicable to early clinical trials. Exp Cell Res 2012; 318:1252-9. [PMID: 22483936 DOI: 10.1016/j.yexcr.2012.03.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 03/19/2012] [Accepted: 03/19/2012] [Indexed: 01/12/2023]
Abstract
The development of biomarkers of cell death to reflect tumor biology and drug-induced response has garnered interest with the development of several classes of drugs aimed at decreasing the cellular threshold for apoptosis and exploiting pre-existing oncogenic stresses. These novel anticancer drugs, directly targeted to the apoptosis regulatory machinery and aimed at abrogating survival signaling pathways, are entering early clinical trials provoking the question of how to monitor their impact on cancer patients. The parallel development of drugs with predictive biomarkers and their incorporation into early clinical trials are anticipated to support the pharmacological audit trail, to speed the development and reduce the attrition rate of novel drugs whose objective is to provoke tumor cell death. Tumor biopsies are an ideal matrix to measure apoptosis, but surrogate less invasive biomarkers such as blood samples and functional imaging are less challenging to acquire clinically. Archetypal and exploratory examples illustrating the importance of biomarkers to drug development are given. This review explores the substantive challenges associated with the validation, deployment, interpretation and utility of biomarkers of cell death and reviews recent advances in their incorporation in preclinical and early clinical trial contexts.
Collapse
Affiliation(s)
- Emma Dean
- Paterson Institute for Cancer Research, The University of Manchester, Withington, Manchester, UK.
| | | | | | | |
Collapse
|
190
|
Cheng AL, Shen YC, Zhu AX. Targeting fibroblast growth factor receptor signaling in hepatocellular carcinoma. Oncology 2012; 81:372-80. [PMID: 22269894 DOI: 10.1159/000335472] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 11/16/2011] [Indexed: 01/22/2023]
Abstract
Hepatocellular carcinoma (HCC) is the primary type of liver cancer, and both the age-adjusted incidence and mortality of HCC have steadily increased in recent years. Advanced HCC is associated with a very poor survival rate. Despite accumulating data regarding the risk factors for HCC, the mechanisms that contribute to HCC tumorigenesis remain poorly understood. Signaling through the fibroblast growth factor (FGF) family is involved in fibrosis and its progression to cirrhosis of the liver, which is a risk factor for the development of HCC. Furthermore, several alterations in FGF/FGF receptor (FGFR) signaling correlate with the outcomes of HCC patients, suggesting that signaling through this family of proteins contributes to the development or progression of HCC tumors. Currently, there are no established systemic treatments for patients with advanced HCC in whom sorafenib treatment has failed or who were unable to tolerate it. Recently, several multikinase inhibitors that target FGFRs have demonstrated some early evidence of antitumor activity in phase I/II trials. Therefore, this review discusses the molecular implications of FGFR-mediated signaling in HCC and summarizes the clinical evidence for novel FGFR-targeted therapies for HCC currently being studied in clinical trials.
Collapse
Affiliation(s)
- Ann-Lii Cheng
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.
| | | | | |
Collapse
|