101
|
Servida F, Lecis D, Scavullo C, Drago C, Seneci P, Carlo-Stella C, Manzoni L, Polli E, Lambertenghi Deliliers G, Delia D, Onida F. Novel second mitochondria-derived activator of caspases (Smac) mimetic compounds sensitize human leukemic cell lines to conventional chemotherapeutic drug-induced and death receptor-mediated apoptosis. Invest New Drugs 2010; 29:1264-75. [PMID: 20614162 DOI: 10.1007/s10637-010-9475-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Accepted: 06/07/2010] [Indexed: 01/26/2023]
Abstract
The Inhibitor of Apoptosis Proteins (IAPs) are important regulators of programmed cell death. XIAP is the most potent among them and is over-expressed in several hematological malignancies. Its activity is endogenously antagonized by SMAC/DIABLO, and also by small molecules mimicking Smac that can induce apoptosis in tumor cells. Here we describe the activity of 56 newly synthesized Smac-mimetics in human leukemic cell lines and normal CD34(+) progenitor cells. Our compounds bind to XIAP with high affinity, reduce the levels of cIAP1 and are cytotoxic at nanomolar or low micromolar concentrations. Furthermore, the Smac-mimetics synergize with Cytarabine, Etoposide and especially with TRAIL in combination treatments. Apoptosis activation was clearly detectable by the occurrence of sub G(1) apoptotic peak and the accumulation of cleaved PARP, caspase 8 and caspase 3. Interestingly, the down-regulation of XIAP sensitized Jurkat cells to drugs too, confirming the role of this protein in drug-resistance. In conclusion, while being very active in leukemic cells, our Smac-mimetics have modest effects on normal hematopoietic progenitors, suggesting their promising therapeutic potential as a new class of anticancer drugs in onco-hematology, particularly when combined with TRAIL, to overcome the resistance of cancer cells.
Collapse
Affiliation(s)
- Federica Servida
- Fondazione Matarelli, Dipartimento di Farmacologia Chemioterapia e Tossicologia Medica, Università degli Studi di Milano, Via Vanvitelli 32, 20129, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Lunov O, Syrovets T, Büchele B, Jiang X, Röcker C, Tron K, Nienhaus GU, Walther P, Mailänder V, Landfester K, Simmet T. The effect of carboxydextran-coated superparamagnetic iron oxide nanoparticles on c-Jun N-terminal kinase-mediated apoptosis in human macrophages. Biomaterials 2010; 31:5063-71. [DOI: 10.1016/j.biomaterials.2010.03.023] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 03/04/2010] [Indexed: 01/18/2023]
|
103
|
Targeting X-linked inhibitor of apoptosis protein to increase the efficacy of endoplasmic reticulum stress-induced apoptosis for melanoma therapy. J Invest Dermatol 2010; 130:2250-8. [PMID: 20520630 DOI: 10.1038/jid.2010.146] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Melanoma remains notoriously resistant to current chemotherapeutics, leaving an acute need for novel therapeutic approaches. The aim of this study was to determine the prognostic and therapeutic significance of X-linked inhibitor of apoptosis protein (XIAP) in melanoma through correlation of XIAP expression with disease stage, RAS/RAF mutational status, clinical outcome, and susceptibility to endoplasmic reticulum (ER) stress-induced cell death. XIAP expression and N-RAS/B-RAF mutational status were retrospectively determined in a cohort of 55 primary cutaneous melanocytic lesions selected and grouped according to the American Joint Committee on Cancer staging system. Short hairpin RNA interference of XIAP was used to analyze the effect of XIAP expression on ER stress-induced apoptosis in response to fenretinide or bortezomib in vitro. The results showed that XIAP positivity increased with progressive disease stage, although there was no significant correlation between XIAP positivity and combined N-RAS/B-RAF mutational status or clinical outcome. However, XIAP knockdown significantly increased ER stress-induced apoptosis of melanoma cells in a caspase-dependant manner. The correlation of XIAP expression with disease stage, as well as data showing that XIAP knockdown significantly increases fenretinide and bortezomib-induced apoptosis of metastatic melanoma cells, suggests that XIAP may prove to be an effective therapeutic target for melanoma therapy.
Collapse
|
104
|
Lecis D, Drago C, Manzoni L, Seneci P, Scolastico C, Mastrangelo E, Bolognesi M, Anichini A, Kashkar H, Walczak H, Delia D. Novel SMAC-mimetics synergistically stimulate melanoma cell death in combination with TRAIL and Bortezomib. Br J Cancer 2010; 102:1707-16. [PMID: 20461078 PMCID: PMC2883696 DOI: 10.1038/sj.bjc.6605687] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND XIAP (X-linked inhibitor of apoptosis protein) is an anti-apoptotic protein exerting its activity by binding and suppressing caspases. As XIAP is overexpressed in several tumours, in which it apparently contributes to chemoresistance, and because its activity in vivo is antagonised by second mitochondria-derived activator of caspase (SMAC)/direct inhibitor of apoptosis-binding protein with low pI, small molecules mimicking SMAC (so called SMAC-mimetics) can potentially overcome tumour resistance by promoting apoptosis. METHODS Three homodimeric compounds were synthesised tethering a monomeric SMAC-mimetic with different linkers and their affinity binding for the baculoviral inhibitor repeats domains of XIAP measured by fluorescent polarisation assay. The apoptotic activity of these molecules, alone or in combination with tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and/or Bortezomib, was tested in melanoma cell lines by MTT viability assays and western blot analysis of activated caspases. RESULTS We show that in melanoma cell lines, which are typically resistant to chemotherapeutic agents, XIAP knock-down sensitises cells to TRAIL treatment in vitro, also favouring the accumulation of cleaved caspase-8. We also describe a new series of 4-substituted azabicyclo[5.3.0]alkane monomeric and dimeric SMAC-mimetics that target various members of the IAP family and powerfully synergise at submicromolar concentrations with TRAIL in inducing cell death. Finally, we show that the simultaneous administration of newly developed SMAC-mimetics with Bortezomib potently triggers apoptosis in a melanoma cell line resistant to the combined effect of SMAC-mimetics and TRAIL. CONCLUSION Hence, the newly developed SMAC-mimetics effectively synergise with TRAIL and Bortezomib in inducing cell death. These findings warrant further preclinical studies in vivo to verify the anticancer effectiveness of the combination of these agents.
Collapse
Affiliation(s)
- D Lecis
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Via G Venezian 1, 20133 Milano, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Van Herreweghe F, Festjens N, Declercq W, Vandenabeele P. Tumor necrosis factor-mediated cell death: to break or to burst, that's the question. Cell Mol Life Sci 2010; 67:1567-79. [PMID: 20198502 PMCID: PMC11115929 DOI: 10.1007/s00018-010-0283-0] [Citation(s) in RCA: 159] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 01/20/2010] [Indexed: 02/06/2023]
Abstract
In this review, we discuss the signal-transduction pathways of three major cellular responses induced by tumor necrosis factor (TNF): cell survival through NF-kappaB activation, apoptosis, and necrosis. Recruitment and activation of caspases plays a crucial role in the initiation and execution of TNF-induced apoptosis. However, experimental inhibition of caspases reveals an alternative cell death pathway, namely necrosis, also called necroptosis, suggesting that caspases actively suppress the latter outcome. TNF-induced necrotic cell death crucially depends on the kinase activity of receptor interacting protein serine-threonine kinase 1 (RIP1) and RIP3. It was recently demonstrated that ubiquitination of RIP1 determines whether it will function as a pro-survival or pro-cell death molecule. Deeper insight into the mechanisms that control the molecular switches between cell survival and cell death will help us to understand why TNF can exert so many different biological functions in the etiology and pathogenesis of human diseases.
Collapse
Affiliation(s)
- Franky Van Herreweghe
- Unit For Molecular Signalling and Cell Death, Department for Molecular Biomedical Research, VIB, Technologiepark 927, 9052 Ghent (Zwijnaarde), Belgium
- Unit for Molecular Signalling and Cell Death, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Ghent, Belgium
| | - Nele Festjens
- Unit for Molecular Glycobiology, Department for Molecular Biomedical Research, VIB, Ghent University, Technologiepark 927, 9052 Ghent, Belgium
- Laboratory for Protein Biochemistry and Biomolecular Engineering, Department of Biochemistry and Microbiology, Ghent University, Ledeganckstraat 35, 9052 Ghent, Belgium
| | - Wim Declercq
- Unit For Molecular Signalling and Cell Death, Department for Molecular Biomedical Research, VIB, Technologiepark 927, 9052 Ghent (Zwijnaarde), Belgium
- Unit for Molecular Signalling and Cell Death, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Ghent, Belgium
| | - Peter Vandenabeele
- Unit For Molecular Signalling and Cell Death, Department for Molecular Biomedical Research, VIB, Technologiepark 927, 9052 Ghent (Zwijnaarde), Belgium
- Unit for Molecular Signalling and Cell Death, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Ghent, Belgium
| |
Collapse
|
106
|
Kauh J, Fan S, Xia M, Yue P, Yang L, Khuri FR, Sun SY. c-FLIP degradation mediates sensitization of pancreatic cancer cells to TRAIL-induced apoptosis by the histone deacetylase inhibitor LBH589. PLoS One 2010; 5:e10376. [PMID: 20442774 PMCID: PMC2860986 DOI: 10.1371/journal.pone.0010376] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Accepted: 04/07/2010] [Indexed: 11/19/2022] Open
Abstract
Great efforts have been made to develop novel and efficacious therapeutics against pancreatic cancer to improve the treatment outcomes. Tumor-necrosis factor-related apoptosis-inducing ligand (TRAIL) is such a therapeutic cytokine with selective killing effect toward malignant cells. However, some human pancreatic cancers are intrinsically resistant to TRAIL-mediated apoptosis or therapy. In this study, we have shown that the histone deacetylase inhibitor LBH589 can synergize with TRAIL to augment apoptosis even in TRAIL-resistant cells. LBH589 decreased c-FLIP levels in every tested cell line and survivin levels in some of the tested cell lines. Enforced expression of ectopic c-FLIP, but not survivin, abolished the cooperative induction of apoptosis by the combination of LBH589 and TRAIL, indicating that c-FLIP downregulation plays a critical role in LBH589 sensitization of pancreatic cancer cells to TRAIL. Moreover, LBH589 decreased c-FLIP stability and the presence of the proteasome inhibitor MG132 prevented c-FLIP from reduction by LBH589. Correspondingly, we detected increased levels of ubiqutinated c-FLIP in LBH589-treated cells. These data thus indicate that LBH589 promotes ubiqutin/proteasome-mediated degradation of c-FLIP, leading to downregulation of c-FLIP. Collectively, LBH589 induces c-FLIP degradation and accordingly sensitizes pancreatic cancer cells to TRAIL-induced apoptosis, highlighting a novel therapeutic regimen against pancreatic cancer.
Collapse
Affiliation(s)
- John Kauh
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, United States of America
| | - Songqing Fan
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, United States of America
| | - Mingjing Xia
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, United States of America
| | - Ping Yue
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, United States of America
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, United States of America
| | - Fadlo R. Khuri
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, United States of America
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
107
|
Schüler S, Fritsche P, Diersch S, Arlt A, Schmid RM, Saur D, Schneider G. HDAC2 attenuates TRAIL-induced apoptosis of pancreatic cancer cells. Mol Cancer 2010; 9:80. [PMID: 20398369 PMCID: PMC2867820 DOI: 10.1186/1476-4598-9-80] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Accepted: 04/16/2010] [Indexed: 12/31/2022] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant tumors with a dismal prognosis and no effective conservative therapeutic strategies. Although it is demonstrated that histone deacetylases (HDACs), especially the class I HDACs HDAC1, 2 and 3 are highly expressed in this disease, little is known about HDAC isoenzyme specific functions. Results Depletion of HDAC2, but not HDAC1, in the pancreatic cancer cell lines MiaPaCa2 and Panc1 resulted in a marked sensitization towards the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Correspondingly, the more class I selective HDAC inhibitor (HDACI) valproic acid (VPA) synergized with TRAIL to induce apoptosis of MiaPaCa2 and Panc1 cells. At the molecular level, an increased expression of the TRAIL receptor 1 (DR5), accelerated processing of caspase 8, pronounced cleavage of the BH3-only protein Bid, and increased effector caspase activation was observed in HDAC2-depleted and TRAIL-treated MiaPaCa2 cells. Conclusions Our data characterize a novel HDAC2 function in PDAC cells and point to a strategy to overcome TRAIL resistance of PDAC cells, a prerequisite to succeed with a TRAIL targeted therapy in clinical settings.
Collapse
Affiliation(s)
- Susanne Schüler
- Technische Universität München, Klinikum rechts der Isar, II, Medizinische Klinik, Ismaninger Str, 22, 81675 Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
108
|
Dineen SP, Roland CL, Greer R, Carbon JG, Toombs JE, Gupta P, Bardeesy N, Sun H, Williams N, Minna JD, Brekken RA. Smac mimetic increases chemotherapy response and improves survival in mice with pancreatic cancer. Cancer Res 2010; 70:2852-61. [PMID: 20332237 DOI: 10.1158/0008-5472.can-09-3892] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Failure of chemotherapy in the treatment of pancreatic cancer is often due to resistance to therapy-induced apoptosis. A major mechanism for such resistance is the expression and activity of inhibitors of apoptosis proteins (IAP). Smac (second mitochondria-derived activator of caspase) is a mitochondrial protein that inhibits IAPs. We show that JP1201, a Smac mimetic, is a potent enhancer of chemotherapy in robust mouse models of pancreatic cancer. Combination of JP1201 with gemcitabine reduced primary and metastatic tumor burden in orthotopic xenograft and syngenic tumor models, induced regression of established tumors, and prolonged survival in xenograft and transgenic models of pancreatic cancer. The effect of JP1201 was phenocopied by XIAP small interfering RNA in vitro and correlated with elevated levels of tumor necrosis factor alpha protein in vivo. The continued development of JP1201 and other strategies designed to enhance therapy-induced apoptosis in pancreatic cancer is warranted.
Collapse
Affiliation(s)
- Sean P Dineen
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical School, Dallas, TX, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Ding W, Cai T, Zhu H, Wu R, Tu C, Yang L, Lu W, He Q, Yang B. Synergistic antitumor effect of TRAIL in combination with sunitinib in vitro and in vivo. Cancer Lett 2010; 293:158-66. [PMID: 20137855 DOI: 10.1016/j.canlet.2010.01.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2009] [Revised: 12/25/2009] [Accepted: 01/11/2010] [Indexed: 11/28/2022]
Abstract
The present data showed that sunitinib potentiated the in vitro and in vivo anticancer capabilities of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), also known as Apo2 ligand. Interactions between sunitinib and TRAIL were examined in colon cancer SW620 cells and lung cancer 95-D cells. The average combination index (CI) values of the anti-proliferation abilities on each cancer cell line were less than 1.0, demonstrating the synergism of the combination of sunitinib and TRAIL. Western blot experiments indicated that TRAIL and sunitinib synergistically enhanced apoptosis by simultaneously activating the extrinsic and intrinsic pathways. The decrease in the expression levels of anti-apoptotic proteins cFLIP, XIAP and Mcl-1 were probably involved in this apoptosis enhancement. Furthermore, treatment of colon cancer SW620-bearing nude mice with sunitinib plus TRAIL resulted in more significant tumor growth inhibition (52.8%), comparing with the moderate inhibition in TRAIL-treated (35.3%) or sunitinib-treated groups (26.7%) (p<0.05). These results indicate that the combination of TRAIL with sunitinib seems highly encouraging and warrants further investigation in a clinical setting.
Collapse
Affiliation(s)
- Wanjing Ding
- School of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Xiang G, Wen X, Wang H, Chen K, Liu H. Expression of X-linked inhibitor of apoptosis protein in human colorectal cancer and its correlation with prognosis. J Surg Oncol 2010; 100:708-12. [PMID: 19777490 DOI: 10.1002/jso.21408] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND X-linked inhibitor of apoptosis protein (XIAP) is a member of the inhibitor of apoptosis family of proteins and deregulation of XIAP can result in tumorigenicity. The objective of this study was to evaluate the prognostic significance of XIAP expression in colorectal cancer (CRC). METHODS RT-PCR was performed to detect the expression of XIAP mRNA in CRC cells and tissues. The expression of XIAP protein in tissues was measured by immunohistochemistry. The correlation of XIAP expression with clinicopathologic factors and prognosis of CRC patients was evaluated. RESULTS CRC cells showed significantly higher levels of XIAP mRNA expression than normal human intestinal epithelial cell. The expression level of XIAP mRNA in CRC samples was significantly higher than that in corresponding non-tumor samples. XIAP staining was positive in the cytoplasm of CRC cells. Higher XIAP protein expression was significantly correlated with tumor differentiation (P = 0.016), venous invasion (P = 0.039), and Duke's staging (P = 0.002). Moreover, XIAP-high group showed lower disease-free (P = 0.0136) and overall survival (P = 0.0084) rates than XIAP-low group. Multivariate analysis indicated that the status of XIAP expression was an independent prognostic factor for CRC (P = 0.0206; HR: 2.730; 95% CI: 1.226-5.445). CONCLUSION The status of XIAP expression might become an independent prognostic marker for CRC.
Collapse
Affiliation(s)
- Guoan Xiang
- Department of General Surgery, The Second People's Hospital of Guangdong Province, Guangzhou, China
| | | | | | | | | |
Collapse
|
111
|
Holoch PA, Griffith TS. TNF-related apoptosis-inducing ligand (TRAIL): a new path to anti-cancer therapies. Eur J Pharmacol 2009; 625:63-72. [PMID: 19836385 PMCID: PMC2783837 DOI: 10.1016/j.ejphar.2009.06.066] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 06/08/2009] [Accepted: 06/22/2009] [Indexed: 12/31/2022]
Abstract
Since its discovery in 1995, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a member of the tumor necrosis factor super family, has been under intense focus because of its remarkable ability to induce apoptosis in malignant human cells while leaving normal cells unscathed. Consequently, activation of the apoptotic signaling pathway from the death-inducing TRAIL receptors provides an attractive, biologically-targeted approach to cancer therapy. A great deal of research has focused on deciphering the TRAIL receptor signaling cascade and intracellular regulation of this pathway, as many human tumor cells possess mechanisms of resistance to TRAIL-induced apoptosis. This review focuses on the current state of knowledge regarding TRAIL signaling and resistance, the preclinical development of therapies targeted at TRAIL receptors and modulators of the pathway, and the results of clinical trials for cancer treatment that have emerged from this base of knowledge. TRAIL-based approaches to cancer therapy vary from systemic administration of recombinant, soluble TRAIL protein with or without the combination of traditional chemotherapy, radiation or novel anti-cancer agents to agonistic monoclonal antibodies directed against functional TRAIL receptors to TRAIL gene transfer therapy. A better understanding of TRAIL resistance mechanisms may allow for the development of more effective therapies that exploit this cell-mediated pathway to apoptosis.
Collapse
Affiliation(s)
- Peter A Holoch
- Department of Urology, University of Iowa, 375 Newton Road, Iowa City, IA 52242, USA
| | | |
Collapse
|
112
|
Mellier G, Huang S, Shenoy K, Pervaiz S. TRAILing death in cancer. Mol Aspects Med 2009; 31:93-112. [PMID: 19995571 DOI: 10.1016/j.mam.2009.12.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Accepted: 12/02/2009] [Indexed: 12/13/2022]
Abstract
The observation that certain types of cancer express death receptors on their cell surface has triggered heightened interest in exploring the potential of receptor ligation as a novel anti-cancer modality, and since the expression is somewhat restricted to cancer cells the therapeutic implications are very promising. One such death receptor ligand belonging to the tumor necrosis receptor (TNF) superfamily, TNF-related apoptosis-inducing ligand (TRAIL), has been in the limelight as a tumor selective molecule that transmits death signal via ligation to its receptors (TRAIL-R1 and TRAIL-R2 or death receptors 4 and 5; DR4 and DR5). Interestingly, TRAIL-induced apoptosis exhibits hallmarks of extrinsic as well as intrinsic death pathways, and, therefore, is subject to regulation both at the cell surface receptor level as well as more downstream at the post-mitochondrial level. Despite the remarkable selectivity of DR expression on cancer cell surface, development of resistance to TRAIL-induced apoptosis remains a major challenge. Therefore, unraveling the cellular and molecular mechanisms of TRAIL resistance as well as identifying strategies to overcome this problem for an effective therapeutic response remains the cornerstone of many research endeavors. This review aims at presenting an overview of the biology, function and translational relevance of TRAIL with a specific view to discussing the various regulatory mechanisms and the current trends in reverting TRAIL resistance of cancer cells with the obvious implication of an improved clinical outcome.
Collapse
Affiliation(s)
- Gregory Mellier
- Department of Physiology, Yong Loo Lin School of Medicine, Singapore
| | | | | | | |
Collapse
|
113
|
Loeder S, Zenz T, Schnaiter A, Mertens D, Winkler D, Döhner H, Debatin KM, Stilgenbauer S, Fulda S. A novel paradigm to trigger apoptosis in chronic lymphocytic leukemia. Cancer Res 2009; 69:8977-86. [PMID: 19920200 DOI: 10.1158/0008-5472.can-09-2604] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Evasion of apoptosis is a hallmark of chronic lymphocytic leukemia (CLL), calling for new strategies to bypass resistance. Here, we provide first evidence that small-molecule X-linked inhibitor of apoptosis (XIAP) inhibitors in combination with the death receptor ligand tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) present a novel approach to trigger apoptosis in CLL, including subgroups with resistant disease or unfavorable prognosis. XIAP, cellular IAP (cIAP) 1, and cIAP2 are expressed at high levels in primary CLL samples. Proof-of-concept studies in CLL cell lines show that subtoxic concentrations of XIAP inhibitors significantly enhance TRAIL-induced apoptosis and also sensitize for CD95-mediated apoptosis. Importantly also in primary CLL samples, XIAP inhibitor acts in concert with TRAIL to trigger apoptosis in 18 of 27 (67%) cases. This XIAP inhibitor-induced and TRAIL-induced apoptosis involves caspase-3 activation and is blocked by the caspase inhibitor zVAD.fmk. The cooperative interaction of XIAP inhibitor and TRAIL is even evident in distinct subgroups of patients with poor prognostic features (i.e., with 17p deletion, TP53 mutation, chemotherapy-refractory disease, or unmutated V(H) genes). Interestingly, cases with unmutated V(H) genes were significantly more sensitive to XIAP inhibitor-induced and TRAIL-induced apoptosis compared with V(H) gene-mutated samples, pointing to a role of B-cell receptor signaling in apoptosis regulation. By showing that XIAP inhibitors in combination with TRAIL present a new strategy to trigger apoptosis even in resistant forms and poor prognostic subgroups of CLL, our findings have important implications for the development of apoptosis-based therapies in CLL.
Collapse
|
114
|
Varfolomeev E, Alicke B, Elliott JM, Zobel K, West K, Wong H, Scheer JM, Ashkenazi A, Gould SE, Fairbrother WJ, Vucic D. X chromosome-linked inhibitor of apoptosis regulates cell death induction by proapoptotic receptor agonists. J Biol Chem 2009; 284:34553-60. [PMID: 19854829 DOI: 10.1074/jbc.m109.040139] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Proapoptotic receptor agonists cause cellular demise through the activation of the extrinsic and intrinsic apoptotic pathways. Inhibitor of apoptosis (IAP) proteins block apoptosis induced by diverse stimuli. Here, we demonstrate that IAP antagonists in combination with Fas ligand (FasL) or the death receptor 5 (DR5) agonist antibody synergistically stimulate death in cancer cells and inhibit tumor growth. Single-agent activity of IAP antagonists relies on tumor necrosis factor-alpha signaling. By contrast, blockade of tumor necrosis factor-alpha does not affect the synergistic activity of IAP antagonists with FasL or DR5 agonist antibody. In most cancer cells, proapoptotic receptor agonist-induced cell death depends on amplifying the apoptotic signal via caspase-8-mediated activation of Bid and subsequent activation of the caspase-9-dependent mitochondrial apoptotic pathway. In the investigated cancer cell lines, induction of apoptosis by FasL or DR5 agonist antibody can be inhibited by knockdown of Bid. However, knockdown of X chromosome-linked IAP (XIAP) or antagonism of XIAP allows FasL or DR5 agonist antibody to induce activation of effector caspases efficiently without the need for mitochondrial amplification of the apoptotic signal and thus rescues the effect of Bid knockdown in these cells.
Collapse
Affiliation(s)
- Eugene Varfolomeev
- Department of Protein Engineering, Genentech, Inc., South San Francisco, California 94080, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Ndubaku C, Varfolomeev E, Wang L, Zobel K, Lau K, Elliott LO, Maurer B, Fedorova AV, Dynek JN, Koehler M, Hymowitz SG, Tsui V, Deshayes K, Fairbrother WJ, Flygare JA, Vucic D. Antagonism of c-IAP and XIAP proteins is required for efficient induction of cell death by small-molecule IAP antagonists. ACS Chem Biol 2009; 4:557-66. [PMID: 19492850 DOI: 10.1021/cb900083m] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The inhibitor of apoptosis (IAP) proteins are critical regulators of cancer cell survival, which makes them attractive targets for therapeutic intervention in cancers. Herein, we describe the structure-based design of IAP antagonists with high affinities and selectivity (>2000-fold) for c-IAP1 over XIAP and their functional characterization as activators of apoptosis in tumor cells. Although capable of inducing cell death and preventing clonogenic survival, c-IAP-selective antagonists are significantly less potent in promoting apoptosis when compared to pan-selective compounds. However, both pan-IAP- and c-IAP-selective antagonists stimulate c-IAP1 and c-IAP2 degradation and activation of NF-kappaB pathways with comparable potencies. Therefore, although compounds that specifically target c-IAP1 and c-IAP2 are capable of inducing apoptosis, antagonism of the c-IAP proteins and XIAP is required for efficient induction of cancer cell death by IAP antagonists.
Collapse
Affiliation(s)
- Chudi Ndubaku
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Eugene Varfolomeev
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Lan Wang
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Kerry Zobel
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Kevin Lau
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Linda O. Elliott
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Brigitte Maurer
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Anna V. Fedorova
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Jasmin N. Dynek
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Michael Koehler
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Sarah G. Hymowitz
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Vickie Tsui
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Kurt Deshayes
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Wayne J. Fairbrother
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - John A. Flygare
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Domagoj Vucic
- Departments of Medicinal Chemistry and Protein Engineering, Genentech, Inc., South San Francisco, California 94080
| |
Collapse
|
116
|
Histone deacetylase inhibitors cooperate with IFN-gamma to restore caspase-8 expression and overcome TRAIL resistance in cancers with silencing of caspase-8. Oncogene 2009; 28:3097-110. [PMID: 19597472 DOI: 10.1038/onc.2009.161] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Evasion of apoptosis can be caused by epigenetic silencing of caspase-8, a key component of the extrinsic apoptosis pathway. Loss of caspase-8 correlates with poor prognosis in medulloblastoma, which highlights the relevance of strategies to upregulate caspase-8 to break apoptosis resistance. Here, we develop a new combinatorial approach, that is treatment using histone deacetylase inhibitors (HDACI) together with interferon (IFN)-gamma, to restore caspase-8 expression and to overcome resistance to the death-receptor ligand TNF-related apoptosis-inducing ligand (TRAIL) in medulloblastoma in vitro and in vivo. HDACI, for example, valproic acid (VA), suberoylanilide hydroxamic acid (SAHA) and MS-275, cooperate with IFN-gamma to upregulate caspase-8 in cancer cells lacking caspase-8, thereby restoring sensitivity to TRAIL-induced apoptosis. Molecular studies show that VA promotes histone acetylation and acts in concert with IFN-gamma to stimulate caspase-8 promoter activity. The resulting increase in caspase-8 mRNA and protein expression leads to enhanced TRAIL-induced activation of caspase-8 at the death-inducing signaling complex, mitochondrial outer-membrane permeabilization and caspase-dependent cell death. Intriguingly, pharmacological or genetic inhibition of caspase-8 also abolishes the VA/IFN-gamma-mediated sensitization for TRAIL-induced apoptosis. It is important to note that VA and IFN-gamma restore caspase-8 expression and sensitivity to TRAIL in primary medulloblastoma samples and significantly potentiate TRAIL-mediated suppression of medulloblastoma growth in vivo. These findings provide the rationale for further (pre)clinical evaluation of VA and IFN-gamma to restore caspase-8 expression and apoptosis sensitivity in cancers with caspase-8 silencing and open new perspectives to overcome TRAIL resistance.
Collapse
|
117
|
Geel TM, Meiss G, van der Gun BT, Kroesen BJ, de Leij LF, Zaremba M, Silanskas A, Kokkinidis M, Pingoud A, Ruiters MH, McLaughlin PM, Rots MG. Endonucleases induced TRAIL-insensitive apoptosis in ovarian carcinoma cells. Exp Cell Res 2009; 315:2487-95. [PMID: 19540229 DOI: 10.1016/j.yexcr.2009.06.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 06/08/2009] [Accepted: 06/08/2009] [Indexed: 11/16/2022]
Abstract
TRAIL induced apoptosis of tumor cells is currently entering phase II clinical settings, despite the fact that not all tumor types are sensitive to TRAIL. TRAIL resistance in ovarian carcinomas can be caused by a blockade upstream of the caspase 3 signaling cascade. We explored the ability of restriction endonucleases to directly digest DNA in vivo, thereby circumventing the caspase cascade. For this purpose, we delivered enzymatically active endonucleases via the cationic amphiphilic lipid SAINT-18((R)):DOPE to both TRAIL-sensitive and insensitive ovarian carcinoma cells (OVCAR and SKOV-3, respectively). Functional nuclear localization after delivery of various endonucleases (BfiI, PvuII and NucA) was indicated by confocal microscopy and genomic cleavage analysis. For PvuII, analysis of mitochondrial damage demonstrated extensive apoptosis both in SKOV-3 and OVCAR. This study clearly demonstrates that cellular delivery of restriction endonucleases holds promise to serve as a novel therapeutic tool for the treatment of resistant ovarian carcinomas.
Collapse
Affiliation(s)
- Tessa M Geel
- Department of Pathology and Medical Biology, Groningen University Institute for Drug Exploration, University Medical Center Groningen, Hanzeplein 1, Groningen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|