101
|
Siegemund M, Seifert O, Zarani M, Džinić T, De Leo V, Göttsch D, Münkel S, Hutt M, Pfizenmaier K, Kontermann RE. An optimized antibody-single-chain TRAIL fusion protein for cancer therapy. MAbs 2016; 8:879-91. [PMID: 27064440 DOI: 10.1080/19420862.2016.1172163] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Fusion proteins combining oligomeric assemblies of a genetically obtained single-chain (sc) variant of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) with antibodies directed against tumor-associated antigens represent a promising strategy to overcome the limited therapeutic activity of conventional soluble TRAIL. To further improve the scTRAIL module in order to obtain a robust, thermostable molecule of high activity, we performed a comprehensive analysis of the minimal TNF homology domain (THD) and optimized linkers between the 3 TRAIL subunits constituting a scTRAIL. Through a stepwise mutagenesis of the N- and C-terminal region and the joining linker sequences, we generated bioactive scTRAIL molecules comprising a covalent linkage of the C-terminal Val280 and the N-terminal position 122 by only 2 amino acid residues in combination with conservative exchanges at positions 122 and 279. The increased thermal stability and solubility of such optimized scTRAIL molecules translated into increased bioactivity in the diabody-scTRAIL (Db-scTRAIL) format, exemplified here for an epidermal growth factor receptor-specific Db-scTRAIL. Additional modifications within the diabody linkers resulted in a fusion protein exerting high, target-dependent apoptosis induction in tumor cell lines in vitro and potent antitumor activity in vivo. Our results illustrate that protein engineering of scTRAIL and associated peptide linkers provides a promising strategy to develop antibody-scTRAIL fusion proteins as effective antitumor therapeutics.
Collapse
Affiliation(s)
- Martin Siegemund
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany
| | - Oliver Seifert
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany
| | | | | | | | - Doris Göttsch
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany
| | - Sabine Münkel
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany
| | - Meike Hutt
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany
| | - Klaus Pfizenmaier
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany
| | - Roland E Kontermann
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany
| |
Collapse
|
102
|
Seow HF, Yip WK, Fifis T. Advances in targeted and immunobased therapies for colorectal cancer in the genomic era. Onco Targets Ther 2016; 9:1899-920. [PMID: 27099521 PMCID: PMC4821380 DOI: 10.2147/ott.s95101] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Targeted therapies require information on specific defective signaling pathways or mutations. Advances in genomic technologies and cell biology have led to identification of new therapeutic targets associated with signal-transduction pathways. Survival times of patients with colorectal cancer (CRC) can be extended with combinations of conventional cytotoxic agents and targeted therapies. Targeting EGFR- and VEGFR-signaling systems has been the major focus for treatment of metastatic CRC. However, there are still limitations in their clinical application, and new and better drug combinations are needed. This review provides information on EGFR and VEGF inhibitors, new therapeutic agents in the pipeline targeting EGFR and VEGFR pathways, and those targeting other signal-transduction pathways, such as MET, IGF1R, MEK, PI3K, Wnt, Notch, Hedgehog, and death-receptor signaling pathways for treatment of metastatic CRC. Additionally, multitargeted approaches in combination therapies targeting negative-feedback loops, compensatory networks, and cross talk between pathways are highlighted. Then, immunobased strategies to enhance antitumor immunity using specific monoclonal antibodies, such as the immune-checkpoint inhibitors anti-CTLA4 and anti-PD1, as well as the challenges that need to be overcome for increased efficacy of targeted therapies, including drug resistance, predictive markers of response, tumor subtypes, and cancer stem cells, are covered. The review concludes with a brief insight into the applications of next-generation sequencing, expression profiling for tumor subtyping, and the exciting progress made in in silico predictive analysis in the development of a prescription strategy for cancer therapy.
Collapse
Affiliation(s)
- Heng Fong Seow
- Immunology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Wai Kien Yip
- Immunology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Theodora Fifis
- Department of Surgery, University of Melbourne, Melbourne, Australia
| |
Collapse
|
103
|
A bispecific fusion protein and a bifunctional enediyne-energized fusion protein consisting of TRAIL, EGFR peptide ligand, and apoprotein of lidamycin against EGFR and DR4/5 show potent antitumor activity. Anticancer Drugs 2016; 26:64-73. [PMID: 25122444 DOI: 10.1097/cad.0000000000000160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) mainly induces apoptosis through the extrinsic death receptor-induced pathway by ligation with death receptor 4 (DR4) and death receptor 5 (DR5). On the basis of the antitumor activity to cancer cells and no cytotoxity to normal cells of TRAIL and the target of the epidermal growth factor receptor (EGFR) ligand peptide, the study constructed a new bispecific fusion protein and a new bifunctional enediyne-energized fusion protein and investigated their antitumor efficacy. Bispecific fusion protein Ec-LDP-TRAIL showed potent binding activity to cancer cell lines with a high expression of EGFR or DR4/DR5 such as A431 and H460 cells, whereas poor binding activity to NIH/3T3 cells with low expressing EGFR and DR4/DR5. Ec-LDP-TRAIL also showed more potent cytotoxicity to A431 and H460 cells than Ec-LDP, which could result from the TRAIL-inducing apoptosis. Results of an in-vivo efficacy study showed that Ec-LDP-TRAIL at a dose of 10 mg/kg decreased the growth of epidermoid carcinoma A431 xenografts by 80.19% (P < 0.01) on day 26. Immunohistochemical detection of nuclear antigen factor Ki-67 suggested that Ec-LDP-TRAIL effectively induced cell necrosis and inhibited cell proliferation of tumor. From IC50 values, bispecific and bifunctional energized fusion protein Ec-LDP-TRAIL-AE was more potent and selective in its cytotoxicity against different carcinoma cell lines than corresponding lidamycin in vitro and induction of the cleavage of poly(ADP-ribose)polymerase was observed in A431 cells treated with Ec-LDP-TRAIL-AE and lidamycin, respectively. Ec-LDP-TRAIL-AE also significantly inhibited the growth of A431 xenografts in a nude mouse model. These properties suggested that Ec-LDP-TRAIL and Ec-LDP-TRAIL-AE may be promising candidates for targeted cancer therapy.
Collapse
|
104
|
Sheng M, Zhao Y, Wang F, Li S, Wang X, Shou T, Luo Y, Tang W. Targeted drugs for unselected patients with advanced non-small-cell lung cancer: a network meta-analysis. J Thorac Dis 2016; 8:98-115. [PMID: 26904218 DOI: 10.3978/j.issn.2072-1439.2016.01.28] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Currently, targeted therapy has shown encouraging treatment benefits in selected patients with advanced non-small cell lung cancer (NSCLC). However, the comparative benefits of targeted drugs and chemotherapy (CT) treatments in unselected patients are not clear. We therefore conduct a network meta-analysis to assess the relative efficacy and safety of these regimens. METHODS PubMed, EMBASE, Cochrane Library and abstracts from major scientific meetings were searched for eligible literatures. The odds ratio (OR) for objective response rate (ORR) and safety was used for pooling effect sizes. Bayesian network meta-analysis was conducted to calculate the efficacy and safety of all included treatments. All tests of statistical significance were two sided. RESULTS A total of 13,060 patients from 24 randomized controlled trials (RCT) were assessed. The targeted agents included bevacizumab (Bev), gefitinib (Gef), erlotinib (Erl) and cetuximab (Cet). Network meta-analysis showed that Bev + CT had a statistically significantly higher incidence of ORR relative to the other six different treatments, including placebo (OR =6.47; 95% CI, 3.85-10.29), Erl (OR =2.81; 95% CI, 2.08-3.70), CT (OR =1.92; 95% CI, 1.61-2.28), Gef (OR =1.40; 95% CI, 1.10-1.75), Erl + CT (OR =1.46; 95% CI, 1.17-1.80) and Gef + CT (OR =1.75; 95% CI, 1.36-2.22), whereas placebo and Erl were associated with statistically significantly lower incidence of ORR. Trend analyses of rank probability revealed that Bev + CT had the highest probability of being the best treatment arm in term of ORR, followed by Cet + CT. Meanwhile, Cet + CT showed significant severer rash and thrombocytopenia compared with Bev + CT. Gef was probable to be the rank 3 for ORR but was associated with relatively low risk for grade ≥3 toxicities. CONCLUSIONS Our study suggested that Bev + CT may offer better ORR in the treatment of unselected patients with advanced NSCLC. Future studies will be needed to investigate whether the increase of ORR with targeted drugs would be translated into survival benefits.
Collapse
Affiliation(s)
- Miaomiao Sheng
- 1 Laboratory of Molecular Genetics of Aging & Tumor, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China ; 2 First People's Hospital of Yunnan Province, Kunming 650032, China
| | - Yueguang Zhao
- 1 Laboratory of Molecular Genetics of Aging & Tumor, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China ; 2 First People's Hospital of Yunnan Province, Kunming 650032, China
| | - Fang Wang
- 1 Laboratory of Molecular Genetics of Aging & Tumor, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China ; 2 First People's Hospital of Yunnan Province, Kunming 650032, China
| | - Shanshan Li
- 1 Laboratory of Molecular Genetics of Aging & Tumor, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China ; 2 First People's Hospital of Yunnan Province, Kunming 650032, China
| | - Xiaojie Wang
- 1 Laboratory of Molecular Genetics of Aging & Tumor, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China ; 2 First People's Hospital of Yunnan Province, Kunming 650032, China
| | - Tao Shou
- 1 Laboratory of Molecular Genetics of Aging & Tumor, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China ; 2 First People's Hospital of Yunnan Province, Kunming 650032, China
| | - Ying Luo
- 1 Laboratory of Molecular Genetics of Aging & Tumor, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China ; 2 First People's Hospital of Yunnan Province, Kunming 650032, China
| | - Wenru Tang
- 1 Laboratory of Molecular Genetics of Aging & Tumor, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China ; 2 First People's Hospital of Yunnan Province, Kunming 650032, China
| |
Collapse
|
105
|
Novel Water-Borne Polyurethane Nanomicelles for Cancer Chemotherapy: Higher Efficiency of Folate Receptors Than TRAIL Receptors in a Cancerous Balb/C Mouse Model. Pharm Res 2016; 33:1426-39. [PMID: 26908046 DOI: 10.1007/s11095-016-1884-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/16/2016] [Indexed: 01/17/2023]
Abstract
PURPOSE Since the introduction of nanocarriers, the delivery of chemotherapeutic agents for treatment of patients with cancer has been possible with better effectiveness. The latest findings are also support that further enhancement in therapeutic effectiveness of these nanocarriers can be attained, if surface decoration with proper targeting agents is considered. METHODS This study aimed at treating a variety of 4T1 murine breast cancer cell line, mainly demonstrating high folate and TRAIL receptor expression of cancerous cells. The therapeutic efficacy of paclitaxel loaded Cremophore EL (Taxol®), paclitaxel loaded waterborne polyurethane nanomicelles (PTX-PU) and paclitaxel loaded waterborne polyurethane nanomicelles conjugated with folate (PTX-PU-FA) and TRAIL (PTX-PU-TRAIL) on treating 4T1 cell was also compared. RESULTS The findings that worth noting are: PTX-PU outperformed Taxol® in a Balb/C mouse model, furthermore, tumor growth was adequately curbed by folate and TRAIL-decorated nanomicelles rather than the unconjugated formulation. Tumors of mice treated with PTX-PU-FA and PTX-PU-TRAIL shrank substantially compared to those treated with Taxol®, PTX-PU and PTX-PU-TRAIL (average 573 mm(3) versus 2640, 846, 717 mm(3) respectively), 45 days subsequent to tumor inoculation. The microscopic study of hematoxylin-eosin stained tumors tissue and apoptotic cell fraction substantiated that the most successful therapeutic effects have been observed for the mice treated with PTX-PU-FA (about 90% in PTX-PU-FA versus 75%, 60%, 15% in PTX-PU-TRAIL, PTX-PU, and Taxol® group respectively). CONCLUSIONS Using folate-targeted nanocarriers to treat cancers characterized by a high level of folate ligand expression is well substantiated by the findings of this study.
Collapse
|
106
|
Miles MA, Shekhar TM, Hall NE, Hawkins CJ. TRAIL causes deletions at the HPRT and TK1 loci of clonogenically competent cells. Mutat Res 2016; 787:15-31. [PMID: 26943263 DOI: 10.1016/j.mrfmmm.2016.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 01/05/2016] [Accepted: 02/02/2016] [Indexed: 12/22/2022]
Abstract
When chemotherapy and radiotherapy are effective, they function by inducing DNA damage in cancerous cells, which respond by undergoing apoptosis. Some adverse effects can result from collateral destruction of non-cancerous cells, via the same mechanism. Therapy-related cancers, a particularly serious adverse effect of anti-cancer treatments, develop due to oncogenic mutations created in non-cancerous cells by the DNA damaging therapies used to eliminate the original cancer. Physiologically achievable concentrations of direct apoptosis inducing anti-cancer drugs that target Bcl-2 and IAP proteins possess negligible mutagenic activity, however death receptor agonists like TRAIL/Apo2L can provoke mutations in surviving cells, probably via caspase-mediated activation of the nuclease CAD. In this study we compared the types of mutations sustained in the HPRT and TK1 loci of clonogenically competent cells following treatment with TRAIL or the alkylating agent ethyl methanesulfonate (EMS). As expected, the loss-of-function mutations in the HPRT or TK1 loci triggered by exposure to EMS were almost all transitions. In contrast, only a minority of the mutations identified in TRAIL-treated clones lacking HPRT or TK1 activity were substitutions. Almost three quarters of the TRAIL-induced mutations were partial or complete deletions of the HPRT or TK1 genes, consistent with sub-lethal TRAIL treatment provoking double strand breaks, which may be mis-repaired by non-homologous end joining (NHEJ). Mis-repair of double-strand breaks following exposure to chemotherapy drugs has been implicated in the pathogenesis of therapy-related cancers. These data suggest that TRAIL too may provoke oncogenic damage to the genomes of surviving cells.
Collapse
Affiliation(s)
- Mark A Miles
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, Victoria, Australia; La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Tanmay M Shekhar
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, Victoria, Australia; La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Nathan E Hall
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia; Life Sciences Computation Centre, Victorian Life Sciences Computation Initiative, Melbourne, Victoria, Australia
| | - Christine J Hawkins
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, Victoria, Australia; La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia.
| |
Collapse
|
107
|
Baig S, Seevasant I, Mohamad J, Mukheem A, Huri HZ, Kamarul T. Potential of apoptotic pathway-targeted cancer therapeutic research: Where do we stand? Cell Death Dis 2016; 7:e2058. [PMID: 26775709 PMCID: PMC4816162 DOI: 10.1038/cddis.2015.275] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/17/2015] [Accepted: 08/25/2015] [Indexed: 12/12/2022]
Abstract
Underneath the intricacy of every cancer lies mysterious events that impel the tumour cell and its posterity into abnormal growth and tissue invasion. Oncogenic mutations disturb the regulatory circuits responsible for the governance of versatile cellular functions, permitting tumour cells to endure deregulated proliferation, resist to proapoptotic insults, invade and erode normal tissues and above all escape apoptosis. This disruption of apoptosis has been highly implicated in various malignancies and has been exploited as an anticancer strategy. Owing to the fact that apoptosis causes minimal inflammation and damage to the tissue, apoptotic cell death-based therapy has been the centre of attraction for the development of anticancer drugs. Increased understanding of the molecular pathways underlying apoptosis has enabled scientists to establish unique approaches targeting apoptosis pathways in cancer therapeutics. In this review, we reconnoitre the two major pathways (intrinsic and extrinsic) targeted cancer therapeutics, steering toward chief modulators of these pathways, such as B-cell lymphoma 2 protein family members (pro- and antiapoptotic), inhibitor of apoptosis proteins, and the foremost thespian of extrinsic pathway regulator, tumour necrosis factor-related apoptosis-inducing agent. Together, we also will have a look from clinical perspective to address the agents (drugs) and therapeutic strategies adopted to target these specific proteins/pathways that have entered clinical trials.
Collapse
Affiliation(s)
- S Baig
- Department of Orthopaedic Surgery, Tissue Engineering Group, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
- Institute of Biological Sciences, Faculty of Science, University of Malaysia, Kuala Lumpur 50603, Malaysia
- Department of Orthopaedic Surgery, University of Malaya, Tissue Engineering Group, Faculty of Medicine, Kuala Lumpur 50603, Malaysia. Tel: +60 3 7967 7022; Fax: +60 3 7949 4642; E-mail: (SB) or Tel: +60 3 7949 2061; Fax: +60 3 7949 4642; E-mail: (TK)
| | - I Seevasant
- Department of Orthopaedic Surgery, Tissue Engineering Group, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - J Mohamad
- Institute of Biological Sciences, Faculty of Science, University of Malaysia, Kuala Lumpur 50603, Malaysia
| | - A Mukheem
- Department of Orthopaedic Surgery, Tissue Engineering Group, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - H Z Huri
- Clinical Investigation Centre, University of Malaya Medical Centre, Kuala Lumpur 50603, Malaysia
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - T Kamarul
- Department of Orthopaedic Surgery, Tissue Engineering Group, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
- Clinical Investigation Centre, University of Malaya Medical Centre, Kuala Lumpur 50603, Malaysia
- Department of Orthopaedic Surgery, University of Malaya, Tissue Engineering Group, Faculty of Medicine, Kuala Lumpur 50603, Malaysia. Tel: +60 3 7967 7022; Fax: +60 3 7949 4642; E-mail: (SB) or Tel: +60 3 7949 2061; Fax: +60 3 7949 4642; E-mail: (TK)
| |
Collapse
|
108
|
Goldar S, Khaniani MS, Derakhshan SM, Baradaran B. Molecular mechanisms of apoptosis and roles in cancer development and treatment. Asian Pac J Cancer Prev 2016; 125:487-9. [PMID: 25824729 DOI: 10.1172/jci80420] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Programmed cell death (PCD) or apoptosis is a mechanism which is crucial for all multicellular organisms to control cell proliferation and maintain tissue homeostasis as well as eliminate harmful or unnecessary cells from an organism. Defects in the physiological mechanisms of apoptosis may contribute to different human diseases like cancer. Identification of the mechanisms of apoptosis and its effector proteins as well as the genes responsible for apoptosis has provided a new opportunity to discover and develop novel agents that can increase the sensitivity of cancer cells to undergo apoptosis or reset their apoptotic threshold. These novel targeted therapies include those targeting anti-apoptotic Bcl-2 family members, p53, the extrinsic pathway, FLICE-inhibitory protein (c-FLIP), inhibitor of apoptosis (IAP) proteins, and the caspases. In recent years a number of these novel agents have been assessed in preclinical and clinical trials. In this review, we introduce some of the key regulatory molecules that control the apoptotic pathways, extrinsic and intrinsic death receptors, discuss how defects in apoptotic pathways contribute to cancer, and list several agents being developed to target apoptosis.
Collapse
Affiliation(s)
- Samira Goldar
- Department of Biochemistry and Clinical Labratorary, Division of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran E-mail :
| | | | | | | |
Collapse
|
109
|
French R, Hayward O, Jones S, Yang W, Clarkson R. Cytoplasmic levels of cFLIP determine a broad susceptibility of breast cancer stem/progenitor-like cells to TRAIL. Mol Cancer 2015; 14:209. [PMID: 26667821 PMCID: PMC4678708 DOI: 10.1186/s12943-015-0478-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 12/03/2015] [Indexed: 01/05/2023] Open
Abstract
Background The clinical application of TRAIL receptor agonists as a novel cancer therapy has been tempered by heterogeneity in tumour responses. This is illustrated in breast cancer, where TRAIL is cytotoxic in cell lines of mesenchymal origin but refractory in lines with an epithelial-like phenotype. However, it is now evident that intra-tumour heterogeneity includes a minority subpopulation of tumour-initiating stem/progenitor-like cells (CSCs) that possess mesenchymal characteristics. We hypothesised therefore that TRAIL may target these phenotypically distinct CSC-like cells that are common to most - if not all - breast cancers, thus impacting on the source of malignancy in a much broader range of breast tumour subtypes than previously envisaged. Methods We used colony formation, tumoursphere, flow cytometry and xenograft tumour initiation assays to observe the TRAIL sensitivity of CSC-like cells in a panel of two mesenchymal-like (TRAIL-sensitive) and four epithelial-like (TRAIL-resistant) breast cancer cell lines. Subcellular levels of the endogenous TRAIL inhibitor, cFLIP, were determined by western blot and immunofluorescence microscopy. The effect of the subcellular redistribution of cFLIP on TRAIL sensitivity and Wnt signalling was determined using cFLIP localisation mutants and the TOPFlash reporter assay respectively. Results TRAIL universally suppressed the clonal expansion of stem/progenitors in all six of the breast cancer cell lines tested, irrespective of their phenotype or overall sensitivity to TRAIL. A concomitant reduction in tumour initiation was confirmed in the TRAIL-resistant epithelial cell line, MCF-7, following serial dilution xenotransplantation. Furthermore TRAIL sensitivity of breast CSCs was inversely proportional to the relative cytoplasmic levels of cFLIP while overexpression of cFLIP in the cytosol using subcellular localization mutants of cFLIP protected these cells from cytotoxicity. The accumulation of nuclear cFLIP on the other hand did not influence TRAIL cytotoxicity but instead promoted Wnt-dependent signalling. Conclusion These data propose a novel role for TRAIL as a selective CSC agent with a broad specificity for both epithelial and mesenchymal breast tumour subtypes. Furthermore we identify a dual role for cFLIP in the maintenance of breast CSC viability, dependent upon its subcellular distribution. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0478-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rhiannon French
- European Cancer Stem Cell Research Institute Cardiff University School of Biosciences, Haydn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| | - Olivia Hayward
- European Cancer Stem Cell Research Institute Cardiff University School of Biosciences, Haydn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| | - Samuel Jones
- European Cancer Stem Cell Research Institute Cardiff University School of Biosciences, Haydn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| | - William Yang
- European Cancer Stem Cell Research Institute Cardiff University School of Biosciences, Haydn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| | - Richard Clarkson
- European Cancer Stem Cell Research Institute Cardiff University School of Biosciences, Haydn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
110
|
Melanoma-Directed Activation of Apoptosis Using a Bispecific Antibody Directed at MCSP and TRAIL Receptor-2/Death Receptor-5. J Invest Dermatol 2015; 136:541-544. [PMID: 26967487 DOI: 10.1016/j.jid.2015.11.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
111
|
Jia D, Yang H, Tao Z, Wan L, Cheng J, Lu X. Preparation and characterization of a novel variant of human tumor necrosis factor-related apoptosis-inducing ligand from the rhesus monkey, Macaca mulatta. Appl Microbiol Biotechnol 2015; 100:3035-47. [DOI: 10.1007/s00253-015-7143-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 10/27/2015] [Accepted: 11/01/2015] [Indexed: 01/10/2023]
|
112
|
Sensitization of Melanoma Cells for Death Ligand TRAIL Is Based on Cell Cycle Arrest, ROS Production, and Activation of Proapoptotic Bcl-2 Proteins. J Invest Dermatol 2015; 135:2794-2804. [DOI: 10.1038/jid.2015.250] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 06/10/2015] [Accepted: 06/16/2015] [Indexed: 12/26/2022]
|
113
|
Activation of surrogate death receptor signaling triggers peroxynitrite-dependent execution of cisplatin-resistant cancer cells. Cell Death Dis 2015; 6:e1926. [PMID: 26492363 PMCID: PMC4632318 DOI: 10.1038/cddis.2015.299] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 09/14/2015] [Accepted: 09/15/2015] [Indexed: 01/03/2023]
Abstract
Platinum-based drugs remain as the cornerstone of cancer chemotherapy; however, development of multidrug resistance presents a therapeutic challenge. This study aims at understanding the molecular mechanisms underlying resistance to cisplatin and unraveling surrogate signaling networks that could revert sensitivity to apoptosis stimuli. We made use of three different sets of cell lines, A549 and H2030 non-small-cell lung cancer (NSCLC) and A2780 ovarian cancer cells and their cisplatin-resistant variants. Here we report that cisplatin-resistant cell lines displayed a multidrug-resistant phenotype. Changes in mitochondrial metabolism and defective mitochondrial signaling were unraveled in the resistant cells. More interestingly, a marked increase in sensitivity of the resistant cells to death receptor-induced apoptosis, in particular TRAIL (TNF-related apoptosis-inducing ligand)-mediated execution, was observed. Although this was not associated with an increase in gene transcription, a significant increase in the localization of TRAIL death receptor, DR4, to the lipid raft subdomains of plasma membrane was detected in the resistant variants. Furthermore, exposure of cisplatin-resistant cells to TRAIL resulted in upregulation of inducible nitric oxide synthase (iNOS) and increase in nitric oxide (NO) production that triggered the generation of peroxynitrite (ONOO−). Scavenging ONOO− rescued cells from TRAIL-induced apoptosis, thereby suggesting a critical role of ONOO− in TRAIL-induced execution of cisplatin-resistant cells. Notably, preincubation of cells with TRAIL restored sensitivity of resistant cells to cisplatin. These data provide compelling evidence for employing strategies to trigger death receptor signaling as a second-line treatment for cisplatin-resistant cancers.
Collapse
|
114
|
Huanna T, Tao Z, Xiangfei W, Longfei A, Yuanyuan X, Jianhua W, Cuifang Z, Manjing J, Wenjing C, Shaochuan Q, Feifei X, Naikang L, Jinchao Z, Chen W. GALNT14 mediates tumor invasion and migration in breast cancer cell MCF-7. Mol Carcinog 2015; 54:1159-71. [PMID: 24962947 DOI: 10.1002/mc.22186] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 04/20/2014] [Accepted: 05/06/2014] [Indexed: 11/10/2022]
Abstract
Aberrant glycosylation is a hallmark of most human cancers and affects many cellular properties, including cell proliferation, apoptosis, differentiation, transformation, migration, invasion, and immune responses. Here, we report that N-acetylgalactosaminyltransferase14 (GALNT14), which mediates the initial step of mucin-type O-glycosylation and is heterogeneously expressed in most breast cancers, plays a critical role in the invasion and migration of breast cancers by regulating the activity of MMP-2 and expression of some EMT genes. We have modulated the expression of GALNT14 by RNAi and overexpression in MCF-7 cells. Overexpression of GALNT14 significantly enhanced cell migration and invasion and promoted the proliferation of breast cancer cells. Knockdown of GALNT14 reduced clonogenicity and attenuates cell migration and cell invasion. The mRNAs for N-cadherin, vimentin, E-cadherin, MMP-2, VEGF, and TGF-β were determined by RT-qPCR involving GALNT14-overexpressing or knockdown MCF-7 cells. Expression profiling revealed the upregulation of N-cadherin, vimentin, MMP-2, VEGF, TGF-β and the downregulation of E-cadherin in GALNT14 overexpressing cells, with the opposite seen in GALNT14 knockdowns. Gelatin zymography analysis further indicated that overexpression of GALNT14 increased MMP-2 activity in MCF-7 cells. Conversely, downregulation of GALNT14 reduced MMP-2 activity. Promoter analysis revealed that GALNT14 stimulates MMP-2 expression through the AP-1-binding site. Western blot analyses showed that knockdown of GALNT14 significantly reduced the expression of an oncoprotein mucin 1 (MUC1). These findings indicate that GALNT14 contributes to breast cancer invasion by altering the cell proliferation, motility, expression levels of EMT genes, and by stimulating MMP-2 activity, suggesting GALNT14 may be a potential target for breast cancer treatment.
Collapse
Affiliation(s)
- Tian Huanna
- College of Life Sciences, Hebei University, Baoding, Hebei, PR China
- Basic Medical Institute, Chengde Medical College, Chengde, Hebei, PR China
| | - Zuo Tao
- College of Life Sciences, Hebei University, Baoding, Hebei, PR China
| | - Wang Xiangfei
- College of Life Sciences, Hebei University, Baoding, Hebei, PR China
| | - An Longfei
- College of Life Sciences, Hebei University, Baoding, Hebei, PR China
| | - Xie Yuanyuan
- College of Life Sciences, Hebei University, Baoding, Hebei, PR China
| | - Wang Jianhua
- College of Life Sciences, Hebei University, Baoding, Hebei, PR China
| | - Zhang Cuifang
- College of Life Sciences, Hebei University, Baoding, Hebei, PR China
| | - Jiao Manjing
- College of Life Sciences, Hebei University, Baoding, Hebei, PR China
| | - Cao Wenjing
- College of Life Sciences, Hebei University, Baoding, Hebei, PR China
| | - Qin Shaochuan
- College of Life Sciences, Hebei University, Baoding, Hebei, PR China
| | - Xu Feifei
- College of Life Sciences, Hebei University, Baoding, Hebei, PR China
| | - Li Naikang
- College of Life Sciences, Hebei University, Baoding, Hebei, PR China
| | - Zhang Jinchao
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding, Hebei, PR China
- Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science, Hebei University, Baoding, Hebei, PR China
| | - Wu Chen
- College of Life Sciences, Hebei University, Baoding, Hebei, PR China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding, Hebei, PR China
| |
Collapse
|
115
|
Reddy A, Growney JD, Wilson NS, Emery CM, Johnson JA, Ward R, Monaco KA, Korn J, Monahan JE, Stump MD, Mapa FA, Wilson CJ, Steiger J, Ledell J, Rickles RJ, Myer VE, Ettenberg SA, Schlegel R, Sellers WR, Huet HA, Lehár J. Gene Expression Ratios Lead to Accurate and Translatable Predictors of DR5 Agonism across Multiple Tumor Lineages. PLoS One 2015; 10:e0138486. [PMID: 26378449 PMCID: PMC4574744 DOI: 10.1371/journal.pone.0138486] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 08/30/2015] [Indexed: 12/16/2022] Open
Abstract
Death Receptor 5 (DR5) agonists demonstrate anti-tumor activity in preclinical models but have yet to demonstrate robust clinical responses. A key limitation may be the lack of patient selection strategies to identify those most likely to respond to treatment. To overcome this limitation, we screened a DR5 agonist Nanobody across >600 cell lines representing 21 tumor lineages and assessed molecular features associated with response. High expression of DR5 and Casp8 were significantly associated with sensitivity, but their expression thresholds were difficult to translate due to low dynamic ranges. To address the translational challenge of establishing thresholds of gene expression, we developed a classifier based on ratios of genes that predicted response across lineages. The ratio classifier outperformed the DR5+Casp8 classifier, as well as standard approaches for feature selection and classification using genes, instead of ratios. This classifier was independently validated using 11 primary patient-derived pancreatic xenograft models showing perfect predictions as well as a striking linearity between prediction probability and anti-tumor response. A network analysis of the genes in the ratio classifier captured important biological relationships mediating drug response, specifically identifying key positive and negative regulators of DR5 mediated apoptosis, including DR5, CASP8, BID, cFLIP, XIAP and PEA15. Importantly, the ratio classifier shows translatability across gene expression platforms (from Affymetrix microarrays to RNA-seq) and across model systems (in vitro to in vivo). Our approach of using gene expression ratios presents a robust and novel method for constructing translatable biomarkers of compound response, which can also probe the underlying biology of treatment response.
Collapse
Affiliation(s)
- Anupama Reddy
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
- * E-mail:
| | - Joseph D. Growney
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Nick S. Wilson
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Caroline M. Emery
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Jennifer A. Johnson
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Rebecca Ward
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Kelli A. Monaco
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Joshua Korn
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - John E. Monahan
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Mark D. Stump
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Felipa A. Mapa
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Christopher J. Wilson
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Janine Steiger
- Horizon CombinatoRx, Cambridge, MA, United States of America
| | - Jebediah Ledell
- Horizon CombinatoRx, Cambridge, MA, United States of America
| | | | - Vic E. Myer
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Seth A. Ettenberg
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Robert Schlegel
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - William R. Sellers
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Heather A. Huet
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Joseph Lehár
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| |
Collapse
|
116
|
Delivery of tumor-homing TRAIL sensitizer with long-acting TRAIL as a therapy for TRAIL-resistant tumors. J Control Release 2015; 220:671-81. [PMID: 26381901 DOI: 10.1016/j.jconrel.2015.09.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 09/02/2015] [Accepted: 09/09/2015] [Indexed: 11/22/2022]
Abstract
Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) has attracted great interest as a cancer therapy because it selectively induces death receptor (DR)-mediated apoptosis in cancer cells while sparing normal tissue. However, recombinant human TRAIL demonstrates limited therapeutic efficacy in clinical trials, possibly due to TRAIL-resistance of primary cancers and its inherent short half-life. Here we introduce drug delivery approaches to maximize in vivo potency of TRAIL in TRAIL-resistant tumor xenografts by (1) extending the half-life of the ligand with PEGylated TRAIL (TRAILPEG) and (2) concentrating a TRAIL sensitizer, selected from in vitro screening, in tumors via tumor-homing nanoparticles. Antitumor efficacy of TRAILPEG with tumor-homing sensitizer was evaluated in HCT116 and HT-29 colon xenografts. Western blot, real-time PCR, immunohistochemistry and cell viability assays were employed to investigate mechanisms of action and antitumor efficacy of the combination. We discovered that doxorubicin (DOX) sensitizes TRAIL-resistant HT-29 colon cancer cells to TRAIL by upregulating mRNA expression of DR5 by 60% in vitro. Intravenously administered free DOX does not effectively upregulate DR5 in tumor tissues nor demonstrate synergy with TRAILPEG in HT-29 xenografts, but rather introduces significant systemic toxicity. Alternatively, when DOX was encapsulated in hyaluronic acid-based nanoparticles (HAC/DOX) and intravenously administered with TRAILPEG, DR-mediated apoptosis was potentiated in HT-29 tumors by upregulating DR5 protein expression by 70% and initiating both extrinsic and intrinsic apoptotic pathways with reduced systemic toxicity compared to HAC/DOX or free DOX combined with TRAILPEG (80% vs. 40% survival rate; 75% vs. 34% tumor growth inhibition). This study demonstrates a unique approach to overcome TRAIL-based therapy drawbacks using sequential administration of a tumor-homing TRAIL sensitizer and long-acting TRAILPEG.
Collapse
|
117
|
Amarante-Mendes GP, Griffith TS. Therapeutic applications of TRAIL receptor agonists in cancer and beyond. Pharmacol Ther 2015; 155:117-31. [PMID: 26343199 DOI: 10.1016/j.pharmthera.2015.09.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
TRAIL/Apo-2L is a member of the TNF superfamily first described as an apoptosis-inducing cytokine in 1995. Similar to TNF and Fas ligand, TRAIL induces apoptosis in caspase-dependent manner following TRAIL death receptor trimerization. Because tumor cells were shown to be particularly sensitive to this cytokine while normal cells/tissues proved to be resistant along with being able to synthesize and release TRAIL, it was rapidly appreciated that TRAIL likely served as one of our major physiologic weapons against cancer. In line with this, a number of research laboratories and pharmaceutical companies have attempted to exploit the ability of TRAIL to kill cancer cells by developing recombinant forms of TRAIL or TRAIL receptor agonists (e.g., receptor-specific mAb) for therapeutic purposes. In this review article we will describe the biochemical pathways used by TRAIL to induce different cell death programs. We will also summarize the clinical trials related to this pathway and discuss possible novel uses of TRAIL-related therapies. In recent years, the physiological importance of TRAIL has expanded beyond being a tumoricidal molecule to one critical for a number of clinical settings - ranging from infectious disease and autoimmunity to cardiovascular anomalies. We will also highlight some of these conditions where modulation of the TRAIL/TRAIL receptor system may be targeted in the future.
Collapse
Affiliation(s)
- Gustavo P Amarante-Mendes
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, SP, Brazil; Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia, Brazil.
| | - Thomas S Griffith
- Department of Urology, Masonic Cancer Center, Center for Immunology, University of Minnesota, Minneapolis, MN, USA; Minneapolis VA Health Care System, Minneapolis, MN 55417, USA.
| |
Collapse
|
118
|
Hylander BL, Sen A, Beachy SH, Pitoniak R, Ullas S, Gibbs JF, Qiu J, Prey JD, Fetterly GJ, Repasky EA. Tumor priming by Apo2L/TRAIL reduces interstitial fluid pressure and enhances efficacy of liposomal gemcitabine in a patient derived xenograft tumor model. J Control Release 2015; 217:160-9. [PMID: 26342663 DOI: 10.1016/j.jconrel.2015.08.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 08/10/2015] [Accepted: 08/24/2015] [Indexed: 12/20/2022]
Abstract
Interstitial fluid pressure (IFP) is elevated in tumors and high IFP, a negative cancer prognosticator, is known to limit the uptake and efficacy of anti-tumor therapeutics. Approaches that alter the tumor microenvironment and enhance uptake of therapeutics are collectively referred to as tumor "priming". Here we show that the cytotoxic biological therapy Apo2L/TRAIL can prime the tumor microenvironment and significantly lower IFP in three different human tumor xenograft models (Colo205, MiaPaca-2 and a patient gastrointestinal adenocarcinoma tumor xenograft). We found that a single dose of Apo2L/TRAIL resulted in a wave of apoptosis which reached a maximum at 8h post-treatment. Apoptotic debris subsequently disappeared concurrent with an increase in macrophage infiltration. By 24h post-treatment, treated tumors appeared less condensed with widening of the stromal areas which increased at 48 and 72h. Analysis of tumor vasculature demonstrated a significant increase in overall vessel size at 48 and 72h although the number of vessels did not change. Notably, IFP was significantly reduced in these tumors by 48h after Apo2L/TRAIL treatment. Administration of gemcitabine at this time resulted in increased tumor uptake of both gemcitabine and liposomal gemcitabine and significantly improved anti-tumor efficacy of liposomal gemcitabine. These results suggest that Apo2L/TRAIL has a potential as a tumor priming agent and provides a rationale for developing a sequencing schema for combination therapy such that an initial dose of Apo2L/TRAIL would precede administration of gemcitabine or other therapies.
Collapse
Affiliation(s)
- Bonnie L Hylander
- Dept. of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA.
| | - Arindam Sen
- Dept. of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Sarah H Beachy
- Dept. of Cell Stress, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Rose Pitoniak
- Dept. of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Soumya Ullas
- Dept. of Cell Stress, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - John F Gibbs
- Dept. of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Jingxin Qiu
- Dept. of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Joshua D Prey
- Dept. of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | | |
Collapse
|
119
|
Chen J, Lu Y, Zheng Y. Incidence and risk of hypertension with bevacizumab in non-small-cell lung cancer patients: a meta-analysis of randomized controlled trials. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:4751-60. [PMID: 26316712 PMCID: PMC4547635 DOI: 10.2147/dddt.s87258] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Aim A study was conducted to determine the overall risk and incidence of hypertension with bevacizumab in non-small-cell lung cancer (NSCLC) patients. Materials and methods Electronic databases such as the Embase, PubMed, and Cochrane Library were searched for related trials. Statistical analyses were conducted to calculate the overall incidence rates, odds ratios (ORs), and 95% confidence intervals (CIs) by using either random-effect or fixed-effect models depending on the heterogeneity. Results A total of 3,155 subjects from nine studies were included. The overall incidences of all-grade and high-grade hypertension in NSCLC patients were 19.55% (95% CI 10.17%–34.3%) and 6.95% (95% CI 5.81%–8.30%). Bevacizumab use was associated with a significantly increased risk in all-grade hypertension (OR 8.07, 95% CI 3.87–16.85; P=0.0002) and high-grade hypertension (OR 5.93, 95% CI 3.41–10.32; P<0.0001). No evidence of publication bias was determined for the ORs of hypertension in our meta-analysis. Conclusion Bevacizumab is associated with a significantly increased risk of hypertension development in NSCLC patients. Early monitoring and effective management of hypertension might be important steps for the safe use of this drug.
Collapse
Affiliation(s)
- Jian Chen
- Intensive Care Unit, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yingfeng Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yunliang Zheng
- Research Center for Clinical Pharmacy, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
120
|
Wang H, Yang T, Wu X. 5-Fluorouracil preferentially sensitizes mutant KRAS non-small cell lung carcinoma cells to TRAIL-induced apoptosis. Mol Oncol 2015; 9:1815-24. [PMID: 26130327 DOI: 10.1016/j.molonc.2015.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/01/2015] [Accepted: 06/09/2015] [Indexed: 12/12/2022] Open
Abstract
Mutations in the KRAS gene are very common in non-small cell lung cancer (NSCLC), but effective therapies targeting KRAS have yet to be developed. Interest in tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a potent inducer of cell death, has increased following the observation that TRAIL can selectively kill a wide variety of human cancer cells without killing normal cells both in vitro and in xenograft models. However, results from clinical trials of TRAIL-based therapy are disappointingly modest at best and many have demonstrated a lack of therapeutic benefit. Current research has focused on selecting a subpopulation of cancer patients who may benefit from TRAIL-based therapy and identifying best drugs to work with TRAIL. In the current study, we found that NSCLC cells with a KRAS mutation were highly sensitive to treatment with TRAIL and 5-fluorouracil (5FU). Compared with other chemotherapeutic agents, 5FU displayed the highest synergy with TRAIL in inducing apoptosis in mutant KRAS NSCLC cells. We also found that, on a mechanistic level, 5FU preferentially repressed survivin expression and induced expression of TRAIL death receptor 5 to sensitize NSCLC cells to TRAIL. The combination of low-dose 5FU and TRAIL strongly inhibited xenograft tumor growth in mice. Our results suggest that the combination of TRAIL and 5FU may be beneficial for patients with mutant KRAS NSCLC.
Collapse
Affiliation(s)
- Haizhen Wang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, USA
| | - Tao Yang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, USA
| | - Xiangwei Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, USA.
| |
Collapse
|
121
|
Lim B, Allen JE, Prabhu VV, Talekar MK, Finnberg NK, El-Deiry WS. Targeting TRAIL in the treatment of cancer: new developments. Expert Opin Ther Targets 2015; 19:1171-85. [DOI: 10.1517/14728222.2015.1049838] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
122
|
Zhang M, Harashima N, Moritani T, Huang W, Harada M. The Roles of ROS and Caspases in TRAIL-Induced Apoptosis and Necroptosis in Human Pancreatic Cancer Cells. PLoS One 2015; 10:e0127386. [PMID: 26000607 PMCID: PMC4441514 DOI: 10.1371/journal.pone.0127386] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 04/15/2015] [Indexed: 01/22/2023] Open
Abstract
Death signaling provided by tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) can induce death in cancer cells with little cytotoxicity to normal cells; this cell death has been thought to involve caspase-dependent apoptosis. Reactive oxygen species (ROS) are also mediators that induce cell death, but their roles in TRAIL-induced apoptosis have not been elucidated fully. In the current study, we investigated ROS and caspases in human pancreatic cancer cells undergoing two different types of TRAIL-induced cell death, apoptosis and necroptosis. TRAIL treatment increased ROS in two TRAIL-sensitive pancreatic cancer cell lines, MiaPaCa-2 and BxPC-3, but ROS were involved in TRAIL-induced apoptosis only in MiaPaCa-2 cells. Unexpectedly, inhibition of ROS by either N-acetyl-L-cysteine (NAC), a peroxide inhibitor, or Tempol, a superoxide inhibitor, increased the annexin V-/propidium iodide (PI)+ early necrotic population in TRAIL-treated cells. Additionally, both necrostatin-1, an inhibitor of receptor-interacting protein kinase 1 (RIP1), and siRNA-mediated knockdown of RIP3 decreased the annexin V-/PI+ early necrotic population after TRAIL treatment. Furthermore, an increase in early apoptosis was induced in TRAIL-treated cancer cells under inhibition of either caspase-2 or -9. Caspase-2 worked upstream of caspase-9, and no crosstalk was observed between ROS and caspase-2/-9 in TRAIL-treated cells. Together, these results indicate that ROS contribute to TRAIL-induced apoptosis in MiaPaCa-2 cells, and that ROS play an inhibitory role in TRAIL-induced necroptosis of MiaPaCa-2 and BxPC-3 cells, with caspase-2 and -9 playing regulatory roles in this process.
Collapse
Affiliation(s)
- Min Zhang
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Shengli Street, Yinchuan, China
- Department of Immunology, Shimane University Faculty of Medicine, Izumi, Shimane, Japan
| | - Nanae Harashima
- Department of Immunology, Shimane University Faculty of Medicine, Izumi, Shimane, Japan
| | - Tamami Moritani
- Department of Immunology, Shimane University Faculty of Medicine, Izumi, Shimane, Japan
| | - Weidong Huang
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Shengli Street, Yinchuan, China
| | - Mamoru Harada
- Department of Immunology, Shimane University Faculty of Medicine, Izumi, Shimane, Japan
- * E-mail:
| |
Collapse
|
123
|
Immunotherapy with liposome-bound TRAIL overcomes partial protection to soluble TRAIL-induced apoptosis offered by down-regulation of Bim in leukemic cells. Clin Transl Oncol 2015; 17:657-67. [PMID: 25967100 DOI: 10.1007/s12094-015-1295-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 04/30/2015] [Indexed: 01/02/2023]
Abstract
PURPOSE Human Apo2-Ligand/TRAIL secreted by natural killer cells and cytotoxic T lymphocytes plays an important role immunosurveillance controlling tumor growth and metastasis. Moreover, the fact that Apo2L/TRAIL is capable of inducing cell death in tumor cells but not in normal cells makes this death ligand a promising anti-tumor agent. Previous data from our group demonstrated that Apo2L/TRAIL was physiologically released as transmembrane protein inserted in lipid vesicles, called exosomes. Recently, we demonstrated that artificial lipid nanoparticles coated with bioactive Apo2L/TRAIL (LUV-TRAIL) resembling the natural exosomes, greatly improved Apo2L/TRAIL activity and were able to induce apoptosis in hematological malignancies. In this study, we have deepened in the underlying mechanism of action of LUV-TRAIL in hematologic cells. METHODS/PATIENTS Cytotoxic ability of LUV-TRAIL was assessed on Jurkat cells either over-expressing the anti-apoptotic protein Mcl1 or down-regulating the pro-apoptotic protein Bim previously generated in our laboratory. We also tested LUV-TRAIL cytotoxic ability against primary human leukemic cells from T-cell ALL patient. RESULTS Silencing Bim but not Mcl-1 over-expression partially protects Jurkat cells from apoptosis induced by sTRAIL. LUV-TRAIL induced caspase-8 and caspase-3 activation and killed Jurkat-Mcl1 and Jurkat-shBim more efficiently than sTRAIL independently of the mitochondrial pathway. On the other hand, LUV-TRAIL were clearly more cytotoxic against primary leukemic cells from a T-cell ALL patient than sTRAIL. CONCLUSION Tethering Apo2L/TRAIL to the surface of lipid nanoparticles greatly increases its bioactivity and could be of potential use in anti-tumor therapeutics.
Collapse
|
124
|
Investigation of Prognostic Factors Affecting Efficacy in Carboplatin- and Paclitaxel-based First-line Chemotherapies for Advanced Non-small-cell Lung Cancer. TUMORI JOURNAL 2015; 101:424-32. [DOI: 10.5301/tj.5000333] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2015] [Indexed: 11/20/2022]
Abstract
Aims and Background First-line chemotherapies for advanced non-small-cell lung cancer (NSCLC) are platinum-based regimens. An analysis of efficacy outcomes has not yet been systematically performed and fully evaluated using large patient cohorts in each of the platinum-based chemotherapies. The present meta-analysis aims to investigate prognostic factors affecting overall survival (OS), progression-free survival (PFS) or time to progression (TTP), and overall response rate (ORR) in carboplatin and paclitaxel-based first-line chemotherapies for advanced NSCLC. Methods We performed a literature search in PubMed for randomized phase II and III clinical trials in patients with NSCLC treated with carboplatin and paclitaxel as first-line chemotherapy published from January 2000 to December 2013 to investigate prognostic factors affecting OS, PFS or TTP, and ORR by linear regression analysis and logistic regression analysis. Results We identified 61 treatment arms in 53 phase II and III clinical trials for the analysis. Asian region was found to be a prognostic factor that affects longer OS in treatment with carboplatin and paclitaxel as first-line chemotherapy. In addition, we identified weekly administration schedule of paclitaxel, Asian region, and lower percentage of patients with adenocarcinoma as factors affecting higher ORR. Conclusions Our findings of prognostic factors affecting ORR and OS in carboplatin and paclitaxel-based chemotherapies as first-line therapy should be considered in the interpretation of efficacy results in global phase II and III clinical trials.
Collapse
|
125
|
Enhancing the antitumor efficacy of a cell-surface death ligand by covalent membrane display. Proc Natl Acad Sci U S A 2015; 112:5679-84. [PMID: 25902490 DOI: 10.1073/pnas.1418962112] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
TNF superfamily death ligands are expressed on the surface of immune cells and can trigger apoptosis in susceptible cancer cells by engaging cognate death receptors. A recombinant soluble protein comprising the ectodomain of Apo2 ligand/TNF-related apoptosis-inducing ligand (Apo2L/TRAIL) has shown remarkable preclinical anticancer activity but lacked broad efficacy in patients, possibly owing to insufficient exposure or potency. We observed that antibody cross-linking substantially enhanced cytotoxicity of soluble Apo2L/TRAIL against diverse cancer cell lines. Presentation of the ligand on glass-supported lipid bilayers enhanced its ability to drive receptor microclustering and apoptotic signaling. Furthermore, covalent surface attachment of Apo2L/TRAIL onto liposomes--synthetic lipid-bilayer nanospheres--similarly augmented activity. In vivo, liposome-displayed Apo2L/TRAIL achieved markedly better exposure and antitumor activity. Thus, covalent synthetic-membrane attachment of a cell-surface ligand enhances efficacy, increasing therapeutic potential. These findings have translational implications for liposomal approaches as well as for Apo2L/TRAIL and other clinically relevant TNF ligands.
Collapse
|
126
|
Dai X, Zhang J, Arfuso F, Chinnathambi A, Zayed ME, Alharbi SA, Kumar AP, Ahn KS, Sethi G. Targeting TNF-related apoptosis-inducing ligand (TRAIL) receptor by natural products as a potential therapeutic approach for cancer therapy. Exp Biol Med (Maywood) 2015; 240:760-73. [PMID: 25854879 DOI: 10.1177/1535370215579167] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to selectively induce apoptotic cell death in various tumor cells by engaging its death-inducing receptors (TRAIL-R1 and TRAIL-R2). This property has led to the development of a number of TRAIL-receptor agonists such as the soluble recombinant TRAIL and agonistic antibodies, which have shown promising anticancer activity in preclinical studies. However, besides activating caspase-dependent apoptosis in several cancer cells, TRAIL may also activate nonapoptotic signal transduction pathways such as nuclear factor-kappa B, mitogen-activated protein kinases, AKT, and signal transducers and activators of transcription 3, which may contribute to TRAIL resistance that is being now frequently encountered in various cancers. TRAIL resistance can be overcome by the application of efficient TRAIL-sensitizing pharmacological agents. Natural compounds have shown a great potential in sensitizing cells to TRAIL treatment through suppression of distinct survival pathways. In this review, we have summarized both apoptotic and nonapoptotic pathways activated by TRAIL, as well as recent advances in developing TRAIL-receptor agonists for cancer therapy. We also briefly discuss combination therapies that have shown great potential in overcoming TRAIL resistance in various tumors.
Collapse
Affiliation(s)
- Xiaoyun Dai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jingwen Zhang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Frank Arfuso
- School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Western Australia 6009, Australia
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - M E Zayed
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Western Australia 6009, Australia Cancer Science Institute of Singapore, Centre for Translational Medicine, Singapore 117599, Singapore Department of Biological Sciences, University of North Texas, Denton, TX 76203, USA
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Western Australia 6009, Australia Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| |
Collapse
|
127
|
Dulanermin with rituximab in patients with relapsed indolent B-cell lymphoma: an open-label phase 1b/2 randomised study. LANCET HAEMATOLOGY 2015; 2:e166-74. [PMID: 26687959 DOI: 10.1016/s2352-3026(15)00026-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/18/2015] [Accepted: 02/19/2015] [Indexed: 01/03/2023]
Abstract
BACKGROUND Dulanermin-a non-polyhistidine-tagged soluble recombinant human apoptosis ligand 2 (Apo2L) or tumour-necrosis-factor-related apoptosis-inducing-ligand (TRAIL)-has pro-apoptotic activity in a range of cancers and synergistic preclinical activity with rituximab against lymphoma in vivo. We aimed to assess the safety, pharmacokinetics, and efficacy of dulanermin and rituximab in patients with relapsed indolent B-cell non-Hodgkin lymphoma. METHODS We did an open-label phase 1b/2 randomised study. Four study centres in the USA enrolled patients into phase 1b, and 27 study centres in the USA, Italy, Australia, France, Czech Republic, New Zealand, and Poland enrolled patients into phase 2. In phase 1b, patients (age ≥18 years) with indolent B-cell non-Hodgkin lymphoma with stable disease or better lasting at least 6 months after the most recent rituximab-containing regimen were included. In phase 2, patients (age ≥18 years) with follicular lymphoma grades 1-3a were included. In phase 1b, patients received 4 mg/kg or 8 mg/kg intravenous dulanermin on days 1-5 of up to four 21-day cycles and intravenous rituximab 375 mg/m(2) weekly for up to eight doses. In phase 2, patients were randomly assigned (1:1:1) centrally by an interactive voice response system to dulanermin (8 mg/kg for a maximum of four 21-day cycles), rituximab (375 mg/m(2) weekly for up to eight doses), or both in combination, stratified by baseline follicular lymphoma International Prognostic Index (0-3 vs 4-5) and geographic site (USA vs non-USA). The primary endpoints of the phase 1b study were the safety, tolerability, and pharmacokinetics of dulanermin with rituximab. The primary endpoint of phase 2 was the proportion of patients who achieved an objective response. All patients who received any dose of study drug were included in safety analyses. Efficacy analyses were per protocol. Treatment was open label; all patients and investigators were unmasked to treatment allocation. This study is registered with ClinicalTrials.gov, NCT00400764. FINDINGS Between June 6, 2006, and Feb 15, 2007, 12 patients were enrolled in phase 1b, and between April 4, 2007, and April 20, 2009, 60 patients were enrolled in phase 2, of whom 59 were included in safety analyses and 58 in efficacy analyses. No dose-limiting toxic effects were noted in phase 1b. The most common grade 1-2 adverse events in phase 1b were fatigue (nine; 75%), rash (five; 42%), and chills, decreased appetite, diarrhoea, and nausea (four each; 33%). 19 grade 3 or higher adverse effects were noted in five (42%) patients, with 14 occurring in one patient. After treatment with 8 mg/kg of dulanermin, in six patients the mean serum peak concentration was 80 μg/mL, dropping below the minimum detectable concentration (2 ng/mL) within 24 h after the dose. The mean steady state peak and trough concentrations of rituximab were 461 μg/mL (SD 97.5) and 303 μg/mL (92.8), respectively. In phase 2, eight (14%) of 59 patients experienced 12 grade 3 or higher adverse events. In phase 2, objective responses were noted in 14 of 22 (63.6%, 95% CI 41.8-81.3) patients treated with rituximab only, 16 of 25 (64.0%, 43.1-81.5) treated with dulanermin and rituximab, and one of 11 (9.1%, 0.5-39.0) treated with dulanermin only. The study was terminated early, on May 5, 2010, because of an absence of efficacy in the combination group. INTERPRETATION The addition of dulanermin to rituximab in patients with indolent B-cell non-Hodgkin lymphoma was tolerable but did not lead to increased objective responses. This combination is not being developed further in non-Hodgkin lymphoma. FUNDING Genentech and Amgen.
Collapse
|
128
|
Quintavalle C, Condorelli G. Dulanermin in cancer therapy: still much to do. Transl Lung Cancer Res 2015; 1:158-9. [PMID: 25806175 DOI: 10.3978/j.issn.2218-6751.2012.02.03] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 02/29/2012] [Indexed: 01/25/2023]
Affiliation(s)
- Cristina Quintavalle
- Department of Cellular and Molecular Biology and Pathology, "Federico II" University of Naples, Naples, Italy ; ; IEOS, CNR, Naples, Italy
| | - Gerolama Condorelli
- Department of Cellular and Molecular Biology and Pathology, "Federico II" University of Naples, Naples, Italy ; ; IEOS, CNR, Naples, Italy ; ; Facoltà di Scienze Biotecnologiche, "Federico II" University of Naples, Naples, Italy
| |
Collapse
|
129
|
Twomey JD, Kim SR, Zhao L, Bozza WP, Zhang B. Spatial dynamics of TRAIL death receptors in cancer cells. Drug Resist Updat 2015; 19:13-21. [PMID: 25840763 DOI: 10.1016/j.drup.2015.02.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 02/15/2015] [Accepted: 02/20/2015] [Indexed: 12/24/2022]
Abstract
TNF-related apoptosis inducing ligand (TRAIL) selectively induces apoptosis in cancer cells without harming most normal cells. Currently, multiple clinical trials are underway to evaluate the antitumor activity of recombinant human TRAIL (rhTRAIL) and agonistic antibodies that target death receptors (DRs) 4 or 5. It is encouraging that these products have shown a tolerated safety profile in early phase studies. However, their therapeutic potential is likely limited by the emergence of tumor drug resistance phenomena. Increasing evidence indicates that TRAIL DRs are deficient on the plasma membrane of some cancer cells despite their total protein expression. Notably, the lack of surface DR4/DR5 is sufficient to render cancers resistant to TRAIL-induced apoptosis, regardless of the status of other apoptosis signaling components. The current review highlights recent findings on the dynamic expression of TRAIL death receptors, including the regulatory roles of endocytosis, autophagy, and Ras GTPase-mediated signaling events. This information could aid in the identification of novel predictive biomarkers of tumor response as well as the development of combinational drugs to overcome or bypass tumor drug resistance to TRAIL receptor-targeted therapies.
Collapse
Affiliation(s)
- Julianne D Twomey
- Division of Biotechnology Review and Research IV, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Su-Ryun Kim
- Division of Biotechnology Review and Research IV, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Liqun Zhao
- Division of Biotechnology Review and Research IV, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - William P Bozza
- Division of Biotechnology Review and Research IV, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Baolin Zhang
- Division of Biotechnology Review and Research IV, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States.
| |
Collapse
|
130
|
Unexpected hepatotoxicity in a phase I study of TAS266, a novel tetravalent agonistic Nanobody® targeting the DR5 receptor. Cancer Chemother Pharmacol 2015; 75:887-95. [DOI: 10.1007/s00280-015-2712-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/19/2015] [Indexed: 11/26/2022]
|
131
|
Fulda S. Safety and tolerability of TRAIL receptor agonists in cancer treatment. Eur J Clin Pharmacol 2015; 71:525-7. [DOI: 10.1007/s00228-015-1823-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/11/2015] [Indexed: 01/22/2023]
|
132
|
Choi SH, Tamura K, Khajuria RK, Bhere D, Nesterenko I, Lawler J, Shah K. Antiangiogenic variant of TSP-1 targets tumor cells in glioblastomas. Mol Ther 2015; 23:235-43. [PMID: 25358253 PMCID: PMC4445617 DOI: 10.1038/mt.2014.214] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 10/28/2014] [Indexed: 12/11/2022] Open
Abstract
Three type-1 repeat (3TSR) domain of thrombospondin-1 is known to have anti-angiogenic effects by targeting tumor-associated endothelial cells, but its effect on tumor cells is unknown. This study explored the potential of 3TSR to target glioblastoma (GBM) cells in vitro and in vivo. We show that 3TSR upregulates death receptor (DR) 4/5 expression in a CD36-dependent manner and primes resistant GBMs to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced caspase-8/3/7 mediated apoptosis. We engineered human mesenchymal stem cells (MSC) for on-site delivery of 3TSR and a potent and secretable variant of TRAIL (S-TRAIL) in an effort to simultaneously target tumor cells and associated endothelial cells and circumvent issues of systemic delivery of drugs across the blood-brain barrier. We show that MSC-3TSR/S-TRAIL inhibits tumor growth in an expanded spectrum of GBMs. In vivo, a single administration of MSC-3TSR/S-TRAIL significantly targets both tumor cells and vascular component of GBMs, inhibits tumor progression, and extends survival of mice bearing highly vascularized GBM. The ability of 3TSR/S-TRAIL to simultaneously act on tumor cells and tumor-associated endothelial cells offers a great potential to target a broad spectrum of cancers and translate 3TSR/TRAIL therapies into clinics.
Collapse
Affiliation(s)
- Sung Hugh Choi
- Molecular Neurotherapy and Imaging Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kaoru Tamura
- Molecular Neurotherapy and Imaging Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rajiv Kumar Khajuria
- Molecular Neurotherapy and Imaging Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Deepak Bhere
- Molecular Neurotherapy and Imaging Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Irina Nesterenko
- Molecular Neurotherapy and Imaging Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jack Lawler
- Division of Experimental Pathology, Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Khalid Shah
- Molecular Neurotherapy and Imaging Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
133
|
Uckun FM, Myers DE, Qazi S, Ozer Z, Rose R, D'Cruz OJ, Ma H. Recombinant human CD19L-sTRAIL effectively targets B cell precursor acute lymphoblastic leukemia. J Clin Invest 2015; 125:1006-18. [PMID: 25621496 DOI: 10.1172/jci76610] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 12/11/2014] [Indexed: 11/17/2022] Open
Abstract
Patients with B cell precursor acute lymphoblastic leukemia (BPL) respond well to chemotherapy at initial diagnosis; however, therapeutic options are limited for individuals with BPL who relapse. Almost all BPL cells express CD19, and we recently cloned the gene encoding a natural ligand of the human CD19 receptor (CD19L). We hypothesized that fusion of CD19L to the soluble extracellular domain of proapoptotic TNF-related apoptosis-inducing ligand (sTRAIL) would markedly enhance the potency of sTRAIL and specifically induce BPL cell apoptosis due to membrane anchoring of sTRAIL and simultaneous activation of the CD19 and TRAIL receptor (TRAIL-R) apoptosis signaling pathways. Here, we demonstrate that recombinant human CD19L-sTRAIL was substantially more potent than sTRAIL and induced apoptosis in primary leukemia cells taken directly from BPL patients. CD19L-sTRAIL effectively targeted and eliminated in vivo clonogenic BPL xenograft cells, even at femtomolar-picomolar concentrations. In mice, CD19L-sTRAIL exhibited a more favorable pharmacokinetic (PK) profile than sTRAIL and was nontoxic at doses ranging from 32 fmol/kg to 3.2 pmol/kg. CD19L-sTRAIL showed potent in vivo antileukemic activity in NOD/SCID mouse xenograft models of relapsed and chemotherapy-resistant BPL at nontoxic fmol/kg dose levels. Together, these results suggest that recombinant human CD19L-sTRAIL has clinical potential as a biotherapeutic agent against BPL.
Collapse
|
134
|
Sun L, Ma JT, Zhang SL, Zou HW, Han CB. Efficacy and safety of chemotherapy or tyrosine kinase inhibitors combined with bevacizumab versus chemotherapy or tyrosine kinase inhibitors alone in the treatment of non-small cell lung cancer: a systematic review and meta-analysis. Med Oncol 2015; 32:473. [PMID: 25603953 DOI: 10.1007/s12032-014-0473-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 12/18/2014] [Indexed: 11/29/2022]
Abstract
The meta-analysis evaluated the efficacy and safety of chemotherapy or tyrosine kinase inhibitors combined with bevacizumab versus chemotherapy or tyrosine kinase inhibitors alone in the treatment of non-small cell lung cancer (NSCLC). The PubMed/MEDLINE, Ovid, Web of Science, CNKI, and the Cochrane Library database were searched for eligible randomized controlled trials comparing the combination of chemotherapy or epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) with bevacizumab to chemotherapy or EGFR-TKI alone. Main outcome measures were overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and adverse effects. The pooled data were analyzed by STATA 12.0 and expressed as hazard ratio (HR) or risk ratio (RR), with their corresponding 95 % confidence intervals (95 % CI). Nine eligible trials comprising 3,547 patients (1,779 for bevacizumab and 1,768 for controls) were included in the study. Chemotherapy or TKIs in combination with bevacizumab significantly prolonged PFS (HRpfs 0.72, 95 % CIpfs 0.66-0.79, P pfs < 0.001) and OS (HRos 0.90, 95 % CIos 0.82-0.99, P os = 0.029) as first-line treatment for NSCLC compared with chemotherapy or TKIs alone. Bevacizumab combination regimens significantly prolonged PFS (HR 0.62, 95 % CI 0.52-0.74, P < 0.001) as second-line treatment; however, no benefit regarding OS was observed with the addition of bevacizumab (HR 0.94, 95 % CI 0.78-1.12, P = 0.479). The bevacizumab group showed increased ORR in both first- and second-line treatments. The high-dose bevacizumab subgroup in combination with chemotherapy showed a statistically significant improvement in OS, PFS, and ORR (HRos 0.89, 95 % CIos 0.80-0.99, P os 0.037; HRpfs 0.71, 95 % CIpfs 0.64-0.79, P pfs < 0.01, RRorr 1.85, 95 % CIorr 1.59-2.15, P orr < 0.001, respectively); however, the low-dose bevacizumab subgroup did not show enhanced OS (HRos 0.91, 95 % CIos 0.77-1.07, P os = 0.263), and a moderate improvement of PFS and ORR (HRpfs 0.85, 95 % CIpfs 0.72-1.00, P pfs = 0.049; RRorr 1.60, 95 % CIorr 1.28-2.0, P orr < 0.001). Erlotinib in combination with bevacizumab significantly prolonged PFS (HR 0.60, P < 0.001, 95 % CI 0.51-0.71) and increased ORR (RR 1.21, 95 % CI 0.98-1.49, P = 0.067) compared with erlotinib alone. A higher incidence of grade ≥3 adverse events such as proteinuria, hypertension, and hemorrhage was observed in the bevacizumab combination group than in the control group without bevacizumab (P all < 0.05). The addition of bevacizumab to chemotherapy or erlotinib can significantly improve PFS and ORR both in first- and second-line treatments of advanced NSCLC, with an acceptable risk of bleeding events, hypertension, proteinuria, and rash. Combination therapy with bevacizumab and chemotherapy is beneficial regarding OS; however, whether bevacizumab plus erlotinib can prolong OS need further validation.
Collapse
Affiliation(s)
- Li Sun
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, People's Republic of China
| | | | | | | | | |
Collapse
|
135
|
Möller Y, Siegemund M, Beyes S, Herr R, Lecis D, Delia D, Kontermann R, Brummer T, Pfizenmaier K, Olayioye MA. EGFR-targeted TRAIL and a Smac mimetic synergize to overcome apoptosis resistance in KRAS mutant colorectal cancer cells. PLoS One 2014; 9:e107165. [PMID: 25198428 PMCID: PMC4157814 DOI: 10.1371/journal.pone.0107165] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 08/04/2014] [Indexed: 11/18/2022] Open
Abstract
TRAIL is a death receptor ligand that induces cell death preferentially in tumor cells. Recombinant soluble TRAIL, however, performs poorly as an anti-cancer therapeutic because oligomerization is required for potent biological activity. We previously generated a diabody format of tumor-targeted TRAIL termed DbαEGFR-scTRAIL, comprising single-stranded TRAIL molecules (scTRAIL) and the variable domains of a humanized variant of the EGFR blocking antibody Cetuximab. Here we define the bioactivity of DbαEGFR-scTRAIL with regard to both EGFR inhibition and TRAIL receptor activation in 3D cultures of Caco-2 colorectal cancer cells, which express wild-type K-Ras. Compared with conventional 2D cultures, Caco-2 cells displayed strongly enhanced sensitivity toward DbαEGFR-scTRAIL in these 3D cultures. We show that the antibody moiety of DbαEGFR-scTRAIL not only efficiently competed with ligand-induced EGFR function, but also determined the apoptotic response by specifically directing DbαEGFR-scTRAIL to EGFR-positive cells. To address how aberrantly activated K-Ras, which leads to Cetuximab resistance, affects DbαEGFR-scTRAIL sensitivity, we generated stable Caco-2tet cells inducibly expressing oncogenic K-RasG12V. In the presence of doxycycline, these cells showed increased resistance to DbαEGFR-scTRAIL, associated with the elevated expression of the anti-apoptotic proteins cIAP2, Bcl-xL and FlipS. Co-treatment of cells with the Smac mimetic SM83 restored the DbαEGFR-scTRAIL-induced apoptotic response. Importantly, this synergy between DbαEGFR-scTRAIL and SM83 also translated to 3D cultures of oncogenic K-Ras expressing HCT-116 and LoVo colorectal cancer cells. Our findings thus support the notion that DbαEGFR-scTRAIL therapy in combination with apoptosis-sensitizing agents may be promising for the treatment of EGFR-positive colorectal cancers, independently of their KRAS status.
Collapse
Affiliation(s)
- Yvonne Möller
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Martin Siegemund
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Sven Beyes
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Ricarda Herr
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Daniele Lecis
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Domenico Delia
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Roland Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Centre for Biological Signalling Studies BIOSS, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Monilola A. Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- * E-mail:
| |
Collapse
|
136
|
Graves JD, Kordich JJ, Huang TH, Piasecki J, Bush TL, Sullivan T, Foltz IN, Chang W, Douangpanya H, Dang T, O'Neill JW, Mallari R, Zhao X, Branstetter DG, Rossi JM, Long AM, Huang X, Holland PM. Apo2L/TRAIL and the death receptor 5 agonist antibody AMG 655 cooperate to promote receptor clustering and antitumor activity. Cancer Cell 2014; 26:177-89. [PMID: 25043603 DOI: 10.1016/j.ccr.2014.04.028] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 02/28/2014] [Accepted: 04/30/2014] [Indexed: 12/15/2022]
Abstract
Death receptor agonist therapies have exhibited limited clinical benefit to date. Investigations into why Apo2L/TRAIL and AMG 655 preclinical data were not predictive of clinical response revealed that coadministration of Apo2L/TRAIL with AMG 655 leads to increased antitumor activity in vitro and in vivo. The combination of Apo2L/TRAIL and AMG 655 results in enhanced signaling and can sensitize Apo2L/TRAIL-resistant cells. Structure determination of the Apo2L/TRAIL-DR5-AMG 655 ternary complex illustrates how higher order clustering of DR5 is achieved when both agents are combined. Enhanced agonism generated by combining Apo2L/TRAIL and AMG 655 provides insight into the limited efficacy observed in previous clinical trials and suggests testable hypotheses to reconsider death receptor agonism as a therapeutic strategy.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Cell Survival
- Crystallography, X-Ray
- Drug Resistance, Neoplasm
- Drug Synergism
- Humans
- Mice
- Models, Molecular
- Protein Multimerization
- Protein Structure, Quaternary
- Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors
- Receptors, TNF-Related Apoptosis-Inducing Ligand/chemistry
- Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
- Signal Transduction
- TNF-Related Apoptosis-Inducing Ligand/chemistry
- TNF-Related Apoptosis-Inducing Ligand/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
| | | | - Tzu-Hsuan Huang
- Therapeutic Innovation Unit, Amgen Inc., Cambridge, MA 02142, USA
| | - Julia Piasecki
- Therapeutic Innovation Unit, Amgen Inc., Seattle, WA 98119, USA
| | - Tammy L Bush
- Therapeutic Innovation Unit, Amgen Inc., Cambridge, MA 02142, USA
| | - Timothy Sullivan
- Therapeutic Innovation Unit, Amgen Inc., Cambridge, MA 02142, USA
| | - Ian N Foltz
- Department of Biologic Discovery, Amgen British Columbia, Burnaby, BC V5A 1V7, Canada
| | - Wesley Chang
- Department of Clinical Immunology, Amgen Inc., South San Francisco, CA 94080, USA
| | | | - Thu Dang
- Therapeutic Innovation Unit, Amgen Inc., Cambridge, MA 02142, USA
| | - Jason W O'Neill
- Department of Biologic Optimization, Amgen Inc., Seattle, WA 98119, USA
| | - Rommel Mallari
- Department of Molecular Structure and Characterization, Amgen, South San Francisco, CA, 94080, USA
| | - Xiaoning Zhao
- Department of Molecular Structure and Characterization, Amgen, South San Francisco, CA, 94080, USA
| | | | - John M Rossi
- Department of Molecular Sciences and Computational Biology, Amgen Inc., Thousand Oaks, CA 91320, USA
| | - Alexander M Long
- Department of Molecular Structure and Characterization, Amgen Inc., Cambridge, MA 02142, USA
| | - Xin Huang
- Department of Molecular Structure and Characterization, Amgen Inc., Cambridge, MA 02142, USA
| | - Pamela M Holland
- Therapeutic Innovation Unit, Amgen Inc., Cambridge, MA 02142, USA.
| |
Collapse
|
137
|
Wu G, Ji Z, Li H, Lei Y, Jin X, Yu Y, Sun M. Selective TRAIL-induced cytotoxicity to lung cancer cells mediated by miRNA response elements. Cell Biochem Funct 2014; 32:547-56. [PMID: 25132116 DOI: 10.1002/cbf.3042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 05/14/2014] [Accepted: 05/28/2014] [Indexed: 12/18/2022]
Abstract
Lung cancer is among the most common cancers, and the current therapeutic strategies are still inefficient in most cases. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising biological agent for cancer treatment because of its potent pro-apoptotic effect on cancer cells. However, TRAIL also induces apoptosis in normal cells and therefore may cause toxicity to normal tissues if clinically applied. To address this issue, we inserted microRNA response elements (MREs) of miR-133a, miR-137 and miR-449a, which are all underexpressed in lung cancer cells, into an adenoviral vector to regulate TRAIL expression. This MRE-regulated vector (Ad-TRAIL-MRE) was able to express TRAIL in a lung-cancer-specific fashion. No TRAIL expression was detected in normal cells. Consistently, Ad-TRAIL-MRE exerted cytotoxicity to lung cancer cells, rather than normal cells, perhaps via inducing selective apoptosis. The selective TRAIL-mediated growth-inhibiting effect was further confirmed in a tumour xenograft model. Also, Ad-TRAIL-MRE only resulted in very low hepatotoxicity when applied. Collectively, we generated a novel TRAIL-expressing adenoviral vector that was regulated by MREs. This strategy permits TRAIL expression in a lung-cancer-specific manner and is worth further studying for clinical trials.
Collapse
Affiliation(s)
- Guodong Wu
- Department of Cardiovascular, Regenerative Medicine and Tissue Engineering, First Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | | | | | | | | | | | | |
Collapse
|
138
|
Di X, Zhang G, Zhang Y, Takeda K, Rivera Rosado LA, Zhang B. Accumulation of autophagosomes in breast cancer cells induces TRAIL resistance through downregulation of surface expression of death receptors 4 and 5. Oncotarget 2014; 4:1349-64. [PMID: 23988408 PMCID: PMC3824535 DOI: 10.18632/oncotarget.1174] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) induces apoptosis through death receptors (DRs) 4 and/or 5 expressed on the surface of target cells. We have previously shown that deficiency of DR4 and DR5 on the surface membrane is a critical mechanism of cancer cell resistance to the recombinant human TRAIL and its receptor agonistic antibodies, which are being evaluated clinically for treating cancers. In certain cancer cells, DR4 and DR5 were found to be mislocalized in intracellular compartments yet to be characterized. Here, we report a novel role of autophagy in the regulation of dynamics of TRAIL death receptors. We first assessed basal levels of autophagosomes in a panel of 11 breast cancer cell lines using complementary approaches (LC3 immunoblotting, RFP-LC3 fluorescence microscopy, and electron microscopy). We found high levels of basal autophagosomes in TRAIL resistant breast cancer cell lines (e.g. BT474 and AU565) and relevant mouse xenograft models under nutrition-rich conditions. Notably, DR4 and DR5 co-localized with LC3-II in the autophagosomes of TRAIL-resistant cells. Disruption of basal autophagosomes successfully restored the surface expression of the death receptors which was accompanied by sensitization of TRAIL-resistant cells to TRAIL induced apoptosis. By contrast, TRAIL-sensitive cell lines (MDA-MB-231) are characterized by high levels of surface DR4/DR5 and an absence of basal autophagosomes. Inhibition of lysosomal activity induced an accumulation of autophagosomes and a decrease in surface DR4 and DR5, and the cells became less sensitive to TRAIL-induced apoptosis. These findings demonstrate a novel role for the basal autophagosomes in the regulation of TRAIL death receptors. Further studies are warranted to explore the possibility of using autophagosome markers such as LC3-II/LC3-I ratios for prediction of tumor resistance to TRAIL related therapies. The results also provide a rationale for future non-clinical and clinical studies testing TRAIL agonists in combination with agents that directly inhibit autophagosome assembly.
Collapse
Affiliation(s)
- Xu Di
- Division of Therapeutic Proteins, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, MD, United States
| | | | | | | | | | | |
Collapse
|
139
|
Synergistic Antitumor Activity of Recombinant Human Apo2L/Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) in Combination with Carboplatin and Pemetrexed in Malignant Pleural Mesothelioma. J Thorac Oncol 2014; 9:1008-1017. [DOI: 10.1097/jto.0000000000000198] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
140
|
Lemke J, von Karstedt S, Zinngrebe J, Walczak H. Getting TRAIL back on track for cancer therapy. Cell Death Differ 2014; 21:1350-64. [PMID: 24948009 PMCID: PMC4131183 DOI: 10.1038/cdd.2014.81] [Citation(s) in RCA: 369] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/14/2014] [Accepted: 04/15/2014] [Indexed: 02/07/2023] Open
Abstract
Unlike other members of the TNF superfamily, the TNF-related apoptosis-inducing ligand (TRAIL, also known as Apo2L) possesses the unique capacity to induce apoptosis selectively in cancer cells in vitro and in vivo. This exciting discovery provided the basis for the development of TRAIL-receptor agonists (TRAs), which have demonstrated robust anticancer activity in a number of preclinical studies. Subsequently initiated clinical trials testing TRAs demonstrated, on the one hand, broad tolerability but revealed, on the other, that therapeutic benefit was rather limited. Several factors that are likely to account for TRAs' sobering clinical performance have since been identified. First, because of initial concerns over potential hepatotoxicity, TRAs with relatively weak agonistic activity were selected to enter clinical trials. Second, although TRAIL can induce apoptosis in several cancer cell lines, it has now emerged that many others, and importantly, most primary cancer cells are resistant to TRAIL monotherapy. Third, so far patients enrolled in TRA-employing clinical trials were not selected for likelihood of benefitting from a TRA-comprising therapy on the basis of a valid(ated) biomarker. This review summarizes and discusses the results achieved so far in TRA-employing clinical trials in the light of these three shortcomings. By integrating recent insight on apoptotic and non-apoptotic TRAIL signaling in cancer cells, we propose approaches to introduce novel, revised TRAIL-based therapeutic concepts into the cancer clinic. These include (i) the use of recently developed highly active TRAs, (ii) the addition of efficient, but cancer-cell-selective TRAIL-sensitizing agents to overcome TRAIL resistance and (iii) employing proteomic profiling to uncover resistance mechanisms. We envisage that this shall enable the design of effective TRA-comprising therapeutic concepts for individual cancer patients in the future.
Collapse
Affiliation(s)
- J Lemke
- 1] Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK [2] Clinic of General and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - S von Karstedt
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - J Zinngrebe
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - H Walczak
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| |
Collapse
|
141
|
Jiang J, Liu X, Deng L, Zhang P, Wang G, Wang S, Liu H, Su Y. GMP production and characterization of leucine zipper-tagged tumor necrosis factor-related apoptosis-inducing ligand (LZ-TRAIL) for phase I clinical trial. Eur J Pharmacol 2014; 740:722-32. [PMID: 24929054 DOI: 10.1016/j.ejphar.2014.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 05/30/2014] [Accepted: 06/02/2014] [Indexed: 11/28/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) exhibits potent antitumor activity in a wide range of cancers without deleterious side effects on normal tissues. Several TRAIL derivatives have been developed to improve its pharmacokinetics and therapeutic effects through strategies such as adding a leucine zipper to increase the circulation half-life. To obtain clinical grade LZ-TRAIL for phase I clinical trial, a single batch of 30 L bioreactor culture was performed using the Escherichia coli BL21 (DE3) strain expressing the recombinant LZ-TRAIL. A robust LZ-TRAIL production fermentation process was developed, which could be scaled up from 5L to 50 L, and had a titer of approximately 1.4 g/l. A four-step purification strategy was carried out to obtain a final product with over 95% purity and 45% yield. The final material was filter sterilized, aseptically vialed, and stored at 4°C, and comprehensively characterized using multiple assays (vialed product was sterile, purity was 95%, aggregates were <5%, potency revealed IC50 of 9 nM on MDA-MB-231 cells, and the endotoxin level was <0.25 U/mg). The purity, composition, and functional activities of the molecule were confirmed. in vivo investigations indicated that LZ-TRAIL has better antitumor potency in three Xenograft tumor models compared to TRAIL (95-281). LZ-TRAIL also showed improved pharmacokinetic and safety profiles in cynomolgus monkeys without abnormalities associated with drug exposure. In conclusion, the scalable synthesis of LZ-TRAIL is useful for production of phase I clinical trial material. These preclinical investigations warrant further clinical development of this product for cancer therapy.
Collapse
Affiliation(s)
- Jing Jiang
- Department of Pharmacology, Binzhou Medical University, Shandong Province, Yantai 256603, China.
| | - Xiaobin Liu
- Shandong Simcere-Medgenn Biopharmaceutical Co., Ltd., Shandong Province, Yantai 264006, China
| | - Leixiu Deng
- Shandong Simcere-Medgenn Biopharmaceutical Co., Ltd., Shandong Province, Yantai 264006, China
| | - Peipei Zhang
- Shandong Simcere-Medgenn Biopharmaceutical Co., Ltd., Shandong Province, Yantai 264006, China
| | - Guangjun Wang
- Shandong Simcere-Medgenn Biopharmaceutical Co., Ltd., Shandong Province, Yantai 264006, China
| | - Shifu Wang
- Shandong Simcere-Medgenn Biopharmaceutical Co., Ltd., Shandong Province, Yantai 264006, China
| | - Honghao Liu
- Shandong Simcere-Medgenn Biopharmaceutical Co., Ltd., Shandong Province, Yantai 264006, China
| | - Yunpeng Su
- Simcere Pharmaceutical R&D Department; Jiangsu Simcere-Medgenn Biopharmaceutical Co., Ltd., Jiangsu Province, Nanjing 210000, China.
| |
Collapse
|
142
|
TRAIL-R2-specific antibodies and recombinant TRAIL can synergise to kill cancer cells. Oncogene 2014; 34:2138-2144. [PMID: 24909167 PMCID: PMC4240732 DOI: 10.1038/onc.2014.156] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 03/14/2014] [Accepted: 04/04/2014] [Indexed: 12/15/2022]
Abstract
TRAIL induces apoptosis in cancer cells whilst sparing normal tissues. Despite promising pre-clinical results, few patients responded to treatment with recombinant TRAIL (Apo2L/Dulanermin) or TRAIL-R2-specific antibodies, such as conatumumab (AMG655). It is unknown whether this was due to intrinsic TRAIL resistance within primary human cancers or insufficient agonistic activity of the TRAIL-R-targeting drugs. FcγR-mediated crosslinking increases the cancer-cell-killing activity of TRAIL-R2-specific antibodies in vivo. We tested this phenomenon using FcγR-expressing immune cells from patients with ovarian cancer. However, even in the presence of high numbers of FcγR-expressing immune cells, as found in ovarian cancer ascites, AMG655-induced apoptosis was not enabled to any significant degree, indicating that this concept may not translate into clinical use. On the basis of these results we next set out to determine whether AMG655 possibly interferes with apoptosis induction by endogenous TRAIL which could be expressed by immune cells. To do so, we tested how AMG655 affected apoptosis induction by recombinant TRAIL. This, however, resulted in the surprising discovery of a striking synergy between AMG655 and non-tagged TRAIL (Apo2L/TRAIL) in killing cancer cells. This combination was as effective in killing cancer cells as highly active recombinant isoleucine-zipper-tagged TRAIL (iz-TRAIL). The increased killing efficiency was due to enhanced formation of the TRAIL death-inducing signalling complex (DISC), enabled by concomitant binding of Apo2L/TRAIL and AMG655 to TRAIL-R2. The synergy of AMG655 with Apo2L/TRAIL extended to primary ovarian cancer cells and was further enhanced by combination with the proteasome inhibitor bortezomib or a SMAC mimetic. Importantly, primary human hepatocytes were not killed by the AMG655-Apo2L/TRAIL combination, also not when further combined with bortezomib or a SMAC mimetic. We therefore propose that clinical-grade non-tagged recombinant forms of TRAIL, such as dulanermin, could be combined with antibodies such as AMG655 to introduce a highly active TRAIL-R2-agonistic therapy into the cancer clinic.
Collapse
|
143
|
Cancer therapeutics: Targeting the apoptotic pathway. Crit Rev Oncol Hematol 2014; 90:200-19. [DOI: 10.1016/j.critrevonc.2013.12.012] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 12/05/2013] [Accepted: 12/12/2013] [Indexed: 01/20/2023] Open
|
144
|
Lu M, Marsters S, Ye X, Luis E, Gonzalez L, Ashkenazi A. E-cadherin couples death receptors to the cytoskeleton to regulate apoptosis. Mol Cell 2014; 54:987-998. [PMID: 24882208 DOI: 10.1016/j.molcel.2014.04.029] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 02/11/2014] [Accepted: 04/22/2014] [Indexed: 01/14/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is a cellular process essential to the development and maintenance of solid tissues. In cancer, EMT suppresses apoptosis, but the mechanisms remain unclear. EMT selectively attenuated apoptosis signaling via the death receptors DR4 and DR5. Loss of the epithelial cell adhesion protein E-cadherin recapitulated this outcome, whereas homotypic E-cadherin engagement promoted apoptotic signaling via DR4/DR5, but not Fas. Depletion of α-catenin, which couples E-cadherin to the actin cytoskeleton, or actin polymerization inhibitors similarly attenuated DR4/DR5-induced apoptosis. E-cadherin bound specifically to ligated DR4/DR5, requiring extracellular cadherin domain 1 and calcium. E-cadherin augmented DR4/DR5 clustering and assembly of the death-inducing signaling complex (DISC), increasing caspase-8 activation in high molecular weight cell fractions. Conversely, EMT attenuated DR4/DR5-mediated DISC formation and caspase-8 stimulation. Consistent with these findings, epithelial cancer cell lines expressing higher E-cadherin levels displayed greater sensitivity to DR4/DR5-mediated apoptosis. These results have potential implications for tissue homeostasis as well as cancer therapy.
Collapse
Affiliation(s)
- Min Lu
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Scot Marsters
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Xiaofen Ye
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Elizabeth Luis
- Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Lino Gonzalez
- Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Avi Ashkenazi
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
145
|
Seifert O, Pollak N, Nusser A, Steiniger F, Rüger R, Pfizenmaier K, Kontermann RE. Immuno-LipoTRAIL: Targeted delivery of TRAIL-functionalized liposomal nanoparticles. Bioconjug Chem 2014; 25:879-87. [PMID: 24766622 DOI: 10.1021/bc400517j] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The TNF-related apoptosis-inducing ligand (TRAIL) is a powerful inducer of apoptosis in tumor cells; however, clinical studies with recombinant soluble TRAIL were rather disappointing. Here, we developed TRAIL-functionalized liposomes (LipoTRAIL, LT) to mimic membrane-displayed TRAIL for efficient activation of death receptors DR4 and DR5 and enhanced induction of apoptosis, which were combined with an anti-EGFR single-chain Fv fragment (scFv) for targeted delivery to EGFR-positive tumor cells. These immuno-LipoTRAILs (ILTs) bound specifically to EGFR-expressing cells (Colo205) and exhibited increased cytotoxicity compared with that of nontargeted LTs. Compared to that of the soluble TRAIL, the plasma half-life of the functionalized liposomes was strongly extended, and increased antitumor activity of LT and ILT was demonstrated in a xenograft tumor model. Thus, we established a multifunctional liposomal TRAIL formulation (ILT) with improved pharmacokinetic and pharmacodynamic behavior, characterized by targeted delivery and increased induction of apoptosis due to multivalent TRAIL presentation.
Collapse
Affiliation(s)
- Oliver Seifert
- Institut für Zellbiologie und Immunologie, Universität Stuttgart , Allmandring 31, 70569 Stuttgart, Germany
| | | | | | | | | | | | | |
Collapse
|
146
|
The novel thymidylate synthase inhibitor trifluorothymidine (TFT) and TRAIL synergistically eradicate non-small cell lung cancer cells. Cancer Chemother Pharmacol 2014; 73:1273-83. [PMID: 24744163 DOI: 10.1007/s00280-014-2465-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/02/2014] [Indexed: 12/18/2022]
Abstract
PURPOSE TRAIL, a tumor selective anticancer agent, may be used for the treatment of non-small cell lung cancer (NSCLC). However, TRAIL resistance is frequently encountered. Here, the combined use of TRAIL with trifluorothymidine (TFT), a thymidylate synthase inhibitor, was examined for sensitizing NSCLC cells to TRAIL. METHODS Interactions between TRAIL and TFT were studied in NSCLC cells using growth inhibition and apoptosis assays. Western blotting and flow cytometry were used to investigate underlying mechanisms. RESULTS The combined treatment of TFT and TRAIL showed synergistic cytotoxicity in A549, H292, H322 and H460 cells. For synergistic activity, the sequence of administration was important; TFT treatment followed by TRAIL exposure did not show sensitization. Combined TFT and TRAIL treatment for 24 h followed by 48 h of TFT alone was synergistic in all cell lines, with combination index values below 0.9. The treatments affected cell cycle progression, with TRAIL inducing a G1 arrest and TFT, a G2/M arrest. TFT activated Chk2 and reduced Cdc25c levels known to cause G2/M arrest. TRAIL-induced caspase-dependent apoptosis was enhanced by TFT, whereas TFT alone mainly induced caspase-independent death. TFT increased the expression of p53 and p21/WAF1, and p53 was involved in the increase of TRAIL-R2 surface expression. TFT also caused downregulation of cFLIP and XIAP and increased Bax expression. CONCLUSIONS TFT enhances TRAIL-induced apoptosis in NSCLC cells by sensitizing the apoptotic machinery at different levels in the TRAIL pathway. Our findings suggest a possible therapeutic benefit of the combined use of TFT and TRAIL in NSCLC.
Collapse
|
147
|
Garimella SV, Gehlhaus K, Dine JL, Pitt JJ, Grandin M, Chakka S, Nau MM, Caplen NJ, Lipkowitz S. Identification of novel molecular regulators of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in breast cancer cells by RNAi screening. Breast Cancer Res 2014; 16:R41. [PMID: 24745479 PMCID: PMC4053258 DOI: 10.1186/bcr3645] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/02/2014] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) binds to its receptors, TRAIL-receptor 1 (TRAIL-R1) and TRAIL-receptor 2 (TRAIL-R2), leading to apoptosis by activation of caspase-8 and the downstream executioner caspases, caspase-3 and caspase-7 (caspase-3/7). Triple-negative breast cancer (TNBC) cell lines with a mesenchymal phenotype are sensitive to TRAIL, whereas other breast cancer cell lines are resistant. The underlying mechanisms that control TRAIL sensitivity in breast cancer cells are not well understood. Here, we performed small interfering RNA (siRNA) screens to identify molecular regulators of the TRAIL pathway in breast cancer cells. METHODS We conducted siRNA screens of the human kinome (691 genes), phosphatome (320 genes), and about 300 additional genes in the mesenchymal TNBC cell line MB231. Forty-eight hours after transfection of siRNA, parallel screens measuring caspase-8 activity, caspase-3/7 activity, or cell viability were conducted in the absence or presence of TRAIL for each siRNA, relative to a negative control siRNA (siNeg). A subset of genes was screened in cell lines representing epithelial TNBC (MB468), HER2-amplified breast cancer (SKBR3), and estrogen receptor-positive breast cancer (T47D). Selected putative negative regulators of the TRAIL pathway were studied by using small-molecule inhibitors. RESULTS The primary screens in MB231 identified 150 genes, including 83 kinases, 4 phosphatases, and 63 nonkinases, as potential negative regulators of TRAIL. The identified genes are involved in many critical cell processes, including apoptosis, growth factor-receptor signaling, cell-cycle regulation, transcriptional regulation, and DNA repair. Gene-network analysis identified four genes (PDPK1, IKBKB, SRC, and BCL2L1) that formed key nodes within the interaction network of negative regulators. A secondary screen of a subset of the genes identified in additional cell lines representing different breast cancer subtypes and sensitivities to TRAIL validated and extended these findings. Further, we confirmed that small-molecule inhibition of SRC or BCL2L1, in combination with TRAIL, sensitizes breast cancer cells to TRAIL-induced apoptosis, including cell lines resistant to TRAIL-induced cytotoxicity. CONCLUSIONS These data identify novel molecular regulators of TRAIL-induced apoptosis in breast cancer cells and suggest strategies for the enhanced application of TRAIL as a therapy for breast cancer.
Collapse
|
148
|
Jin SM, Jang HW, Sohn SY, Kim NK, Joung JY, Cho YY, Kim SW, Chung JH. Role of autophagy in the resistance to tumour necrosis factor-related apoptosis-inducing ligand-induced apoptosis in papillary and anaplastic thyroid cancer cells. Endocrine 2014; 45:256-62. [PMID: 23821365 DOI: 10.1007/s12020-013-9997-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 06/03/2013] [Indexed: 10/26/2022]
Abstract
Current alternative therapies for refractory thyroid cancer such as kinase inhibitors have limitations including incomplete response and toxicity. Although tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) can induce cancer cell-specific apoptosis, various degrees of TRAIL resistance have been reported for different types of thyroid cancer cells. Here, we investigated if modulation of autophagy could improve sensitivity to TRAIL in papillary and anaplastic thyroid cancer cells. Human papillary thyroid cancer cells (TPC-1 cells) and human anaplastic thyroid cancer cells (FRO cells) were treated with TRAIL after transfection with ATG7 siRNA or control siRNA. Levels of autophagy and apoptosis were confirmed by Western blot of ATG7, LC3, caspase-3 and poly (ADP-ribose) polymerase. Viability index was determined by dimethyl-thiazole-diphenyltetrazolium bromide assay. Fraction of apoptotic cells was determined by flow cytometry. In TPC-1 cells, treatment with TRAIL increased the levels of autophagy. A low concentration (20 ng/ml) of TRAIL resulted in significantly decreased viability index and increased apoptosis. However, inhibition of autophagy with ATG7 siRNA desensitised the cells to TRAIL-induced apoptosis. In FRO cells, TRAIL did not increase the levels of autophagy. In contrast to TPC-1 cells, inhibition of autophagy with ATG7 siRNA sensitised FRO cells to TRAIL-induced apoptosis. Autophagy might contribute to the known sensitivity of papillary thyroid cancer cells to TRAIL-induced apoptosis. Inhibition of autophagy in anaplastic thyroid cancer cells could sensitise these cells to TRAIL-induced apoptosis.
Collapse
Affiliation(s)
- Sang-Man Jin
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50, Ilwon-dong, Gangnam-gu, Seoul, 135-710, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
149
|
Sun S, Li Z, Sun L, Yang C, Mei Z, Ouyang W, Yang B, Xie C. Results on efficacy and safety of cancer treatment with or without tumor necrosis factor-related apoptosis-inducing ligand-related agents: A meta-analysis. Mol Clin Oncol 2014; 2:440-448. [PMID: 24772315 DOI: 10.3892/mco.2014.261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/21/2014] [Indexed: 11/05/2022] Open
Abstract
This meta-analysis aimed to evaluate the currently available evidence on the efficacy and safety of cancer treatment with or without tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-related agents. We conducted a systematic search through Medline, Cochrane Library and EMBASE electronic databases and manually searched the Journal of Clinical Oncology to identify randomized controlled trials (RCTs) conducted between 1995 and 2013 comparing the efficacy and safety results of cancer treatment with and without TRAIL-related agents. The methodological quality of the included RCTs was assessed by the Cochrane Risk of Bias assessment tool. The outcome measurements included objective response rate (ORR), clinical benefit rate (CBR)/disease control rate (DCR) and adverse events (AEs). The relevant data were analyzed using Review Manager 5.2 software. Grading of Recommendations Assessment Development and Evaluation was used to assess the quality of evidence and grade of recommendation. Four RCTs, including a total of 596 patients, were ultimately selected and analyzed. There were no statistically significant differences among the 4 RCTs regarding ORR [relative risk (RR)=0.92, 95% confidence interval (CI): 0.73-1.15, P=0.45], CBR/DCR (RR=0.92, 95% CI: 0.81-1.05, P=0.21), progression-free survival [hazard ratio (HR)=0.89, 95% CI: 0.75-1.05, P=0.16], overall survival (HR=0.90, 95% CI: 0.74-1.09, P=0.27), number of patients with any AEs (RR=0.99, 95% CI: 0.96-1.03, P=0.77), number of patients with any severe AEs (RR=0.95, 95% CI: 0.78-1.55, P=0.58), number of patients with ≥grade 3 AEs (RR=1.13, 95% CI: 0.93-1.38, P=0.22) and number of fatal AEs (RR=1.14, 95% CI: 0.71-1.81, P=0.59). The quality of evidence was considered to be moderate and the recommendation grades were weak. In conclusion, the benefits of TRAIL-related agents in the treatment of cancer patients remain uncertain and further clinical trials are required.
Collapse
Affiliation(s)
- Shaoxing Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zonghuan Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Li Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Chunxu Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zijie Mei
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wen Ouyang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Bo Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China ; ; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
150
|
Abstract
Apoptosis is a cell death program that is well-orchestrated for normal tissue homeostasis and for removal of damaged, old or infected cells. It is regulated by intrinsic and extrinsic pathways. The intrinsic pathway responds to signals such as ultraviolet radiation or DNA damage and activates "executioner" caspases through a mitochondria-dependent pathway. The extrinsic pathway is activated by death signals induced, for example, by an infection that activates the immune system or receptor-mediated pathways. The extrinsic pathway signals also cascade down to executioner caspases that cleave target proteins and lead to cell death. Strict control of cellular apoptosis is important for the hematopoietic system as it has a high turnover rate. However, the apoptosis program is often deregulated in hematologic malignancies leading to the accumulation of malignant cells. Therefore, apoptosis pathways have been identified for the development of anticancer therapeutics. We review here the proteins that have been targeted for anticancer drug development in hematologic malignancies. These include BCL-2 family proteins, death ligands and receptors, inhibitor of apoptosis family proteins and caspases. Except for caspase activators, drugs that target each of these classes of proteins have advanced into clinical trials.
Collapse
Affiliation(s)
- Shadia Zaman
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center , Houston, TX , USA
| | | | | |
Collapse
|