1
|
Chen Q, Zhang H. SMAC mimetic BV6 acts in synergy with mTOR inhibitor to increase cisplatin sensitivity in ovarian cancer. Anticancer Drugs 2025; 36:62-71. [PMID: 39423314 DOI: 10.1097/cad.0000000000001664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
The objective of this study is to observe the antitumor efficacy of the second mitochondria-derived activator of caspases (SMAC) mimetic bivalent smac mimetic (BV6) in combination with target of rapamycin (mTOR) inhibitor on DDP (cisplatin) sensitivity. Ovarian cancer cells were exposed to cisplatin, BV6, DDP + BV6, and DDP + BV6 + mTOR inhibitor Rapamycin. Using proteomics and bioinformatics, protein expression profiles in ovarian cancer were determined. Bagg Albino color nude mice were treated with DDP or BV6 alone or in combination, or BV6 + DDP + Rapamycin. The effects of different treatments on ovarian cancer cells and tumor growth were evaluated in vivo and in vitro . Proteomics and bioinformatics analysis revealed significant changes of protein kinase (AKT)/mTOR pathway. Consistently, western blot data indicated that AKT/mTOR axis was gradually activated in BV6-treated ovarian cancer cells and attenuated the cytotoxic effect of BV6. Functional assays showed that DDP or BV6 treatment alone significantly enhanced the sensitivity and inhibited the migration of ovarian cancer cells, but without any synergistic effects. In addition, combination with BV6 and mTOR inhibitor Rapamycin significantly decreased cell viability and inhibited migration of ovarian cancer cells exposed to DDP. Consistently, the xenograft model showed that co-treatment with Rapamycin with BV6 had significantly suppressed tumor growth and metastasis. Our study demonstrated that SMAC analogue BV6 exhibits a strong anticancer effect on ovarian cancer in vitro and in vivo . Combination with Rapamycin overcomes the activation of mTOR pathway by BV6 and increases the chemosensitivity to DDP. These data suggest a potential application of triple combination with DDP + BV6 + Rapamycin in clinical management of ovarian cancer.
Collapse
Affiliation(s)
- Qi Chen
- Department of Gynecological Oncology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | | |
Collapse
|
2
|
Kim HS, Bae S, Lim YJ, So KA, Kim TJ, Bae S, Lee JH. Tephrosin Suppresses the Chemoresistance of Paclitaxel-Resistant Ovarian Cancer via Inhibition of FGFR1 Signaling Pathway. Biomedicines 2023; 11:3155. [PMID: 38137377 PMCID: PMC10740824 DOI: 10.3390/biomedicines11123155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/21/2023] [Accepted: 11/26/2023] [Indexed: 12/24/2023] Open
Abstract
Ovarian cancer is the leading cause of death among gynecologic cancers. Paclitaxel is used as a standard first-line therapeutic agent for ovarian cancer. However, chemotherapeutic resistance and high recurrence rates are major obstacles to treating ovarian cancer. We have found that tephrosin, a natural rotenoid isoflavonoid, can resensitize paclitaxel-resistant ovarian cancer cells to paclitaxel. Cell viability, immunoblotting, and a flow cytometric analysis showed that a combination treatment made up of paclitaxel and tephrosin induced apoptotic death. Tephrosin inhibited the phosphorylation of AKT, STAT3, ERK, and p38 MAPK, all of which simultaneously play important roles in survival signaling pathways. Notably, tephrosin downregulated the phosphorylation of FGFR1 and its specific adapter protein FRS2, but it had no effect on the phosphorylation of the EGFR. Immunoblotting and a fluo-3 acetoxymethyl assay showed that tephrosin did not affect the expression or function of P-glycoprotein. Additionally, treatment with N-acetylcysteine did not restore cell cytotoxicity caused by a treatment combination made up of paclitaxel and tephrosin, showing that tephrosin did not affect the reactive oxygen species scavenging pathway. Interestingly, tephrosin reduced the expression of the anti-apoptotic factor XIAP. This study demonstrates that tephrosin is a potent antitumor agent that can be used in the treatment of paclitaxel-resistant ovarian cancer via the inhibition of the FGFR1 signaling pathway.
Collapse
Affiliation(s)
- Hee Su Kim
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.S.K.); (S.B.); (Y.J.L.); (S.B.)
| | - Sowon Bae
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.S.K.); (S.B.); (Y.J.L.); (S.B.)
| | - Ye Jin Lim
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.S.K.); (S.B.); (Y.J.L.); (S.B.)
| | - Kyeong A So
- Department of Obstetrics and Gynecology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea; (K.A.S.); (T.J.K.)
| | - Tae Jin Kim
- Department of Obstetrics and Gynecology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea; (K.A.S.); (T.J.K.)
| | - Seunghee Bae
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.S.K.); (S.B.); (Y.J.L.); (S.B.)
| | - Jae Ho Lee
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.S.K.); (S.B.); (Y.J.L.); (S.B.)
| |
Collapse
|
3
|
Tavakoli S, Firoozpour L, Davoodi J. The synergistic effect of chimeras consisting of N-terminal smac and modified KLA peptides in inducing apoptosis in breast cancer cell lines. Biochem Biophys Res Commun 2023; 655:138-144. [PMID: 36934589 DOI: 10.1016/j.bbrc.2023.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/26/2023] [Accepted: 03/05/2023] [Indexed: 03/08/2023]
Abstract
Drug resistance is one of the most important obstacles in effective cancer therapy triggered through various mechanisms. One of these mechanisms is caused by the upregulation of Inhibitor of Apoptosis Proteins (IAPs). IAPs, inhibit apoptosis through direct and/or indirect caspase inhibition, which themselves are antagonized by an endogenous protein called Second Mitochondrial-derived Activator of Caspases, Smac/Diablo, mediated by the presence of a tetrapeptide IAP binding motif at its N-terminus. Accordingly, Smac-based peptides are under intense investigation as anti-cancer drugs and have reached Phase 2 clinical trials, although, Smac based peptides or mimetics alone have not been effective as anti-cancer agents. On the other hand, KLA peptide has shown major toxicity against cancer cells through the induction of apoptosis. Consequently, we designed an anti-cancer chimera by fusing an octa-peptide from the N-terminus of mature Smac protein to a modified proapoptotic KLA peptide (KLAKLCKKLAKLCK) to be called Smac-KLA. This chimera, therefore, possesses both proapoptotic and anti-IAP activities. In addition, we dimerized this chimera via intermolecular disulfide bonds in order to enhance their cellular permeability. Both the Smac-KLA monomeric and dimeric peptides exhibited cytotoxic activity against both MCF-7 and MDA-MB231 breast cancer cell lines at low micromolar concentrations. Importantly, the dimerization of the chimeras enhanced their potency 2-4- fold due to higher cellular uptake.
Collapse
Affiliation(s)
- Somayeh Tavakoli
- Institute of Biochemistry and Biophysics, University of Tehran, Postal code: 1417614335, Tehran, Iran
| | - Loghman Firoozpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamshid Davoodi
- Institute of Biochemistry and Biophysics, University of Tehran, Postal code: 1417614335, Tehran, Iran.
| |
Collapse
|
4
|
Guo L, Wang J, Li N, Cui J, Su Y. Peptides for diagnosis and treatment of ovarian cancer. Front Oncol 2023; 13:1135523. [PMID: 37213272 PMCID: PMC10196167 DOI: 10.3389/fonc.2023.1135523] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 04/24/2023] [Indexed: 05/23/2023] Open
Abstract
Ovarian cancer is the most deadly gynecologic malignancy, and its incidence is gradually increasing. Despite improvements after treatment, the results are unsatisfactory and survival rates are relatively low. Therefore, early diagnosis and effective treatment remain two major challenges. Peptides have received significant attention in the search for new diagnostic and therapeutic approaches. Radiolabeled peptides specifically bind to cancer cell surface receptors for diagnostic purposes, while differential peptides in bodily fluids can also be used as new diagnostic markers. In terms of treatment, peptides can exert cytotoxic effects directly or act as ligands for targeted drug delivery. Peptide-based vaccines are an effective approach for tumor immunotherapy and have achieved clinical benefit. In addition, several advantages of peptides, such as specific targeting, low immunogenicity, ease of synthesis and high biosafety, make peptides attractive alternative tools for the diagnosis and treatment of cancer, particularly ovarian cancer. In this review, we focus on the recent research progress regarding peptides in the diagnosis and treatment of ovarian cancer, and their potential applications in the clinical setting.
Collapse
|
5
|
Zaman R, Islam RA, Chowdhury EH. Evolving therapeutic proteins to precisely kill cancer cells. J Control Release 2022; 351:779-804. [DOI: 10.1016/j.jconrel.2022.09.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 10/31/2022]
|
6
|
Priwitaningrum DL, Jentsch J, Bansal R, Rahimian S, Storm G, Hennink WE, Prakash J. Apoptosis-inducing peptide loaded in PLGA nanoparticles induces anti-tumor effects in vivo. Int J Pharm 2020; 585:119535. [PMID: 32534162 DOI: 10.1016/j.ijpharm.2020.119535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 01/17/2023]
Abstract
Induction of apoptosis in tumor cells specifically within the complex tumor microenvironment is highly desirable to kill them efficiently and to enhance the effects of chemotherapy. Second mitochondria-derived activator of caspase (Smac) is a key pro-apoptotic pathway which can be activated with a Smac mimetic peptide. However, in vivo application of peptides is hampered by several limitations such as poor pharmacokinetics, rapid elimination, enzymatic degradation, and insufficient intracellular delivery. In this study, we developed a nanosystem to deliver a Smac peptide to tumor by passive targeting. We first synthesized a chimeric peptide that consists of the 8-mer Smac peptide and a 14-mer cell penetrating peptide (CPP) and then encapsulated the Smac-CPP into polymeric nanoparticles (Smac-CPP-NPs). In vitro, Smac-CPP-NPs were rapidly internalized by 4T1 mammary tumor cells and subsequently released Smac-CPP into the cells, as shown with fluorescence microscopy. Furthermore, Smac-CPP-NPs induced apoptosis in tumor cells, as confirmed with cell viability and caspase 3/7 assays. Interestingly, combination of Smac-CPP-NPs with doxorubicin (dox), a clinically used cytostatic drug, showed combined effects in vitro in 4T1 cells. The effect was significantly better than that of SMAC-CPP-NPs alone as well as empty nanoparticles and dox. In vivo, co-treatment with Smac-CPP-NPs and free dox reduced the tumor growth to 85%. Furthermore, the combination of Smac-CPP-NPs and free dox showed reduced proliferating tumor cells (Ki-67 staining) and increased apoptotic cells (cleaved caspase-3 staining) in tumors. In conclusion, the present study demonstrates that the intracellular delivery of Smac-mimetic peptide using nanoparticle system can be an interesting strategy to attenuate the tumor growth and to potentiate the therapeutic efficacy of chemotherapy in vivo.
Collapse
Affiliation(s)
- Dwi L Priwitaningrum
- Targeted Therapeutics and Nanomedicine, Department of Biomaterials Science and Technology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Faculty of Pharmacy, University of Sumatera Utara, Medan, Indonesia
| | - Julian Jentsch
- Targeted Therapeutics and Nanomedicine, Department of Biomaterials Science and Technology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- Targeted Therapeutics and Nanomedicine, Department of Biomaterials Science and Technology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Sima Rahimian
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Faculty of Pharmacy, University of Sumatera Utara, Medan, Indonesia
| | - Jai Prakash
- Targeted Therapeutics and Nanomedicine, Department of Biomaterials Science and Technology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
7
|
Bae Y, Lee YH, Ko KS, Han J, Choi JS. Smac Gene Delivery by the Glycol Chitosan with Low Molecular Weight Polyethylenimine Induces Apoptosis of Cancer Cells for Combination Therapy with Etoposide. Macromol Res 2019. [DOI: 10.1007/s13233-019-7130-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
8
|
Di Tucci C, Capone C, Galati G, Iacobelli V, Schiavi MC, Di Donato V, Muzii L, Panici PB. Immunotherapy in endometrial cancer: new scenarios on the horizon. J Gynecol Oncol 2019; 30:e46. [PMID: 30887763 PMCID: PMC6424849 DOI: 10.3802/jgo.2019.30.e46] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 12/18/2018] [Indexed: 02/08/2023] Open
Abstract
This extensive review summarizes clinical evidence on immunotherapy and targeted therapy currently available for endometrial cancer (EC) and reports the results of the clinical trials and ongoing studies. The research was carried out collecting preclinical and clinical findings using keywords such as immune environment, tumor infiltrating lymphocytes, programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) expression, immune checkpoint inhibitors, anti-PD-1/PD-L1 antibodies and others' on PubMed. Finally, we looked for the ongoing immunotherapy trials on ClinicalTrials.gov. EC is the fourth most common malignancy in women in developed countries. Despite medical and surgical treatments, survival has not improved in the last decade and death rates have increased for uterine cancer in women. Therefore, identification of clinically significant prognostic risk factors and formulation of new rational therapeutic regimens have great significance for enhancing the survival rate and improving the outcome in patients with advanced or metastatic disease. The identification of genetic alterations, including somatic mutations and microsatellite instability, and the definition of intracellular signaling pathways alterations that have a major role in in tumorigenesis is leading to the development of new therapeutic options for immunotherapy and targeted therapy.
Collapse
Affiliation(s)
- Chiara Di Tucci
- Department of Gynecological and Obstetric Sciences, and Urological Sciences, University of Rome "Sapienza", Umberto I Hospital, Rome, Italy.
| | - Carmela Capone
- Department of Gynecological and Obstetric Sciences, and Urological Sciences, University of Rome "Sapienza", Umberto I Hospital, Rome, Italy
| | - Giulia Galati
- Department of Gynecological and Obstetric Sciences, and Urological Sciences, University of Rome "Sapienza", Umberto I Hospital, Rome, Italy
| | - Valentina Iacobelli
- Department of Gynecological and Obstetric Sciences, and Urological Sciences, University of Rome "Sapienza", Umberto I Hospital, Rome, Italy
| | - Michele C Schiavi
- Department of Gynecological and Obstetric Sciences, and Urological Sciences, University of Rome "Sapienza", Umberto I Hospital, Rome, Italy
| | - Violante Di Donato
- Department of Gynecological and Obstetric Sciences, and Urological Sciences, University of Rome "Sapienza", Umberto I Hospital, Rome, Italy
| | - Ludovico Muzii
- Department of Gynecological and Obstetric Sciences, and Urological Sciences, University of Rome "Sapienza", Umberto I Hospital, Rome, Italy
| | - Pierluigi Benedetti Panici
- Department of Gynecological and Obstetric Sciences, and Urological Sciences, University of Rome "Sapienza", Umberto I Hospital, Rome, Italy
| |
Collapse
|
9
|
Nuclear Imaging Study of the Pharmacodynamic Effects of Debio 1143, an Antagonist of Multiple Inhibitor of Apoptosis Proteins (IAPs), in a Triple-Negative Breast Cancer Model. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2018:8494031. [PMID: 30627061 PMCID: PMC6305031 DOI: 10.1155/2018/8494031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/18/2018] [Indexed: 11/17/2022]
Abstract
Background Debio 1143, a potent orally available SMAC mimetic, targets inhibitors of apoptosis proteins (IAPs) members and is currently in clinical trials. In this study, nuclear imaging evaluated the effects of Debio 1143 on tumor cell death and metabolism in a triple-negative breast cancer (TNBC) cell line (MDA-MB-231)-based animal model. Methods Apoptosis induced by Debio 1143 was assessed by FACS (caspase-3, annexin 5 (A5)), binding of 99mTc-HYNIC-Annexin V, and a cell proliferation assay. 99mTc-HYNIC-Annexin V SPECT and [18F]-FDG PET were also performed in mice xenografted with MDA-MB-231 cells. Results Debio 1143 induced early apoptosis both in vitro and in vivo 6 h after treatment. Debio 1143 inhibited tumor growth, which was associated with a decreased tumor [18F]-FDG uptake when measured during treatment. Conclusions This imaging study combining SPECT and PET showed the early proapoptotic effects of Debio 1143 resulting in a robust antitumor activity in a preclinical TNBC model. These imaging biomarkers represent valuable noninvasive tools for translational and clinical research in TNBC.
Collapse
|
10
|
Panayotopoulou EG, Müller AK, Börries M, Busch H, Hu G, Lev S. Targeting of apoptotic pathways by SMAC or BH3 mimetics distinctly sensitizes paclitaxel-resistant triple negative breast cancer cells. Oncotarget 2018; 8:45088-45104. [PMID: 28187446 PMCID: PMC5542169 DOI: 10.18632/oncotarget.15125] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/24/2017] [Indexed: 12/13/2022] Open
Abstract
Standard chemotherapy is the only systemic treatment for triple-negative breast cancer (TNBC), and despite the good initial response, resistance remains a major therapeutic obstacle. Here, we employed a High-Throughput Screen to identify targeted therapies that overcome chemoresistance in TNBC. We applied short-term paclitaxel treatment and screened 320 small-molecule inhibitors of known targets to identify drugs that preferentially and efficiently target paclitaxel-treated TNBC cells. Among these compounds the SMAC mimetics (BV6, Birinapant) and BH3-mimetics (ABT-737/263) were recognized as potent targeted therapy for multiple paclitaxel-residual TNBC cell lines. However, acquired paclitaxel resistance through repeated paclitaxel pulses result in desensitization to BV6, but not to ABT-263, suggesting that short- and long-term paclitaxel resistance are mediated by distinct mechanisms. Gene expression profiling of paclitaxel-residual, -resistant and naïve MDA-MB-231 cells demonstrated that paclitaxel-residual, as opposed to -resistant cells, were characterized by an apoptotic signature, with downregulation of anti-apoptotic genes (BCL2, BIRC5), induction of apoptosis inducers (IL24, PDCD4), and enrichment of TNFα/NF-κB pathway, including upregulation of TNFSF15, coupled with cell-cycle arrest. BIRC5 and FOXM1 downregulation and IL24 induction was also evident in breast cancer patient datasets following taxane treatment. Exposure of naïve or paclitaxel-resistant cells to supernatants of paclitaxel-residual cells sensitized them to BV6, and treatment with TNFα enhanced BV6 potency, suggesting that sensitization to BV6 is mediated, at least partially, by secreted factor(s). Our results suggest that administration of SMAC or BH3 mimetics following short-term paclitaxel treatment could be an effective therapeutic strategy for TNBC, while only BH3-mimetics could effectively overcome long-term paclitaxel resistance.
Collapse
Affiliation(s)
| | - Anna-Katharina Müller
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Melanie Börries
- Institute of Molecular Medicine and Cell Research (IMMZ), Albert Ludwigs-University, 79104 Freiburg, Germany
| | - Hauke Busch
- Institute of Molecular Medicine and Cell Research (IMMZ), Albert Ludwigs-University, 79104 Freiburg, Germany
| | - Guohong Hu
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Sima Lev
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
11
|
Zhang M, Liu E, Cui Y, Huang Y. Nanotechnology-based combination therapy for overcoming multidrug-resistant cancer. Cancer Biol Med 2017; 14:212-227. [PMID: 28884039 PMCID: PMC5570599 DOI: 10.20892/j.issn.2095-3941.2017.0054] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/03/2017] [Indexed: 12/28/2022] Open
Abstract
Multidrug resistance (MDR) is a major obstacle to successful cancer treatment and is crucial to cancer metastasis and relapse. Combination therapy is an effective strategy for overcoming MDR. However, the different pharmacokinetic (PK) profiles of combined drugs often undermine the combination effect in vivo, especially when greatly different physicochemical properties (e.g., those of macromolecules and small drugs) combine. To address this issue, nanotechnology-based codelivery techniques have been actively explored. They possess great advantages for tumor targeting, controlled drug release, and identical drug PK profiles. Thus, a powerful tool for combination therapy is provided, and the translation from in vitro to in vivo is facilitated. In this review, we present a summary of various combination strategies for overcoming MDR and the nanotechnology-based combination therapy.
Collapse
Affiliation(s)
- Meng Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ergang Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanna Cui
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yongzhuo Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
12
|
Abstract
Inhibitor of Apoptosis (IAP) proteins block programmed cell death and are expressed at high levels in various human cancers, thus making them attractive targets for cancer drug development. Second mitochondrial activator of caspases (Smac) mimetics are small-molecule inhibitors that mimic Smac, an endogenous antagonist of IAP proteins. Preclinical studies have shown that Smac mimetics can directly trigger cancer cell death or, even more importantly, sensitize tumor cells for various cytotoxic therapies, including conventional chemotherapy, radiotherapy, or novel agents. Currently, several Smac mimetics are under evaluation in early clinical trials as monotherapy or in rational combinations (i.e., GDC-0917/CUDC-427, LCL161, AT-406/Debio1143, HGS1029, and TL32711/birinapant). This review discusses the promise as well as some challenges at the translational interface of exploiting Smac mimetics as cancer therapeutics.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany. German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
13
|
Hernandez L, Kim MK, Noonan AM, Sagher E, Kohlhammer H, Wright G, Lyle LT, Steeg PS, Anver M, Bowtell DD, on behalf of the Australian Ovarian Cancer Study Group
57, Annunziata CM. A dual role for Caspase8 and NF- κB interactions in regulating apoptosis and necroptosis of ovarian cancer, with correlation to patient survival. Cell Death Discov 2015; 1:15053. [PMID: 28179987 PMCID: PMC5198842 DOI: 10.1038/cddiscovery.2015.53] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 09/24/2015] [Accepted: 09/26/2015] [Indexed: 12/15/2022] Open
Abstract
Ovarian cancer is a deadly disease characterized by primary and acquired resistance to chemotherapy. We previously associated NF-κB signaling with poor survival in ovarian cancer, and functionally demonstrated this pathway as mediating proliferation, invasion and metastasis. We aimed to identify cooperating pathways in NF-κB-dependent ovarian cancer cells, using genome-wide RNA interference as a loss-of-function screen for key regulators of cell survival with IKKβ inhibition. Functional genomic screen for interactions with NF-κB in ovarian cancer showed that cells depleted of Caspase8 died better with IKKβ inhibition. Overall, low Caspase8 was associated with shorter overall survival in three independent gene expression data sets of ovarian cancers. Conversely, Caspase8 expression was markedly highest in ovarian cancer subtypes characterized by strong T-cell infiltration and better overall prognosis, suggesting that Caspase8 expression increased chemotherapy-induced cell death. We investigated the effects of Caspase8 depletion on apoptosis and necroptosis of TNFα-stimulated ovarian cancer cell lines. Inhibition of NF-κB in ovarian cancer cells switched the effects of TNFα signaling from proliferation to death. Although Caspase8-high cancer cells died by apoptosis, Caspase8 depletion downregulated NF-κB signaling, stabilized RIPK1 and promoted necroptotic cell death. Blockage of NF-κB signaling and depletion of cIAP with SMAC-mimetic further rendered these cells susceptible to killing by necroptosis. These findings have implications for anticancer strategies to improve outcome for women with low Caspase8-expressing ovarian cancer.
Collapse
Affiliation(s)
- L Hernandez
- Women’s Malignancies Branch, National Cancer Institute,
Bethesda, MD
20892-1906, USA
| | - M K Kim
- Women’s Malignancies Branch, National Cancer Institute,
Bethesda, MD
20892-1906, USA
| | - A M Noonan
- Women’s Malignancies Branch, National Cancer Institute,
Bethesda, MD
20892-1906, USA
| | - E Sagher
- Women’s Malignancies Branch, National Cancer Institute,
Bethesda, MD
20892-1906, USA
| | - H Kohlhammer
- Metabolism Branch, Center for Cancer Research, National Cancer Institute,
Bethesda, MD
20892-1906, USA
| | - G Wright
- Biometric Research Branch, Division of Cancer Treatment and Diagnosis, National
Cancer Institute, Bethesda, MD
20892-1906, USA
| | - L T Lyle
- Women’s Malignancies Branch, National Cancer Institute,
Bethesda, MD
20892-1906, USA
| | - P S Steeg
- Women’s Malignancies Branch, National Cancer Institute,
Bethesda, MD
20892-1906, USA
| | - M Anver
- Pathology/Histotechnology Laboratory, LASP, Leidos Biomedical Research, Inc.,
Frederick, MD
21702-1201, USA
| | - D D Bowtell
- Centre for Cancer Genomics and Predictive Medicine, Peter MacCallum Cancer
Centre, East Melbourne, Victoria, Australia
- The Department of Pathology, University of Melbourne, Parkville,
Victoria, Australia
| | - on behalf of the Australian Ovarian Cancer Study Group
57
- Women’s Malignancies Branch, National Cancer Institute,
Bethesda, MD
20892-1906, USA
- Metabolism Branch, Center for Cancer Research, National Cancer Institute,
Bethesda, MD
20892-1906, USA
- Biometric Research Branch, Division of Cancer Treatment and Diagnosis, National
Cancer Institute, Bethesda, MD
20892-1906, USA
- Pathology/Histotechnology Laboratory, LASP, Leidos Biomedical Research, Inc.,
Frederick, MD
21702-1201, USA
- Centre for Cancer Genomics and Predictive Medicine, Peter MacCallum Cancer
Centre, East Melbourne, Victoria, Australia
- The Department of Pathology, University of Melbourne, Parkville,
Victoria, Australia
| | - C M Annunziata
- Women’s Malignancies Branch, National Cancer Institute,
Bethesda, MD
20892-1906, USA
| |
Collapse
|
14
|
Qin S, Xu C, Li S, Yang C, Sun X, Wang X, Tang SC, Ren H. Indomethacin induces apoptosis in the EC109 esophageal cancer cell line by releasing second mitochondria-derived activator of caspase and activating caspase-3. Mol Med Rep 2015; 11:4694-700. [PMID: 25673090 DOI: 10.3892/mmr.2015.3331] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 12/19/2014] [Indexed: 11/05/2022] Open
Abstract
The use of non‑steroidal anti‑inflammatory drugs (NSAIDs) has been associated with a reduced risk of various types of cancer, including esophageal cancer. However, the mechanisms underlying the antineoplastic effects of NSAIDs in esophageal cancer remain to be elucidated. In the present study, a significant inhibition in cell viability was observed in the EC109 cells following treatment with different concentrations of indomethacin, and these effects occurred in a dose‑ and time‑dependent manner. This inhibition was due to the release of second mitochondria‑derived activator of caspase (Smac) into the cytosol and the activation of caspase‑3. Subsequently, flow cytometry was performed to investigate indomethacin‑induced apoptosis following the overexpression or knockdown of Smac, and western blot analysis was performed to determine the expression of Smac and the activation of caspase‑3. Overexpression of Smac was promoted apoptosis, while downregulation of Smac significantly inhibited apoptosis. Western blot analysis demonstrated that indomethacin induced apoptosis through releasing Smac into the cytosol and activating caspase‑3. These results indicated that Smac is essential for the apoptosis induced by indomethacin in esophageal cancer cells.
Collapse
Affiliation(s)
- Sida Qin
- Department of Thoracic Surgery, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Chongwen Xu
- Department of Thoracic Surgery, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shuo Li
- Department of Thoracic Surgery, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Chengcheng Yang
- Department of Oncology, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xin Sun
- Department of Thoracic Surgery, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xifang Wang
- Department of Thoracic Surgery, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shou-Ching Tang
- Department of Hematology and Oncology, Georgia Regents University Cancer Center, Augusta, GA 30912, USA
| | - Hong Ren
- Department of Thoracic Surgery, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
15
|
Abstract
Inhibitor of apoptosis (IAP) proteins are overexpressed in multiple human malignancies, an event that is associated with poor prognosis and treatment resistance. Therefore, IAP proteins represent relevant targets for therapeutic intervention. Second mitochondrial activator of caspases (Smac) is a mitochondrial protein that is released into the cytosol upon the induction of programmed cell death and promotes apoptosis by neutralizing IAP proteins. On the basis of this property, a variety of small-molecule inhibitors have been developed that mimic the binding domain of the native Smac protein to IAP proteins. Evaluation of these Smac mimetics in preclinical studies revealed that they particularly synergize together with agents that trigger the death receptor pathway of apoptosis. Such combinations might therefore be of special interest for being included in the ongoing evaluation of Smac mimetics in early clinical trials.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| |
Collapse
|
16
|
Gatti L, De Cesare M, Ciusani E, Corna E, Arrighetti N, Cominetti D, Belvisi L, Potenza D, Moroni E, Vasile F, Lecis D, Delia D, Castiglioni V, Scanziani E, Seneci P, Zaffaroni N, Perego P. Antitumor Activity of a Novel Homodimeric SMAC Mimetic in Ovarian Carcinoma. Mol Pharm 2013; 11:283-93. [DOI: 10.1021/mp4004578] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Laura Gatti
- Department
of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Michelandrea De Cesare
- Department
of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Emilio Ciusani
- Laboratory
of Clinical Pathology and Medical Genetics, Fondazione IRCCS Istituto Neurologico C. Besta, Via Celoria 11, Milan 20133, Italy
| | - Elisabetta Corna
- Department
of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Noemi Arrighetti
- Department
of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Denis Cominetti
- Department
of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Laura Belvisi
- Department
of Chemistry, Università degli Studi di Milano, Via Golgi
19, Milan 20133, Italy
| | - Donatella Potenza
- Department
of Chemistry, Università degli Studi di Milano, Via Golgi
19, Milan 20133, Italy
| | - Elisabetta Moroni
- Department
of Chemistry, Università degli Studi di Milano, Via Golgi
19, Milan 20133, Italy
| | - Francesca Vasile
- Department
of Chemistry, Università degli Studi di Milano, Via Golgi
19, Milan 20133, Italy
| | - Daniele Lecis
- Department
of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Domenico Delia
- Department
of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Vittoria Castiglioni
- Department
of Veterinary Science and Public Health, Università degli Studi di Milano, Via Celoria 10, Milan 20133, Italy
| | - Eugenio Scanziani
- Department
of Veterinary Science and Public Health, Università degli Studi di Milano, Via Celoria 10, Milan 20133, Italy
| | - Pierfausto Seneci
- Department
of Chemistry, Università degli Studi di Milano, Via Golgi
19, Milan 20133, Italy
| | - Nadia Zaffaroni
- Department
of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Paola Perego
- Department
of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| |
Collapse
|