1
|
Chen TC, Swenson S, Thein TZ, Minea RO, Schönthal AH. Potent Therapeutic Activity of NEO212 in Preclinical Models of Human and Canine Leukaemia and Lymphoma. Vet Comp Oncol 2025. [PMID: 40377133 DOI: 10.1111/vco.13066] [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: 01/31/2025] [Revised: 04/25/2025] [Accepted: 05/05/2025] [Indexed: 05/18/2025]
Abstract
Haematological cancer types, such as leukaemia and lymphoma, represent diseases that are life-threatening to canine and human patients alike, and better treatments are needed. We are developing a novel anticancer agent, NEO212, a conjugate of two cancer drugs, the alkylating agent temozolomide (TMZ) and the monoterpene perillyl alcohol (POH). NEO212 has revealed robust therapeutic activity in preclinical tumour models harbouring different human cancer types. In the comparative preclinical study presented here, a two-species (canine and human) and two-cancer (leukaemia and lymphoma) analysis was performed to determine whether the promising therapeutic activity of NEO212 would span species and cancer types. We investigated the activity of NEO212 in human and canine leukaemia and lymphoma cell lines in vitro and in corresponding mouse models in vivo. Our results show that in vitro NEO212 is significantly more potent than TMZ and POH in all cell lines and exerts activity even against strongly TMZ-resistant tumour cells. In vivo, oral NEO212 strikingly extends the survival of mice harbouring human or canine leukaemia or lymphoma cells. At the same time, NEO212 is well tolerated in dogs at dosages higher than those that achieved therapeutic activity in mouse models. Our study introduces NEO212 as a novel oral cancer drug candidate for both human and veterinary oncology applications.
Collapse
Affiliation(s)
- Thomas C Chen
- Department of Neurosurgery, Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
- USC/Norris Comprehensive Cancer Center, Los Angeles, California, USA
- NeOnc Technologies, Inc., Westlake Village, California, USA
| | - Steve Swenson
- Department of Neurosurgery, Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
| | - Thu Zan Thein
- Department of Neurosurgery, Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
| | - Radu O Minea
- Department of Neurosurgery, Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
- USC/Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Axel H Schönthal
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, USC, Los Angeles, California, USA
| |
Collapse
|
2
|
Zhang L, Wang G, Li Z, Yang J, Li H, Wang W, Li Z, Li H. Molecular pharmacology and therapeutic advances of monoterpene perillyl alcohol. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155826. [PMID: 38897045 DOI: 10.1016/j.phymed.2024.155826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/20/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Perillyl alcohol (POH) is a aroma monoterpene commonly obtained from various plants' essential oil. Recently, increasing researches have demonstrated that POH may be useful, not only as flavor compound, but also as bioactive molecule because of a variety of biological activities. PURPOSE The aim of this review is to summarize the production, pharmacological activities and molecular mechanism, active derivatives, toxicity and parmacokinetics, and industrial application of POH. METHODS A systematic search of published articles up to January 2024 in Web of Science, China Knowledge Network, and PubMed databases is conducted using the following keywords: POH, POH derivatives, biological or pharmacological, production or synthesis, pharmacokinetics, toxicity and application. RESULTS Biotechnological production is considered to be a potential alternative approach to generate POH. POH provides diverse pharmacological benefits, including anticancer, antimicrobial, insecticidal, antioxidant, anti-inflammatory, hypotensive, vasorelaxant, antinociceptive, antiasthmatic, hepatoprotective effects, etc. The underlying mechanisms of action include modulation of NF-κB, JNK/c-Jun, Notch, Akt/mTOR, PI3K/Akt/eNOS, STAT3, Nrf2 and ERS response pathways, mitigation of mitochondrial dysfunction and membrane integrity damage, and inhibition of ROS accumulation, pro-inflammatory cytokines release and NLRP3 activation. What's more, the proteins or genes influenced by POH against diseases refer to Bax, Bcl-2, cyclin D1, CDK, p21, p53, HIF-1α, AP-1, caspase-3, M6P/IGF2R, PARP, VEGF, etc. Some clinical studies report that intranasal delivery of POH is a safe and effective treatment for cancer, but further clinical investigations are needed to confirm other health benefits of POH in human healthy. Depending on these health-promoting properties together with desirable flavor and safety, POH can be employed as dietary supplement, preservative and flavor additive in food and cosmetic fields, as building block in synthesis fields, as anticancer drug in medicinal fields, and as pesticides and herbicides in agricultural fields. CONCLUSION This review systematically summarizes the recent advances in POH and highlights its therapeutic effects and potential mechanisms as well as the clinical settings, which is helpful to develop POH into functional food and new candidate drug for prevention and management of diseases. Future studies are needed to conduct more biological activity studies of POH and its derivatives, and check their clinical efficacy and potential side effects.
Collapse
Affiliation(s)
- Lulu Zhang
- College of Food Science and Technology, Henan University of Technology, Zhengzhou, Henan 450001, PR China; Henan Province Wheat-flour Staple Food Engineering Technology Research Centre, Zhengzhou, Henan 450001, PR China.
| | - Guoguo Wang
- College of Food Science and Technology, Henan University of Technology, Zhengzhou, Henan 450001, PR China; Henan Province Wheat-flour Staple Food Engineering Technology Research Centre, Zhengzhou, Henan 450001, PR China
| | - Zehao Li
- College of Food Science and Technology, Henan University of Technology, Zhengzhou, Henan 450001, PR China; Henan Province Wheat-flour Staple Food Engineering Technology Research Centre, Zhengzhou, Henan 450001, PR China
| | - Jinchu Yang
- Technology Center, China Tobacco Henan Industrial Co., Ltd., Zhengzhou, Henan 450000, PR China.
| | - Haoliang Li
- Technology Center, China Tobacco Henan Industrial Co., Ltd., Zhengzhou, Henan 450000, PR China
| | - Wanying Wang
- College of Food Science and Technology, Henan University of Technology, Zhengzhou, Henan 450001, PR China; Henan Province Wheat-flour Staple Food Engineering Technology Research Centre, Zhengzhou, Henan 450001, PR China
| | - Zhijian Li
- College of Food Science and Technology, Henan University of Technology, Zhengzhou, Henan 450001, PR China; Henan Province Wheat-flour Staple Food Engineering Technology Research Centre, Zhengzhou, Henan 450001, PR China
| | - Hua Li
- College of Food Science and Technology, Henan University of Technology, Zhengzhou, Henan 450001, PR China; Henan Province Wheat-flour Staple Food Engineering Technology Research Centre, Zhengzhou, Henan 450001, PR China.
| |
Collapse
|
3
|
Jatyan R, Singh P, Sahel DK, Karthik YG, Mittal A, Chitkara D. Polymeric and small molecule-conjugates of temozolomide as improved therapeutic agents for glioblastoma multiforme. J Control Release 2022; 350:494-513. [PMID: 35985493 DOI: 10.1016/j.jconrel.2022.08.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/31/2022] [Accepted: 08/12/2022] [Indexed: 11/15/2022]
Abstract
Temozolomide (TMZ), an imidazotetrazine, is a second-generation DNA alkylating agent used as a first-line treatment of glioblastoma multiforme (GBM). It was approved by FDA in 2005 and declared a blockbuster drug in 2008. Although TMZ has shown 100% oral bioavailability and crosses the blood-brain barrier effectively, however it suffers from limitations such as a short half-life (∼1.8 h), rapid metabolism, and lesser accumulation in the brain (∼10-20%). Additionally, development of chemoresistance has been associated with its use. Since it is a potential chemotherapeutic agent with an unmet medical need, advanced delivery strategies have been explored to overcome the associated limitations of TMZ. Nanocarriers including liposomes, solid lipid nanoparticles (SLNs), nanostructure lipid carriers (NLCs), and polymeric nanoparticles have demonstrated their ability to improve its circulation time, stability, tissue-specific accumulation, sustained release, and cellular uptake. Because of the appreciable water solubility of TMZ (∼5 mg/mL), the physical loading of TMZ in these nanocarriers is always challenging. Alternatively, the conjugation approach, wherein TMZ has been conjugated to polymers or small molecules, has been explored with improved outcomes in vitro and in vivo. This review emphasized the practical evidence of the conjugation strategy to improve the therapeutic potential of TMZ in the treatment of glioblastoma multiforme.
Collapse
Affiliation(s)
- Reena Jatyan
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Prabhjeet Singh
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Deepak Kumar Sahel
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Y G Karthik
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India.
| |
Collapse
|
4
|
Chang M, Shang M, Yuan F, Guo W, Wang C. EF24 exerts cytotoxicity against NSCLC via inducing ROS accumulation. Cancer Cell Int 2021; 21:531. [PMID: 34641863 PMCID: PMC8513219 DOI: 10.1186/s12935-021-02240-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/29/2021] [Indexed: 12/11/2022] Open
Abstract
Background The role of Diphenyldifluoroketone (EF24), a synthetic analogue of curcumin with noteworthy antitumor potential, remains unclear in non-small cell lung cancer (NSCLC). Herein, the inhibitory effect of EF24 on NSCLC and its mechanism were studied. Methods Cytotoxicity was measured by MTT assay, colony formation assay and xenograft model. Cell apoptosis and reactive oxygen species (ROS) level were quantified by flow cytometer. Protein level was detected by western blot assay. Mitochondria and autophagosomes were observed using transmission electron microscope and confocal microscopy. Results In-vitro, EF24 significantly induced proliferation inhibition, apoptosis, mitochondrial fission and autophagy of NSCLC cell lines. These cytotoxic effects were significantly attenuated by two reactive oxygen species (ROS) scavengers, indicating its anti-cancer effects largely depend on ROS accumulation. In-vivo, EF24 inhibited tumor growth in a dose-dependent manner. Moreover, no pathological changes of heart, lung, spleen, kidney and liver of mice were observed. Collectively, EF24 induced ROS accumulation, in turn activates cell apoptosis, and then exerts its cytotoxicity on NSCLC cells. Conclusions The results showed that EF24 exerted cytotoxicity against NSCLC via ROS accumulation. Thus, EF24 might serve as a potential anti-cancer agent for the treatment of NSCLC. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02240-z.
Collapse
Affiliation(s)
- Minghui Chang
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Ming Shang
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Fang Yuan
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Wei Guo
- Ultrasound Diagnosis Department, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China.
| | - Cuijuan Wang
- Physical and Chemical Laboratory, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, People's Republic of China.
| |
Collapse
|
5
|
Chien CH, Hsueh WT, Chuang JY, Chang KY. Dissecting the mechanism of temozolomide resistance and its association with the regulatory roles of intracellular reactive oxygen species in glioblastoma. J Biomed Sci 2021; 28:18. [PMID: 33685470 PMCID: PMC7938520 DOI: 10.1186/s12929-021-00717-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is the most common primary malignant brain tumor that is usually considered fatal even with treatment. This is often a result for tumor to develop resistance. Regarding the standard chemotherapy, the alkylating agent temozolomide is effective in disease control but the recurrence will still occur eventually. The mechanism of the resistance is various, and differs in terms of innate or acquired. To date, aberrations in O6-methylguanine-DNA methyltransferase are the clear factor that determines drug susceptibility. Alterations of the other DNA damage repair genes such as DNA mismatch repair genes are also known to affect the drug effect. Together these genes have roles in the innate resistance, but are not sufficient for explaining the mechanism leading to acquired resistance. Recent identification of specific cellular subsets with features of stem-like cells may have role in this process. The glioma stem-like cells are known for its superior ability in withstanding the drug-induced cytotoxicity, and giving the chance to repopulate the tumor. The mechanism is complicated to administrate cellular protection, such as the enhancing ability against reactive oxygen species and altering energy metabolism, the important steps to survive. In this review, we discuss the possible mechanism for these specific cellular subsets to evade cancer treatment, and the possible impact to the following treatment courses. In addition, we also discuss the possibility that can overcome this obstacle.
Collapse
Affiliation(s)
- Chia-Hung Chien
- National Institute of Cancer Research, National Health Research Institutes, 367 Sheng-Li Road, Tainan, 70456, Taiwan
| | - Wei-Ting Hsueh
- Department of Oncology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Jian-Ying Chuang
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan.,The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kwang-Yu Chang
- National Institute of Cancer Research, National Health Research Institutes, 367 Sheng-Li Road, Tainan, 70456, Taiwan. .,Department of Oncology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
6
|
Cho HY, Swenson S, Thein TZ, Wang W, Wijeratne NR, Marín-Ramos NI, Katz JE, Hofman FM, Schönthal AH, Chen TC. Pharmacokinetic properties of the temozolomide perillyl alcohol conjugate (NEO212) in mice. Neurooncol Adv 2020; 2:vdaa160. [PMID: 33392507 PMCID: PMC7764505 DOI: 10.1093/noajnl/vdaa160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background NEO212 is a novel small-molecule anticancer agent that was generated by covalent conjugation of the natural monoterpene perillyl alcohol (POH) to the alkylating agent temozolomide (TMZ). It is undergoing preclinical development as a therapeutic for brain-localized malignancies. The aim of this study was to characterize metabolism and pharmacokinetic (PK) properties of NEO212 in preclinical models. Methods We used mass spectrometry (MS) and modified high-performance liquid chromatography to identify and quantitate NEO212 and its metabolites in cultured glioblastoma cells, in mouse plasma, brain, and excreta after oral gavage. Results Our methods allowed identification and quantitation of NEO212, POH, TMZ, as well as primary metabolites 5-aminoimidazole-4-carboxamide (AIC) and perillic acid (PA). Intracellular concentrations of TMZ were greater after treatment of U251TR cells with NEO212 than after treatment with TMZ. The half-life of NEO212 in mouse plasma was 94 min. In mice harboring syngeneic GL261 brain tumors, the amount of NEO212 was greater in the tumor-bearing hemisphere than in the contralateral normal hemisphere. The brain:plasma ratio of NEO212 was greater than that of TMZ. Excretion of unaltered NEO212 was through feces, whereas its AIC metabolite was excreted via urine. Conclusions NEO212 preferentially concentrates in brain tumor tissue over normal brain tissue, and compared to TMZ has a higher brain:plasma ratio, altogether revealing favorable features to encourage its further development as a brain-targeted therapeutic. Its breakdown into well-characterized, long-lived metabolites, in particular AIC and PA, will provide useful equivalents for PK studies during further drug development and clinical trials with NEO212.
Collapse
Affiliation(s)
- Hee-Yeon Cho
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Steve Swenson
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Thu Zan Thein
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Weijun Wang
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Neloni R Wijeratne
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, California, USA
| | - Nagore I Marín-Ramos
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jonathan E Katz
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, California, USA
| | - Florence M Hofman
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Pathology Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Axel H Schönthal
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Thomas C Chen
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Pathology Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
7
|
Albino de Souza G, de Castro Bezerra F, Martins TD. Photophysical Properties of Fluorescent Self-Assembled Peptide Nanostructures for Singlet Oxygen Generation. ACS OMEGA 2020; 5:8804-8815. [PMID: 32337442 PMCID: PMC7178805 DOI: 10.1021/acsomega.0c00381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/30/2020] [Indexed: 06/11/2023]
Abstract
In this work, a drug delivery system for perillyl alcohol based on the peptide self-assembly containing 3-(2-benzothiazolyl)-7-(diethylamino)coumarin (C6) as a fluorescent additive is obtained, and its photophysical characteristics as well as its release dynamics were studied by steady-state and time-resolved fluorescence spectroscopy. Results proved the dynamics of drug release from the peptide nanostructures and showed that the system formed by the self-assembled peptide and C6, along with perillyl alcohol, presents unique photophysical properties that can be exploited to generate singlet oxygen (1O2) upon irradiation, which is not achieved by the sole components. Through epifluorescence microscopy combined with time-correlated single photon counting fluorescence spectroscopy, the release mechanism was proven to occur upon peptide structure interconversion, which is controlled by environmental changes.
Collapse
Affiliation(s)
- Geovany Albino de Souza
- Chemistry Institute, Federal University of Goiás, Av. Esperança, s/n, Vila Itatiaia, BR 74690900 Goiânia, Goiás, Brazil
| | | | - Tatiana Duque Martins
- Chemistry Institute, Federal University of Goiás, Av. Esperança, s/n, Vila Itatiaia, BR 74690900 Goiânia, Goiás, Brazil
| |
Collapse
|
8
|
Silva-Hirschberg C, Hartman H, Stack S, Swenson S, Minea RO, Davitz MA, Chen TC, Schönthal AH. Cytotoxic impact of a perillyl alcohol-temozolomide conjugate, NEO212, on cutaneous T-cell lymphoma in vitro. Ther Adv Med Oncol 2019; 11:1758835919891567. [PMID: 31839810 PMCID: PMC6900611 DOI: 10.1177/1758835919891567] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 11/04/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Mycosis fungoides (MF) and Sézary syndrome (SS) are subtypes of primary
cutaneous lymphomas and represent complex diseases regarding their
physiopathology and management. Depending on the stage of the disease,
different treatment regimens are applied, but there is no consensus on an
optimal approach. Prognosis for patients with early stage MF is favorable,
but significantly worsens in advanced disease and in SS, where patients
frequently relapse and require multiple therapies. Methods: We investigated the potential anticancer effects of NEO212, a novel compound
generated by covalently conjugating perillyl alcohol (a natural monoterpene)
to temozolomide (an alkylating agent), on MF and SS cell lines in
vitro. HUT-78, HUT-102, and MyLa cells were treated with NEO212
under different conditions, and drug effects on proliferation, viability,
and apoptosis were characterized. Results: NEO212 inhibited proliferation, diminished viability, and stimulated
apoptosis in all cell lines, although with varying degrees of potency in the
different cell lines. It down-regulated c-myc and cyclin D1 proteins, which
are required for cell proliferation, but triggered endoplasmic reticulum
stress and activation of caspases. Pretreatment of cells with antioxidants
ascorbic acid and beta-mercaptoethanol prevented these NEO212-induced
effects. Conclusions: NEO212 exerted promising anticancer effects on SS and MF cell lines. The
generation of reactive oxygen species (ROS) appears to play a key role in
the NEO212-induced cell death process, because the blockage of ROS with
antioxidants prevented caspase activation. We propose that NEO212 should be
investigated further toward clinical testing in these tumor types.
Collapse
Affiliation(s)
- Catalina Silva-Hirschberg
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hannah Hartman
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Samantha Stack
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Steve Swenson
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Radu O Minea
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michael A Davitz
- Leason Ellis, One Barker Avenue, Fifth Floor, White Plains, New York, NY, USA
| | - Thomas C Chen
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Axel H Schönthal
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, HMR-405, Los Angeles, CA 90089, USA
| |
Collapse
|
9
|
Ding S, Song X, Geng X, Liu L, Ma H, Wang X, Wei L, Xie L, Song X. Saliva-derived cfDNA is applicable for EGFR mutation detection but not for quantitation analysis in non-small cell lung cancer. Thorac Cancer 2019; 10:1973-1983. [PMID: 31441578 PMCID: PMC6775000 DOI: 10.1111/1759-7714.13178] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 12/21/2022] Open
Abstract
Background Both quantitative and qualitative aspects of plasma cell‐free DNA (plasma cfDNA, pcfDNA) have been well‐studied as potential biomarkers in non‐small cell lung cancer (NSCLC). Accumulating evidence has proven that saliva also has the potential for the detection and analysis of circulating free DNA (saliva cfDNA, scfDNA). Methods In the current study, we aimed to explore the potential application of scfDNA in NSCLC diagnostics and consistency of epidermal growth factor receptor (EGFR) mutation detection in paired pcfDNA and scfDNA using droplet digital PCR (ddPCR) and analyze the relationship between EGFR mutations and clinical treatment response. Results In the quantitative cohort study, scfDNA concentration in NSCLC patients was no different from that in healthy donors, or in benign patients. ScfDNA concentration was significantly lower than pcfDNA concentration, yet they were not statistically significant in relevance (Spearman's rank correlation r = −0.123, P = 0.269). In the qualitative cohort study, the overall concordance rate of EGFR mutations between pcfDNA and scfDNA was 83.78% (31 of 37; k = 0.602; P < 0.001). EGFR mutation detection in paired pcfDNA and scfDNA was significantly correlated with the clinical treatment response (Spearman's rank correlation r = 0.664, P = 0.002). Conclusions Our results demonstrated that saliva might not be the idea material for a cfDNA quantitative test, and scfDNA concentration is not applicable for NSCLC diagnostics. Conversely, scfDNA was capable of acting as the supplement for EGFR mutations due to the coincidence rate of EGFR mutation detection between scfDNA and pcfDNA.
Collapse
Affiliation(s)
- Shanshan Ding
- Department of Clinical Laboratory, Shandong Cancer Hospital affiliated to Shandong University, Jinan, China.,Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xingguo Song
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xinran Geng
- Department of Clinical Laboratory, Maternity & Child Care Center of Dezhou, Dezhou, China
| | - Lele Liu
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medicine Science, Jinan, China
| | - Hongxin Ma
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiujuan Wang
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ling Wei
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Li Xie
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xianrang Song
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
10
|
Song X, Liu L, Chang M, Geng X, Wang X, Wang W, Chen TC, Xie L, Song X. NEO212 induces mitochondrial apoptosis and impairs autophagy flux in ovarian cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:239. [PMID: 31174569 PMCID: PMC6554966 DOI: 10.1186/s13046-019-1249-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 05/27/2019] [Indexed: 12/21/2022]
Abstract
Background Temozolomide-perillyl alcohol conjugate (NEO212), a novel temozolomide (TMZ) analog, was previously reported to exert its anti-cancer effect in non-small cell lung cancer (NSCLC), and human nasopharyngeal carcinoma (NPC), etc.. In the current study, we intend to illuminate the potential anticancer property and the underly mechanisms of NEO212 in ovarian cancer cells. Methods The cytotoxicity of NEO212 was detected by MTT, colony formation analysis and xenograft model. The proteins involved in cell proliferation, DNA damage, autophagy and lysosomal function were detected by western blots; mitochondria, lysosome and autophagosome were visualized by TEM and/or immunofluorescence; Apoptosis, cell cycle analysis and mitochondrial transmembrane potential were detected by flow cytometry. TFEB translocation was detected by immunofluorescence and western blot. Results NEO212 has the potential anticancer property in ovarian cancer cells, as evidence from cell proliferation inhibition, G2/M arrest, DNA damage, xenograft, mitochondrial dysfunction and apoptosis. Importantly, we observed that although it induced significant accumulation of autophagosomes, NEO212 quenched GFP-LC3 degradation, down-regulated a series of lysosome related gene expression and blocked the autophagic flux, which significantly facilitated it induced apoptosis and was largely because it inhibited the nuclear translocation of transcription factor EB (EB). Conclusions NEO212 inhibited TFEB translocation, and impaired the lysosomal function, implying NEO212 might avoid from autophagy mediated chemo-resistance, thus proposing NEO212 as a potential therapeutic candidate for ovarian cancer. Electronic supplementary material The online version of this article (10.1186/s13046-019-1249-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xingguo Song
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Lisheng Liu
- Key Laboratory of Animal Resistance Research, College of Life Science, Shandong Normal University, 88 East Wenhua Road, Jinan, Shandong, People's Republic of China.,Department of Clinical Laboratory, Shandong Cancer Hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Minghui Chang
- School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China.,Department of Clinical Laboratory, Shandong Cancer Hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Xinran Geng
- Maternity & Child Care Center of Dezhou, Dongdizhong Street 835#, Decheng District, Dezhou, Shandong, People's Republic of China
| | - Xingwu Wang
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Weijun Wang
- Departments of Neurological Surgery, and Pathology, University of Southern California, Los Angeles, California, USA
| | - Thomas C Chen
- Departments of Neurological Surgery, and Pathology, University of Southern California, Los Angeles, California, USA
| | - Li Xie
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China.,Department of Clinical Laboratory, Shandong Cancer Hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Xianrang Song
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China. .,Department of Clinical Laboratory, Shandong Cancer Hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China.
| |
Collapse
|
11
|
Qiu M, Zhang S, Ke L, Tang H, Zeng X, Liu J. JS-K enhances chemosensitivity of prostate cancer cells to Taxol via reactive oxygen species activation. Oncol Lett 2018; 17:757-764. [PMID: 30655827 PMCID: PMC6312932 DOI: 10.3892/ol.2018.9684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 05/15/2018] [Indexed: 12/24/2022] Open
Abstract
The aim of the present study was to investigate the influence of the nitric oxide donor prodrug JS-K (C13H16N6O8) on Taxol-induced apoptosis in prostate cancer cells, and to investigate a potential reactive oxygen species (ROS)-associated mechanism. The effect of JS-K on the anticancer activity of Taxol was assessed in prostate cancer cells; cell viability, colony formation, apoptosis, ROS generation and expression levels of apoptosis-associated proteins were investigated. The function of ROS accumulation in the combined effects of JS-K and Taxol was determined using the antioxidant N-acetylcysteine (NAC) and the pro-oxidant oxidized glutathione (GSSG). The results of the present study demonstrated that JS-K was able to increase Taxol-induced suppression of prostate cancer cell proliferation, apoptosis, ROS accumulation and upregulation of apoptosis-associated proteins. Furthermore, NAC reversed the effect of JS-K on Taxol-induced apoptosis and conversely, the pro-oxidant GSSG exacerbated the effect of JS-K on Taxol-induced apoptosis in prostate cancer cells. In conclusion, JS-K enhances the chemosensitivity of prostate cancer cells to Taxol, via the upregulation of intracellular ROS.
Collapse
Affiliation(s)
- Mingning Qiu
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Sai Zhang
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Longzhi Ke
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Huancheng Tang
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Xin Zeng
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Jianjun Liu
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
12
|
Chang M, Song X, Geng X, Wang X, Wang W, Chen TC, Xie L, Song X. Temozolomide-Perillyl alcohol conjugate impairs Mitophagy flux by inducing lysosomal dysfunction in non-small cell lung Cancer cells and sensitizes them to irradiation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:250. [PMID: 30326943 PMCID: PMC6191917 DOI: 10.1186/s13046-018-0905-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/08/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND Temozolomide-perillyl alcohol conjugate (TMZ-POH), a novel Temozolomide (TMZ) analog developed based on the conjugation of TMZ and perillyl alcohol (POH), displayed strong anticancer potency in multiple cancer types. In this study, we aimed to clarify the relationship between TMZ-POH and autophagy, and explore the underlying mechanisms involved in. METHODS The proteins involved in autophagy, mitochondrial fission, lysosomal function and membrane traffic were detected by western blots; Autophagosome, mitochondria and lysosome were visualized by transmission electron microscope (TEM) and immunostaining; Apoptosis analysis and fluorescence probe detection were applied by flow cytometry. RESULTS TMZ-POH blocked mitophagy flux although the number of autophagosomes which colocalized with mitochondria in the cells was increased via inducing lysosomal dysfunction as evidence from impaired lysosomal acidification, maturation and hampered autophagosome- lysosome fusion, which largely depended on its downregulation on the small GTPase RAB7A via mevalonate pathway. More importantly, our data demonstrated TMZ-POH sensitized cancer cell to irradiation induced apoptosis. CONCLUSIONS Temozolomide-perillyl alcohol conjugate impairs mitophagy flux by inducing lysosomal dysfunction in Non-Small Cell Lung Cancer (NSCLC) cells and sensitizes them to irradiation, thereby proposing TMZ-POH as a potential radiosensitizer.
Collapse
Affiliation(s)
- Minghui Chang
- School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medicine Science, Jinan, Shandong, People's Republic of China.,Department of Clinical Laboratory, Shandong cancer hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Xingguo Song
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong cancer hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Xinran Geng
- Maternity & Child Care Center of Dezhou, Dongdizhong Street 835#, Decheng District, Dezhou, Shandong, People's Republic of China
| | - Xingwu Wang
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong cancer hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Weijun Wang
- Departments of Neurological Surgery, and Pathology, University of Southern California, California, Los Angeles, USA
| | - Thomas C Chen
- Departments of Neurological Surgery, and Pathology, University of Southern California, California, Los Angeles, USA
| | - Li Xie
- Department of Clinical Laboratory, Shandong cancer hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China.,Shandong Provincial Key Laboratory of Radiation Oncology, Shandong cancer hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Xianrang Song
- Department of Clinical Laboratory, Shandong cancer hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China. .,Shandong Provincial Key Laboratory of Radiation Oncology, Shandong cancer hospital affiliated to Shandong University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China.
| |
Collapse
|
13
|
Yu X, Lin H, Wang Y, Lv W, Zhang S, Qian Y, Deng X, Feng N, Yu H, Qian B. d-limonene exhibits antitumor activity by inducing autophagy and apoptosis in lung cancer. Onco Targets Ther 2018; 11:1833-1847. [PMID: 29670359 PMCID: PMC5894671 DOI: 10.2147/ott.s155716] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Purpose d-limonene is a plant extract with widespread application, and it has been recently reported to have antiproliferative and proapoptotic effects on cancer cells. However, the mechanisms by which d-limonene achieves these effects, especially in lung cancer, are not entirely clear. Therefore, the goal of this study was to examine the effects of d-limonene on lung cancer and explore its mechanisms of action. Methods We examined the therapeutic effects of d-limonene on lung cancer cells and in a xenograft animal model by characterizing its effects on the pathways of apoptosis and autophagy. Cell proliferation was measured using the Cell Counting Kit-8, and apoptosis was determined by flow cytometric analysis. Levels of LC3 puncta, an autophagy marker, were analyzed by laser scanning confocal microscopy. Autophagy and apoptosis-related gene expression were assessed by real-time quantitative polymerase chain reaction and Western blot. Results d-limonene inhibited the growth of lung cancer cells and suppressed the growth of transplanted tumors in nude mice. Expression of apoptosis and autophagy-related genes were increased in tumors after treatment with d-limonene. Furthermore, the use of chloroquine, an autophagy inhibitor, and knockdown of the atg5 gene, suppressed the apoptosis induced by d-limonene. Conclusion d-limonene may have a therapeutic effect on lung cancer as it can induce apoptosis of lung cancer cells by promoting autophagy.
Collapse
Affiliation(s)
- Xiao Yu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyan Lin
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Wang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenwen Lv
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Zhang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Qian
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaobei Deng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nannan Feng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Herbert Yu
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Biyun Qian
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Differential expression of peroxiredoxin 3 in laryngeal squamous cell carcinoma. Oncotarget 2018; 8:3471-3480. [PMID: 27966448 PMCID: PMC5356896 DOI: 10.18632/oncotarget.13838] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 11/22/2016] [Indexed: 02/05/2023] Open
Abstract
Peroxiredoxin (PRDX) proteins are involved in carcinogenesis. PRDX3, which is predominantly localized in mitochondria and up-regulated in several human cancers, seems to confer increased treatment resistance and aggressive phenotypes. This study examined the expression profile of PRDX3 and its possible clinical value in laryngeal squamous cell carcinoma (LSCC). The expression of PRDX3 in LSCC samples was confirmed by Western blotting and further analyzed by immunohistochemistry in LSCC samples of different clinical pathological stages. The results showed that up-regulated expression of PRDX3 was observed in LSCC and associated with poor differentiation (P < 0.01), primary tumor location, N category and tumor stage (P < 0.05). Knockdown of PRDX3 in the Hep-2 laryngeal carcinoma epithelial cell line significantly enhanced Hep-2 cells’ apoptosis and inhibited their proliferation and migration. Taken together, our results suggest that PRDX3 has substantial clinical impact on the progression of LSCC and shed new light on the role of PRDX3 in treatment resistance and aggressive phenotypes in LSCC.
Collapse
|
15
|
AAV-Mediated angiotensin 1-7 overexpression inhibits tumor growth of lung cancer in vitro and in vivo. Oncotarget 2018; 8:354-363. [PMID: 27861149 PMCID: PMC5352125 DOI: 10.18632/oncotarget.13396] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/11/2016] [Indexed: 11/25/2022] Open
Abstract
Ang-(1-7) inhibits lung cancer cell growth both in vitro and in vivo. However, the molecular mechanism of action is unclear and also the rapid degradation of Ang-(1-7) in vivo limits its clinical application. Here, we have demonstrated that Ang- (1-7) inhibits lung cancer cell growth by interrupting pre-replicative complex assembly and restrains epithelial-mesenchymal transition via Cdc6 inhibition. Furthermore, we constructed a mutant adeno-associated viral vector AAV8 (Y733F) that produced stable and high efficient Ang-(1-7) expression in a xenograft tumor model. The results show that AAV8-mediated Ang-(1-7) over-expression can remarkably suppress tumor growth in vivo by down-regulating Cdc6 and anti-angiogenesis. Ang-(1-7) over-expression via the AAV8 method may be a promising strategy for lung cancer treatment.
Collapse
|
16
|
Song X, Xie L, Chang M, Geng X, Wang X, Chen TC, Song X. Temozolomide-perillyl alcohol conjugate downregulates O 6-methylguanin DNA methltransferase via inducing ubiquitination-dependent proteolysis in non-small cell lung cancer. Cell Death Dis 2018; 9:202. [PMID: 29426908 PMCID: PMC5833843 DOI: 10.1038/s41419-017-0193-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 11/20/2017] [Accepted: 11/28/2017] [Indexed: 11/27/2022]
Abstract
The DNA repair enzyme O6-methylguanin-DNA-methltransferase (MGMT) is able to remove products of alkylating agent such as O6-meG and emerges as a central determinant of cancer resistance to temozolomide (TMZ). Temozolomide–perillyl alcohol conjugate (TMZ–POH), a novel TMZ analog developed based on the conjugation of TMZ and POH, displayed strong anticancer potency in multiple cancer types, but seemed not to experience the chemoresistance even in cells with high MGMT expression unlike TMZ and other alkylating agents. In this study, we demonstrated TMZ–POH inhibited MGMT dependent on proteasomal pathway and this inhibition is a significant factor in its toxic effect in the non-small cell lung cancer (NSCLC) cells.
Collapse
Affiliation(s)
- Xingguo Song
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Li Xie
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.,Department of Clinical Laboratory, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Minghui Chang
- Department of Clinical Laboratory, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.,School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medicine Science, Jinan, Shandong, China
| | - Xinran Geng
- Maternity & Child Care Center of Dezhou, Dongdizhong Street 835#, Decheng District, Dezhou, Shandong, China
| | - Xingwu Wang
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Thomas C Chen
- Departments of Neurological Surgery and Pathology, University of Southern California, Los Angeles, CA, USA
| | - Xianrang Song
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China. .,Department of Clinical Laboratory, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
17
|
Chang KY, Hsu TI, Hsu CC, Tsai SY, Liu JJ, Chou SW, Liu MS, Liou JP, Ko CY, Chen KY, Hung JJ, Chang WC, Chuang CK, Kao TJ, Chuang JY. Specificity protein 1-modulated superoxide dismutase 2 enhances temozolomide resistance in glioblastoma, which is independent of O 6-methylguanine-DNA methyltransferase. Redox Biol 2017; 13:655-664. [PMID: 28822335 PMCID: PMC5561972 DOI: 10.1016/j.redox.2017.08.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 12/12/2022] Open
Abstract
Acquisition of temozolomide (TMZ) resistance is a major factor leading to the failure of glioblastoma (GBM) treatment. The exact mechanism by which GBM evades TMZ toxicity is not always related to the expression of the DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT), and so remains unclear. In this study, TMZ-resistant variants derived from MGMT-negative GBM clinical samples and cell lines were studied, revealing there to be increased specificity protein 1 (Sp1) expression associated with reduced reactive oxygen species (ROS) accumulation following TMZ treatment. Analysis of gene expression databases along with cell studies identified the ROS scavenger superoxide dismutase 2 (SOD2) as being disease-related. SOD2 expression was also increased, and it was found to be co-expressed with Sp1 in TMZ-resistant cells. Investigation of the SOD2 promoter revealed Sp1 as a critical transcriptional activator that enhances SOD2 gene expression. Co-treatment with an Sp1 inhibitor restored the inhibitory effects of TMZ, and decreased SOD2 levels in TMZ-resistant cells. This treatment strategy restored susceptibility to TMZ in xenograft animals, leading to prolonged survival in an orthotopic model. Thus, our results suggest that Sp1 modulates ROS scavengers as a novel mechanism to increase cancer malignancy and resistance to chemotherapy. Inhibition of this pathway may represent a potential therapeutic target for restoring treatment susceptibility in GBM.
Collapse
Affiliation(s)
- Kwang-Yu Chang
- National Institute of Cancer Research, National Health Research Institutes, Taiwan; Department of Internal Medicine, National Cheng Kung University Hospital, Taiwan
| | - Tsung-I Hsu
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taiwan
| | - Che-Chia Hsu
- Graduate Institute of Medical Science, Taipei Medical University, Taiwan; Department of Cancer Biology, Wake Forest School of Medicine, USA
| | | | - Jr-Jiun Liu
- National Institute of Cancer Research, National Health Research Institutes, Taiwan; The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taiwan
| | - Shao-Wen Chou
- National Institute of Cancer Research, National Health Research Institutes, Taiwan
| | - Ming-Sheng Liu
- National Institute of Cancer Research, National Health Research Institutes, Taiwan
| | | | - Chiung-Yuan Ko
- The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taiwan
| | - Kai-Yun Chen
- The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taiwan
| | - Jan-Jong Hung
- Institute of Bioinformatics and Biosignal Transduction, National Cheng Kung University, Taiwan
| | - Wen-Chang Chang
- Graduate Institute of Medical Science, Taipei Medical University, Taiwan
| | - Cheng-Keng Chuang
- Department of Medicine, Chang Gung University, Taiwan; Department of Urology, Linkou Chang Gung Memorial Hospital, Taiwan
| | - Tzu-Jen Kao
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taiwan; The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taiwan.
| | - Jian-Ying Chuang
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taiwan; The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taiwan.
| |
Collapse
|
18
|
Na/K Pump and Beyond: Na/K-ATPase as a Modulator of Apoptosis and Autophagy. Molecules 2017; 22:molecules22040578. [PMID: 28430151 PMCID: PMC6154632 DOI: 10.3390/molecules22040578] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/29/2017] [Accepted: 03/29/2017] [Indexed: 01/22/2023] Open
Abstract
Lung cancer is a leading cause of global cancer deaths. Na/K-ATPase has been studied as a target for cancer treatment. Cardiotonic steroids (CS) trigger intracellular signalling upon binding to Na/K-ATPase. Normal lung and tumour cells frequently express different pump isoforms. Thus, Na/K-ATPase is a powerful target for lung cancer treatment. Drugs targeting Na/K-ATPase may induce apoptosis and autophagy in transformed cells. We argue that Na/K-ATPase has a role as a potential target in chemotherapy in lung cancer treatment. We discuss the effects of Na/K-ATPase ligands and molecular pathways inducing deleterious effects on lung cancer cells, especially those leading to apoptosis and autophagy.
Collapse
|
19
|
Yi X, Zhang Y, Zhong C, Zhong X, Xiao F. The role of STIM1 in the Cr(vi)-induced [Ca2+]iincrease and cell injury in L-02 hepatocytes. Metallomics 2016; 8:1273-1282. [DOI: 10.1039/c6mt00204h] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|